Sei sulla pagina 1di 293

Spermatogenesis

Biology and Clinical Implications


http://taylorandfrancis.com
Spermatogenesis
Biology and Clinical Implications

Edited by
C. Yan Cheng, PhD
Senior Scientist, Center for Biomedical Research
The Population Council, New York City, New York, USA
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742

© 2019 by Taylor & Francis Group, LLC


CRC Press is an imprint of Taylor & Francis Group, an Informa business

No claim to original U.S. Government works

Printed on acid-free paper

International Standard Book Number-13: 978-1-4987-6411-7 (Pack – Book and eBook)

This book contains information obtained from authentic and highly regarded sources. Reasonable efforts have been made to publish reliable data and infor-
mation, but the author and publisher cannot assume responsibility for the validity of all materials or the consequences of their use. The authors and publish-
ers have attempted to trace the copyright holders of all material reproduced in this publication and apologize to copyright holders if permission to publish in
this form has not been obtained. If any copyright material has not been acknowledged please write and let us know so we may rectify in any future reprint.

Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any electronic,
mechanical, or other means, now known or hereafter invented, including photocopying, microfilming, and recording, or in any information storage or
retrieval system, without written permission from the publishers.

For permission to photocopy or use material electronically from this work, please access www.copyright​.com (http://www.copyright.com/) or contact
the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides
licenses and registration for a variety of users. For organizations that have been granted a photocopy license by the CCC, a separate system of payment has
been arranged.

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for identification and explanation without
intent to infringe.

Library of Congress Cataloging‑in‑Publication Data

Names: Cheng, C. Yan, editor.


Title: Spermatogenesis : biology and clinical implications / edited by C. Yan Cheng.
Other titles: Spermatogenesis (Cheng)
Description: Boca Raton, FL : CRC Press, [2019] | Includes bibliographical references and index.
Identifiers: LCCN 2018003483| ISBN 9781498764117 (pack- hardback and ebook : alk. paper) |
ISBN 9780429488634 (ebook)
Subjects: | MESH: Spermatogenesis | Testis--physiology | Fertility--physiology
Classification: LCC QP255 | NLM WJ 834 | DDC 612.6/1--dc23
LC record available at https://lccn.loc.gov/2018003483

Visit the Taylor & Francis Web site at


http://www.taylorandfrancis.com

and the CRC Press Web site at


http://www.crcpress.com
Dedicated to:
My wife Po-lee and our children
Yan-ho and Chin-ho
http://taylorandfrancis.com
Contents

Preface ix
Contributors xi

1 Golgi apparatus regulation of differentiation: A case study for male germ cells of the rat testis 1
Louis Hermo, Regiana L. Oliveira, Charles E. Smith, Catherine E. Au, and John J. M. Bergeron
2 Androgen regulation of spermatogenesis 40
William H. Walker
3 Testicular immunoregulation: The role of Tyro3, Axl, and Mer receptor tyrosine kinases and pattern recognition receptors 52
Fei Wang, Qian Jiang, and Daishu Han
4 Inflammation and Spermatogenesis 63
Maria Susana Theas, Patricia Verónica Jacobo, Cecilia Valeria Pérez, Vanesa Anabella Guazzone, and Livia Lustig
5 Junctional adhesion molecule (JAM) family: Recent findings and their role and regulation in spermatogenesis 73
Kun Huang and Wing-Yee Lui
6 Sertoli cell immune regulation within the testis 81
Gurvinder Kaur, Kandis Wright, Robin Hannah Greer, Karl Mueller, Allan Haynes, and Jannette M. Dufour
7 The mitotic phase of spermatogenesis: Recent advances and perspectives 94
Kin Lam Fok and Hsiao Chang Chan
8 Genetics of mammalian meiosis 106
Fang Yang and P. Jeremy Wang
9 Roles of membrane and nuclear estrogen receptors in spermatogenesis 116
Paul S. Cooke, Manjunatha K. Nanjappa, Sergei G. Tevosian, and Rex A. Hess
10 Regulation of fertility and infertility in humans 123
Nahid Punjani, Ryan Flannigan, and Peter N. Schlegel
11 Male infertility: Evaluation and treatment 139
Ryan Flannigan and Marc Goldstein
12 Effects of chemical pollutants on spermatogenesis and implications in male infertility 155
Chris KC Wong
13 Advances in our understanding of human spermatogenesis 168
Qing Wen, Elizabeth I. Tang, Tito Jesus, Bruno Silvestrini, and C. Yan Cheng
14 A look into the testis as a reservoir for HIV and ZIKV—A reproductive biologist’s perspective 183
Elizabeth I. Tang, Christopher L. Robinson, Chi Nok Chong, Shuibing Chen, and C. Yan Cheng
15 Cytoskeletons (F-actin) and spermatogenesis 191
Liza O’Donnell and Peter G. Stanton
16 Roles of mTOR signaling in spermatogenesis 201
Lan Ye and Ke Zheng
17 Does planar cell polarity matter during spermatogenesis? 211
Linxi Li, Haiqi Chen, Qingquan Lian, Ren-Shan Ge, and C. Yan Cheng
18 Computational characterization and integrative analysis of proteins involved in spermatogenesis 220
Pranitha Jenardhanan, Manivel Panneerselvam, and Premendu P. Mathur
19 Effects of chemical exposures on testis cell-cell interactions and endocrine function 234
Rachel C. Knight, Jennifer R. Panizzi, and Benson T. Akingbemi
20 Environmental toxicants on Leydig cell function 245
Leping Ye, Xiaoheng Li, Xiaomin Chen, Qingquan Lian, and Ren-Shan Ge

Index 268

vii
http://taylorandfrancis.com
Preface

Over the last two decades, there have been major advances Lastly, there is interest in the field of developing non-
in the study of spermatogenesis. However, a number of hormonal contraceptives for men so that men can play a
these advances come from unexpected groups of inves- more active role in family planning among couples in both
tigators in other fields. These include investigators who developed and developing countries. This has sparked
specialize in studying stem cell biology, clinical urolo- interest, drawing chemists, biologists, clinicians, and
gists and endocrinologists, toxicologists, as well as chem- pharmacologists into the field to better understand sper-
ists, biologists, and clinicians. The rationale behind such matogenesis, so that better new molecules can be synthe-
as unexpected surge of interest in spermatogenesis is as sized and tested prior to clinical studies.16–18
follows. In this book, I attempt to provide a balanced over-
First, there is an intensive interest in the field to under- view of the topic of spermatogenesis from the perspec-
stand the biology of spermatogonial stem cells (SSCs) in tive of a reproductive biologist, while also providing
the testis, since investigators wish to understand if SSCs in some thought-provoking concepts on the latest develop-
rodents (Asingle spermatogonia) or humans (Adark spermato- ments in the field, which will benefit both clinical and
gonia)1–6 can be used to develop functional spermatids basic research scientists in the field. I have organized
in vitro,7–9 which would serve as a useful model to study this book so that the opening chapters are devoted to the
meiosis. These studies thus shed light on understanding basic aspects of hormonal and nonhormonal regulation
the molecular mechanism(s) that regulate meiosis, which of spermatogenesis, immunological regulation of sper-
remains largely unknown in the testis besides the morpho- matogenesis and other recently identified novel pathways,
logical events. There is also interest in the field to establish and meiosis and role of receptors. These are then followed
an in vitro testis model to study testis biology.7,8 In addition, by chapters devoted to the biology of human fertility and
researchers in the field are trying to use SSCs to develop infertility, with a chapter on the role of the testis that
other mammalian cells and/or tissues, such as insulin- serves as a reservoir for HIV and Zika virus. Thereafter,
producing pancreatic cells10 for tissue transplants. For we have chapters on the role of the cytoskeleton, polarity,
instance, a recent report has provided information neces- and mTOR signaling, and new developments in the field
sary to develop functional haploid spermatids from human using computational approaches to study spermatogen-
germline stem cells using a three-dimentional-induced esis. We conclude with chapters on the role of environ-
system.11 This information thus will be exceedingly help- mental toxicants that impede spermatogenesis. In short,
ful for clinicians to help men who cannot father their own all efforts were made to have some of the best investiga-
children due to nonobstructive azoospermia (NOA). In tors in their fields to update our latest developments in
short, many cell biologists are drawn to the field of sper- corresponding fields. As such, this book will serve as an
matogenesis to make important discoveries and advances. important reference work for investigators who wish to
Second, there is an increasing number of clinician-based encompass different aspects of the rapidly developing
scientists who are attracted to the field of spermatogenesis fields pertinent to the study of spermatogenesis. This is
because they need to provide better care to their patients. the goal of this work.
For instance, many patients with NOA who cannot produce I am grateful and indebted to all the colleagues who
functional spermatids were later found to possess normal worked with me selflessly for the past year. I am particu-
SSCs in their testes. The recent report on the production of larly indebted to Robert Peden and Kyle Meyer at the pub-
functional haploid spermatids from SSCs in men11 will pro- lisher’s office, who has provided the needed support and
vide hope to NOA patients since many of these men would patience during the preparation of this work. I am aslo
like to use their own functional haploid spermatids for in grateful to Amy Rodriguez who has worked selflessly to
vitro fertilization to father their own children using eggs assist us in copy-editing all the chapters and preparing all
from their spouses. Furthermore, this technique has been the proofs for the publication of this book.
shown in farm animals, as reported in a recent study in
which functional haploid spermatids were produced using C. Yan Cheng
an in vitro system from SSCs isolated from goat testes.12 New York City, New York
Third, some toxicologists and clinicians are also
attracted to the field of studying spermatogenesis because
studies have shown that the testis is a highly sensitive REFERENCES
endocrine organ to environmental toxicants,13–15 so that 1. Ehmcke J, Schlatt S. A revised model for spermato-
more studies are warranted in order to provide treatments gonial expansion in man: Lessons from non-human
to affected individuals. primates. Reproduction. 2006;132:673–680.

ix
x x

2. Ehmcke J, Wistuba J, Schlatt S. Spermatogonial stem 12. Deng SL, Zhang Y, Yu K et al. Melatonin up-regulates
cells: Questions, models and perspectives. Human the expression of the GATA-4 transcription factor
Reprod Update. 2006;12:275–282. and increases testosterone secretion from Leydig
3. de Rooij DG. The spermatogonial stem cell niche. cells through RORα signaling in an in vitro goal sper-
Microsc Res Tech. 2009;72:580–585. matogonial stem cell differentation culture system.
4. de Rooij DG, Russell LD. All you wanted to know Oncotarget. 2017;8:110592–110605.
about spermatogonia but were afraid to ask. J Androl. 13. Sedha S, Kumar S, Shukla S. Role of oxidative stress
2000;21:776–798. in male reproductive dysfunctions with reference to
5. Oatley JM, Brinster RL. The germline stem cell niche phthalate compounds. Urol J. 2015;12:2304–2316.
unit in mammalian testes. Physiol Rev. 2012;​92:577–595. 14. Gao Y, Mruk DD, Cheng CY. Sertoli cells are the
6. Phillips BT, Gassei K, Orwig KE. Spermatogonial target of environmental toxicants in the testis—A
stem cell regulation and spermatogenesis. Philos mechanistic and therapeutic insight. Expert Opin Ther
Trans R Soc Lond B Biol Sci. 2010;365:1663–1678. Targets 2015;19:1073–1090.
7. Sato T, Katagiri K, Gohbara A et al. In vitro produc- 15. Moffit JS, Her LS, Mineo AM, Knight BL, Phillips
tion of functional sperm in cultured neonatal mouse JA, Thibodeau MS. Assessment of inhibin B as a
testes. Nature. 2011;471:504–507. biomarker of testicular injury following administra-
8. Sato T, Katagiri K, Yoknishi T et al. In vitro produc- tion of carbendazim, cetrorelix, or 1,2-dibromo-3-­
tion of fertile sperm from murine spermatogonial chloropropane in Wistar han rats. Birth Defects Res B
stem cell lines. Nat Commun. 2011;2:472 (DOI:10.1038​ Dev Reprod Toxicol. 2013;98:17–28.
/ncomms1478). 16. Roth MY, Amory JK. Beyond the condom: Frontiers
9. Zhou Q, Wang M, Yuan Y et al. Complete meio- in male contraception. Semin Reprod Med. 2016;34:​
sis from embryonic stem cell-derived germ cells in 183–190.
vitro. Cell Stem Cell. 2016;18:330–340. 17. Gu X, Gupta V, Yang Y et al. Structure–activity
10. Skurikhin EG, Pakhomova AV, Ermakova NN et studies of N-butyl-1-deoxynojirimycin (NB-DNJ)
al. Role of tissue-specific stem and progenitor cells analogues: Discovery of potent and selective amino-
in the regeneration of the pancreas and testicu- cyclopentitol inhibitors of GBA1 and GBA2. Chem
lar tissue in diabetic disorders. Bull Exp Biol Med. Med Chem. 2017;12:1977–1984.
2016;162:146–152. 18. Schonbrunn E, Betzi S, Alam R et al. Development
11. Sun M, Yuan Q, Niu M et al. Efficient generation of of highly potent and selective diaminothiazole
functional haploid spermatids from human germline inhibitors of cyclin-dependent kinases. J Med Chem.
stem cells by three-dimensional-induced system. Cell 2013;56:3768–3782.
Death Differ. 2018;25:747–764.
Contributors

Benson T. Akingbemi C. Yan Cheng


Department of Anatomy, Physiology, and Pharmacology The Mary M. Wohlford Laboratory for Male Contraceptive
Auburn University Research
Auburn, Alabama Center for Biomedical Research
Population Council
Catherine E. Au New York City, New York
Department of Medicine
McGill University Hospital Research Institute Chi Nok Chong
Montreal, Canada Department of Surgery
Weill Cornell Medical College
John J. M. Bergeron New York City, New York
Department of Medicine
McGill University Hospital Research Institute Paul S. Cooke
Montreal, Canada Department of Physiological Sciences
University of Florida
Hsiao Chang Chan Gainesville, Florida
Epithelial Cell Biology Research Center
School of Biomedical Sciences, Faculty of Medicine Jannette M. Dufour
The Chinese University of Hong Kong Cell Biology and Biochemistry
Hong Kong, China Texas Tech University Health Sciences Center
and Lubbock, Texas
School of Biomedical Sciences Core Laboratory
Shenzhen Research Institute Ryan Flannigan
The Chinese University of Hong Kong Department of Urology
Shenzhen, China Weill Cornell Medicine
and New York City, New York
Sichuan University–The Chinese University of Hong Kong
Joint Laboratory for Reproductive Medicine Kin Lam Fok
West China Second University Hospital Epithelial Cell Biology Research Center
Chengdu, China School of Biomedical Sciences, Faculty of Medicine
The Chinese University of Hong Kong
Haiqi Chen Hong Kong, China
The Mary M. Wohlford Laboratory for Male Contraceptive and
Research School of Biomedical Sciences Core Laboratory
Center for Biomedical Research Shenzhen Research Institute
Population Council The Chinese University of Hong Kong
New York City, New York Shenzhen, China

Shuibing Chen Robin Hannah Greer


Department of Surgery Department of Cell Biology and Biochemistry
Weill Cornell Medical College Texas Tech University Health Sciences Center
New York City, New York Lubbock, Texas

Xiaomin Chen Ren-Shan Ge


The Second Affiliated Hospital and Yuying Children’s Hospital The Second Affiliated Hospital and Yuying Children’s Hospital
Wenzhou Medical University and
Wenzhou, Zhejiang Institute of Reproductive Biomedicine
Wenzhou Medical University
Zhejiang, China

xi
xii Contributors

Marc Goldstein Tito Jesus


Department of Urology The Mary M. Wohlford Laboratory for Male Contraceptive
Weill Cornell Medicine Research
New York City, New York Center for Biomedical Research
Population Council
Vanesa Anabella Guazzone New York City, New York
Department of Cellular Biology
University of Buenos Aires Qian Jiang
and Institute of Basic Medical Sciences
Cátedra II de Histología Chinese Academy of Medical Sciences
and and
Instituto de Investigaciones Biomédícas (INBIOMED) School of Basic Medicine
Buenos Aires, Argentina Peking Union Medical College
Beijing, China
Daishu Han
Institute of Basic Medical Sciences Gurvinder Kaur
Chinese Academy of Medical Sciences Department of Cell Biology and Biochemistry
and Texas Tech University Health Sciences Center
School of Basic Medicine Lubbock, Texas
Peking Union Medical College
Beijing, China Rachel C. Knight
Department of Anatomy, Physiology, and Pharmacology
Allan Haynes Auburn University
Department of Urology Auburn, Alabama
Texas Tech University Health Sciences Center
Lubbock, Texas Linxi Li
The Mary M. Wohlford Laboratory for Male Contraceptive
Louis Hermo Research
Department of Anatomy and Cell Biology Center for Biomedical Research
McGill University Population Council
Montreal, Canada New York City, New York
and
Rex A. Hess Institute of Reproductive Biomedicine
Department of Comparative Biosciences Wenzhou Medical University
University of Illinois at Urbana-Champaign Zhejiang, China
Urbana, Illinois
Xiaoheng Li
Kun Huang The Second Affiliated Hospital and Yuying Children’s Hospital
School of Biological Sciences Wenzhou Medical University
The University of Hong Kong Zhejiang, China
Hong Kong, China
Qingquan Lian
Patricia Verónica Jacobo The Second Affiliated Hospital and Yuying Children’s Hospital
Universidad de Buenos Aires. Facultad de Medicina Wenzhou Medical University
Departamento de Biología Celular Zhejiang, China
Cátedra II de Histología
and Wing-Yee Lui
CONICET-Universidad de Buenos Aires School of Biological Sciences
Instituto de Investigaciones Biomédícas (INBIOMED) The University of Hong Kong
Buenos Aires, Argentina Hong Kong, China

Pranitha Jenardhanan
Centre for Bioinformatics
Pondicherry University
Pondicherry, India
Contributors xiii

Livia Lustig Christopher L. Robinson


Department of Cellular Biology Department of Surgery
University of Buenos Aires Weill Cornell Medical College
and New York City, New York
Cátedra II de Histología
and Peter N. Schlegel
Instituto de Investigaciones Biomédícas (INBIOMED) Department of Urology
Buenos Aires, Argentina Weill Cornell Medicine
New York City, New York
Premendu P. Mathur
KIIT University Bruno Silvestrini
Odisha, India S.B.M. Srl Pharmaceuticals
Rome, Italy
Karl Mueller
Department of Cell Biology and Biochemistry Charles E. Smith
Texas Tech University Health Sciences Center McGill University
Lubbock, Texas Department of Anatomy and Cell Biology
Montreal, Canada
Manjunatha K. Nanjappa
Department of Physiological Sciences Peter G. Stanton
University of Florida Hudson Institute of Medical Research
Gainesville, Florida and
Department of Molecular and Translational Science
Liza O’Donnell Monash University
Hudson Institute of Medical Research Clayton, Australia
and
Department of Molecular and Translational Science Elizabeth I. Tang
Monash University The Mary M. Wohlford Laboratory for Male Contraceptive
Clayton, Australia Research
Center for Biomedical Research
Regiana L. Oliveira Population Council
Department of Anatomy and Cell Biology New York City, New York
McGill University
Montreal, Canada Sergei G. Tevosian
Department of Physiological Sciences
Manivel Panneerselvam University of Florida
Centre for Bioinformatics Gainesville, Florida
Pondicherry University
Pondicherry, India Maria Susana Theas
Department of Cellular Biology
Jennifer R. Panizzi University of Buenos Aires
Department of Anatomy, Physiology, and Pharmacology and
Auburn University Cátedra II de Histología
Auburn, Alabama and
Instituto de Investigaciones Biomédícas (INBIOMED)
Cecilia Valeria Pérez Buenos Aires, Argentina
CONICET – University of Buenos Aires
and William H. Walker
Instituto de Investigaciones Biomédícas (INBIOMED) Department of Obstetrics, Gynecology and Reproductive
Buenos Aires, Argentina Sciences
University of Pittsburgh School of Medicine and Magee-
Nahid Punjani Womens Research Institute
Department of Surgery Pittsburgh, Pennsylvania
Division of Urology
University of Western Ontario
London, Canada
xiv Contributors

Fei Wang Kandis Wright


Institute of Basic Medical Sciences Department of Cell Biology and Biochemistry
Chinese Academy of Medical Sciences Texas Tech University Health Sciences Center
School of Basic Medicine Lubbock, Texas
Peking Union Medical College
Beijing, China Fang Yang
Department of Biomedical Sciences
P. Jeremy Wang University of Pennsylvania
Department of Biomedical Sciences Philadelphia, Pennsylvania
University of Pennsylvania
Philadelphia, Pennsylvania Lan Ye
State Key Laboratory of Reproductive Medicine
Qing Wen Nanjing Medical University
The Mary M. Wohlford Laboratory for Male Contraceptive Nanjing, China
Research
Center for Biomedical Research Leping Ye
Population Council The Second Affiliated Hospital and Yuying Children’s Hospital
New York City, New York Wenzhou Medical University
Zhejiang, China
Chris KC Wong
Croucher Institute for Environmental Sciences Ke Zheng
Department of Biology State Key Laboratory of Reproductive Medicine
Hong Kong Baptist University Nanjing Medical University
Hong Kong, China Nanjing, China
Golgi apparatus regulation
of differentiation
1
A case study for male germ cells of the rat testis
LOUIS HERMO, REGIANA L. OLIVEIRA, CHARLES E. SMITH, CATHERINE E. AU,
and JOHN J. M. BERGERON

INTRODUCTION from this seminal paper is celebrated today as the found-


The Golgi apparatus is found in each of the estimated ing paradigm in modern stem cell biology.4
more than 200 different cell types that make up the human
body.1 Golgi apparatus structure visualized by standard DEFINING SPERMATOGENESIS
thin sectioning techniques at the electron microscope Spermatogenesis is defined as a temporal event whereby
(EM) level, which reached its apogee in the latter part of germ cells in the seminiferous epithelium of the testis
the last century, can identify solely and unambiguously undergo a differentiation process into maturing sperma-
any of the differentiated cell types in a given species. tozoa.3,5–10 It occurs over a span of several weeks and is
Remarkably, its structure is strictly conserved with the characterized by a period of mitotic, meiotic, and post-
number and size of cisternae in Golgi stacks and overall meiotic events.3,6 During the mitotic or proliferation
size of the Golgi apparatus under strict control for each of phase, spermatogonia undergo several mitotic divisions
the many differentiated cell types of a given organ of the to gradually increase their numbers and eventually form
human body. This conjecture has been illustrated here for spermatocytes. The latter undergoing two meiotic divi-
the Golgi apparatus using the well-described differentia- sions form haploid spermatids, which metamorphose into
tion of male germ cells in the adult rat testis. spermatozoa involving a series of dramatic structural and
functional modifications defined as the 19 steps of sper-
SPERMATOGENESIS: A DISCOVERY BASED miogenesis, a subdivision of spermatogenesis.10
ON THE GOLGI APPARATUS Cross-sectional profiles of the numerous seminiferous
In the mid-1950s, detailed light microscope (LM) observa- tubules comprising the testis reveal several concentric lay-
tions elucidated the complexity of the differentiation path- ers of germ cells (Figures 1.2 and 1.3).11 These layers consist
way of male germ cells in seminiferous tubules of the testis, of spermatogonia and early spermatocytes localizing to
a process referred to as spermatogenesis. The periodic acid the base of the epithelium, more advanced spermatocytes
Schiff (PAS) technique2 was the breakthrough that enabled and early spermatids in the mid region, and late sperma-
both the steps of germ cell differentiation to be uncovered tids prior to release closest to the lumen. Through the PAS
as well as the discovery of stem cells (Figure 1.1). stain, the different germ cells undergoing differentiation
The PAS stain is specific to vicinyl hydroxyls and when associate with each other to form specific cellular associa-
applied to the testis reveals staining of the Golgi appara- tions or stages of their differentiating process. In the rat, 14
tus and Golgi derived acrosome of germ cells.3 The reac- different stages were identified and represented by roman
tion due to carbohydrate modifications on glycoproteins numerals I–XIV. Moreover, in any given cross-sectional
is modulated by Golgi localized glycosyltransferases. This tubule profile constituting a specific stage, the different
method provides a precise mapping of how germ cells germ cell layers were all at the same step of development,
associate with each  other in cross sections of seminifer- defining what was termed as a generation of germ cells.
ous tubules of the testis, as first described by Leblond and In any given tubular cross-sectional profile, four or five
Clermont3 (Figure 1.2). distinct generations of germ cells could be seen, depend-
In the seminiferous epithelium, germ cells at different ing on the particular stage of the seminiferous epithelium
steps of differentiation associate with each other to form at that precise area of the tubule. Stages I–VIII show five
specific cellular associations referred to as stages of the generations of germ cells, while stages IX–XIV present
cycle of the seminiferous epithelium. The PAS method was four3,6,10 (Figures 1.2, 1.3).
the first test in defining the significance of the Golgi appa- The different generations of germ cells were shown to
ratus in understanding the differentiation process of any be constant for each of the 14 stages of the adult rat tes-
organ undergoing renewal. Even more important was the tis.3 Moreover, each generation defining a distinct stage
first identification of stem cells identified as a constantly (e.g., stage I) in the living organism had to be advancing
replenished population undergoing cell division to gener- in differentiation to give rise to the next stage (e.g., stage
ate other stem cells as well as those destined for further II), until all 14 stages appeared in that given area of the
differentiation. The stem cell renewal theory that resulted tubule. Considering that spermatozoa have to be released

1
2  Golgi apparatus regulation of differentiation

Glycoconjugate

HO2SHN HIO4 RCH (OH) O2SHN

C NH2 + 2R-CHO C NH

HO2SHN Aldehyde RCH (OH) O2SHN


SO3H
Schiff’s reagent Quinoid chromophore (red ~ purplish blue)

Figure 1.1  Chemical ingredients for the Schiff reaction. (Reproduced with permission from Wako Chemicals USA.)

Stage I Stage II Stage VIII Stage IX


19
RB
15
16
1
8
2 9
P P

S L
A PI
Stage III In In Stage X A
Stage IV Stage XI

10
3 4 11
P
16 17
P
L

In A A L
A Stage XII A Stage XIII
InM
Stage VI Di
Stage V

6
Di
518
Di
17
P P
Z
A
B B BM
Stage VII Stage XIV
18
14
7
IM
IIM
P Di
S II
P
A PI
A

Figure 1.2  Schematic drawings of portions of seminiferous tubules, each representing one of the 14 well-defined stages of the
cycle of the seminiferous epithelium as revealed from PAS-hematoxylin stained preparations. Stages are represented by roman
numerals. Spermatogonia: type A, intermediate (In) and B; InM and BM, mitosis of In and B spermatogonia; preleptotene (Pl), lep-
totene (L), zygotene (Z), pachytene (P), diplotene/diakinesis (Di) spermatocytes and secondary spermatocytes (II). IM and IIM, first
and second meiotic divisions. Arabic numbers represent the 19 steps of differentiation of spermatids undergoing spermiogenesis.
RB, residual body. (Reproduced with permission from American Physiological Society. Clermont, Y. Physiol Rev. 1972;52(1):198–236.)
Golgi apparatus and acrosome formation  3

58
15 16 16 17 17 18 19 19

1 2 3 4 5 6 7 8 9 10 11 12 13 14

P P P P P P P P P P P P Di II

In In In M B BM PI PI L L L Z Z P
A4M
A1 A1 A1 A1 A1 A1 A1 A1M A2 A2 A2M A3 A3M
I II III IV V VI VII VIII IX X XI XII XIII XIV
40 17 10 12 29 27 58 30 7 7 7 31 18 15

Figure 1.3  Schematic diagram of the different cellular associations or stages of the cycle of the seminiferous epithelium in adult
rats. Drawings as revealed from PAS-hematoxylin stained preparations. The stages are indicated by roman numerals I–XIV. The verti-
cal columns illustrate the different cell types present in each stage. The cells illustrated are spermatogonia: types A1–4, intermediate
(In), and type B; spermatocytes: preleptotene, leptotene, zygotene, pachytene, and diplotene spermatocytes; and spermatids at
steps 1–19 of spermiogenesis. M next to spermatogonia indicates that these cells undergo mitosis, while II at stage XIV indicates
secondary spermatocytes undergoing meiotic division. (Reproduced with permission from Wiley. Dym, M and Clermont, Y. Am J
Anat. 1970;128:265–282.)

periodically from the seminiferous epithelium, the idea of steps with 19 being identified in rats; the entire process
a cycle of the seminiferous epithelium was proposed. The extends over 22.7 days (Figure 1.3). During spermiogenesis,
sequential occurrence of all 14 stages that occur over time many genes/proteins are expressed, some for the first time,
in a given area of the tubule was defined as a cycle of the with implications in the diverse dynamic events occurring
seminiferous epithelium3 (Figure 1.3).11 at this time point of germ cell development. These events
Using a variety of techniques, the duration of each stage include elaboration of the acrosome and flagellum, nuclear
of the cycle of the seminiferous epithelium was calculated condensation, and modification of specific organelles, such
and shown to be constant but varying in time for differ- as the Golgi apparatus and endoplasmic reticulum (ER),
ent stages. Some stages lasted for a short period of time leading to the formation of spermatozoa destined to be
(i.e.,  stages IX and X [9 hours]), while others lasted lon- released from the seminiferous epithelium with the cyto-
ger (i.e., stage VII [58 hours]) (Figure 1.3). The duration plasmic droplet attached but the residual body detached
of each stage was calculated as well as the total duration and subject to disposal by the Sertoli cells.9,10,14,15
of the cycle; in rats it lasts about 13 days and that of the
complete process of spermatogenesis about 65 days.6,12 GOLGI APPARATUS AND ACROSOME FORMATION
The entire process of spermatogenesis varies for different The most conspicuous event that defines spermatids is the
species and even within similar species. A comprehensive formation of the acrosome, a membrane-bound lysosomal
analysis of the stages of the cycle of the rat and several structure made by the Golgi apparatus. Steps of spermatid
other species is provided in a complete book on the testis.13 differentiation as defined by Golgi identification and acro-
some formation were discovered through the PAS reac-
DEFINING SPERMIOGENESIS tion, which alone enabled a distinction in the rat among
Spermiogenesis is a timely event defining the last phase the transformation of early round spermatids (steps 1–8)
of germ cell differentiation. It begins after the second to late elongating spermatids (steps 9–19).3,10 The PAS
meiotic division of spermatocytes with the formation of staining of the Golgi apparatus and the acrosome is found
haploid spermatids and follows them through their meta- in the original illustration from Yves Clermont,16 shown
morphosis into spermatozoa. It is subdivided into distinct here in Figure 1.4.
4  Golgi apparatus regulation of differentiation

2 3
6

8
10

13 12

15
19b

17 19a RB

Figure 1.4  Schematic representation of the role of the Golgi apparatus in acrosome formation during spermiogenesis as visual-
ized by the PAS method. At step 2, the Golgi apparatus, seen as a small magenta-colored bubble, has formed two small dense proac-
rosomic granules placed next to the nucleus, which at step 3 have fused together to form a single larger dense granule. At step 6 the
acrosome has grown in size and covers a sizeable portion of the nucleus. At step 8 the nucleus occupies an eccentric position next to
one pole of the nucleus, while the Golgi apparatus becomes spherical in shape and floats freely in the cytoplasm. From steps 10–19
the nucleus becomes gradually compacted, elongated, and sickle-shaped. The spherical Golgi apparatus is still evident as a reddish
bubble in the vast cytoplasm of these cells. At step 19a, the cytoplasm has displaced itself along the tail to form the residual body
seen hanging as a sac onto the sperm at the junction of its head and tail. At step 19b, the residual body has completely liberated
itself from the sperm, while a small dilated focal area of cytoplasm at the neck region of the tail constitutes the Hermes body (cyto-
plasmic droplet). It reveals the presence of a small reddish remnant of the Golgi apparatus, as does the residual body (RB) depicted
in step 19a. (Reproduced with permission from Wiley: Hess, RA et al., Andrology, 2015; 3(6):1015–1021.)

As visualized in a LM, small proacrosomic granules After completion, the nucleus and overlying acrosome
derived from the hemispherical Golgi apparatus appear of step 7 spermatids shifts from its central position in the
adjacent to the spherical nucleus from steps 1–3 (Figure cytoplasm to become eccentric and apply itself directly to
1.4). These granules fuse with one another and with con- one pole of the plasma membrane. This event constitutes
tinued production create a larger acrosome (step 4). The the formation of step 8 spermatids and coincides with
latter gradually applies itself to the nuclear surface, and the change in shape of the Golgi apparatus from crescen-
with time and progressive enlargement, it spans more and tic and elongated and closely apposed to the acrosome to
more of the nuclear surface (steps 5, 6) until its formation spherical and removed from it. The spherical Golgi there-
is completed, at which time it covers almost half of the after migrates and floats freely in the expanded cytoplas-
nuclear circumference (step 7). mic lobe of differentiating elongating spermatids.
Defining Golgi proteins localized to spermatogonia: TMED7/p27, TMED4/p25, and TM9SF3  5

During the ensuing decades, the paradigm of the fuse with the acrosome.29,45,46 Two types of β-COP, coat-
“cycle”  and the description of germ cells recognizable omer protein complexers, are found associated with the
by standard light microscopy formed the foundation Golgi apparatus and in the acrosomal membrane of sper-
for the accretion of knowledge on the subcellular struc- matids.46 EM cytochemical markers indicate that nico-
tures and molecular changes accompanying germ cell tinamide adenine dinucleotide phosphatase (NADPase)
differentiation.10,14,17–23 localizes to middle cisternae, thiamine pyrophospha-
tase (TPPase) to the transmost cisternae, and cytidine
GOLGI APPARATUS STRUCTURE AND FUNCTION 5-monophosphatase (CMPase) to the TGNs (Figure
DISTINGUISHES SPERMATOGONIA FROM 1.6).24,48,49 Because the acrosome also shows a cytochemi-
SPERMATOCYTES AND SPERMATIDS cal reaction for CMPase, TPPase, and other phosphatases
The structure of the Golgi apparatus of spermatogonia and such as orotidine 50-monophosphatase, it is suggested
spermatocytes has been described by EM analysis10,24 but that these respective Golgi cisternal elements contribute
the majority of earlier studies were done on spermatids directly to acrosome formation (Figure 1.6).14,24
during acrosome formation (i.e., steps 1–7 of spermiogen-
esis10,14,24–29). During this time, the hemispherical Golgi MOLECULAR DISSECTION OF GOLGI APPARATUS
is closely associated with one pole of the nucleus as it is DURING SPERMATOGENESIS
actively involved in laying down the membrane bound Following an unbiased strategy known as the cell map,50
acrosome onto its surface. Thereafter, beginning at step 8, a profile of Golgi protein expression was determined in
the Golgi apparatus becomes spherical and migrates freely germ cells.51 First, a reproducible method to isolate Golgi
into the cytoplasmic lobe.9,29 This event coincides with apparatus selectively from male germ cells of whole adult
cessation of transcription in the nucleus30 and with the rat testis homogenates was developed.51 Then by quantita-
replacement in the nucleus of histones by protamines.31 tive proteomics, a characterization of over 1000 different
Nevertheless, in elongating spermatids the Golgi appara- proteins in this subcellular fraction was elucidated, with
tus continues to be involved in glycoprotein synthesis.32,33 several representing novel Golgi proteins as well as known
During steps 16–18 spermatids, the Golgi apparatus frag- Golgi proteins.
ments and becomes vacuolated,29 with individual flattened Several of the proteins identified were characterized
cisternae dispersing into the cytoplasm.34 At the last step for the first time requiring the generation of antibodies to
of spermiogenesis, two cytoplasmic distensions of the step a subset to assure that they represented bona fide Golgi
19 spermatid become prominent; that is, the cytoplasmic proteins.51 In total, with monospecific antibodies to 20
droplet, recently renamed the Hermes body35 and residual different proteins obtained from different sources51 and
bodies. The dispersed Golgi cisternae congregate into the concluded to be Golgi localized by LM immunocytochem-
Hermes body (Figure 1.5).34 ical localization, a map of their expression profile during
Historically, the very first EM visualization of Golgi spermatogenesis was realized.
apparatus for any cell type was done for spermatids36 as The insight gained from the detailed analysis of this data
well as very early three-dimensional (3D) images of Golgi will be summarized by an illustration of spermatogene-
apparatus structure.28 The early spermatid Golgi appa- sis truncated to 30 of the 64 distinct cell types identified
ratus is a continuous structure with alternating com- (Figure 1.7). Here germ cells are represented undergoing
pact stacked and noncompact tubular interconnecting mitosis, meiosis, acrosome formation (steps 1–7), posta-
regions24,28,37 with an overall appearance in mammalian crosome Golgi migration (steps 8–18), and ending at the
cells to that of a ribbon. The term Golgi ribbon was first terminal step of spermiogenesis, step 19 spermatids when
introduced by Rambourg and Clermont38 and its usage has the Hermes body is formed (Figure 1.7).
been adopted universally by the scientific community to
define the conserved properties of Golgi apparatus struc- DEFINING GOLGI PROTEINS LOCALIZED TO
ture in all mammalian cell types.39–44 SPERMATOGONIA: TMED7/P27, TMED4/P25,
The structure of the early spermatid Golgi has been AND TM9SF3
described in several studies by Clermont et al.37 and visu- The periodic renewal of type A spermatogonia was recog-
alized diagrammatically (Figure 1.6).9 The stacked regions nized early in the last century,52 but the complex process
of the Golgi ribbon consist of a highly anastomotic Cis of spermatogonial renewal was only beginning to be fully
Golgi network (CGN), followed by nine closely apposed understood 50 years later. In the early 1950s, it was the
flattened cisternae. Morphologically distinct thickened inspirational work of Clermont and Leblond4 that first
cisternae appear on the trans face of the Golgi ribbon and defined stem cells and paved the way for all subsequent
form trans Golgi networks (TGNs). The latter are close significant contributions on their process of renewal that
to the developing acrosome apposed to the nucleus. The over the course of the next 60 years has yielded an exten-
TGNs often reveal a peeling off configuration from the sive amount of information.10,18,53–62
stack with dilated edges showing a bristle coat. The area While there are many descriptions of markers for undif-
in between the trans face of the Golgi apparatus and the ferentiated and differentiated spermatogonia,10 those that
developing acrosome contains vesicular and tubular pro- demarcate the Golgi apparatus of these cells are clearly lack-
files decorated with fuzz or bristle coats, some of which ing. Three of the 20 proteins characterized by Au et al.,51
6  Golgi apparatus regulation of differentiation

An

CD

S
CD CD
Step 19 CD

RB

Step 18

IS
Sertoli Caput CD
G CL ED
cell
RT

Tubule

Step 10 Corpus

N
Testis
AS
G
Step 7
Cauda

Figure 1.5  Diagrammatic representation of the shape, position, and fate of the Golgi apparatus of early and late spermatids dur-
ing spermiogenesis and formation and fate of the cytoplasmic droplet (CD) of sperm as they pass through the epididymal duct. From
steps 1–7, the Golgi (G) forms the acrosomic system (AS) applied to the nucleus (N). From steps 8–16, the Golgi becomes spherical
and occupies the cytoplasmic lobe (CL). In steps 17 and 18, the Golgi apparatus undergoes fragmentation whereupon it is seen as
many randomly dispersed individual Golgi saccular elements (S) floating freely in the CL. At step 19 the Golgi cisternae congregate
into the CD while other organelles enter the residual body (RB), which is phagocytosed by Sertoli cells. The CD is positioned at the
neck region of the sperm flagellum as it forms in the testis at step 19 and remains there as sperm pass through the rete testis (RT),
efferent ducts (ED), and initial segment (IS). In the caput epididymidis, the CD displaces itself along the tail to the annulus (An). In
the corpus it detaches from the sperm leaving most sperm free of CDs in the cauda. (Reproduced with permission from Oko, R et al.,
J Cell Biol. 1993;123(4):809–821.)

although widely distributed throughout spermatogenesis, The TMEDp24 family of Golgi (GOLD domain) mem-
clearly labeled the Golgi apparatus of spermatogonia. These brane proteins regulate trafficking, cargo selection, and
were two members of the transmembrane emp24 domain lipid makeup of Golgi-derived carriers.63–66 They are regu-
(TMEDp24) family of Golgi (GOLD domain) integral lated during differentiation and development in many cell
membrane proteins, TMED4/p25 and TMED7/p27, along types.67–69 Hence TMED4/p25 and TMED7/p27 proteins
with one member of the TM9SF (transmembrane 9 super may regulate germ cell differentiation with their expres-
family) family of proteins, TM9SF3 (Figure 1.8a). sion in the Golgi apparatus of germ cells commencing in
MG160 and GBF1  7

NCZ

VA ER

W VA

CIS-element S
S
TGN
NADPase + W
E
M R
TPPase + ER
CMPase +

E
R
ER

Acrosome

Head cap

Nucleus

Figure 1.6  Schematic representation of the Golgi apparatus of a step 7 spermatid. Represented are stacks of Golgi cisternae
(S) forming the compact zone and dilated tubules extending from the cisternal edges serving to join adjacent stacks that form the
noncompact zone (NCZ). The Golgi stack consists of an anastomotic tubular network on the cis face called the CIS-element, mid cis-
ternae stained for NADPase, a trans cisterna stained for TPPase, and two CMPase+ cisternae called the trans Golgi networks (TGN’s).
The latter reveal bristle-coated buds at their edges and a peeling-off configuration. Endoplasmic reticulum (ER) is seen overlying the
cis element, applied to trans cisterna and the TGN, as well as in the NCZ and core or medulla (M) of the Golgi apparatus. Pan-shaped
spaces of the cis cisternae form wells (W). Vesicular aggregates (VA) also appear on the cis face of the Golgi apparatus. (Reproduced
with permission from Clermont, Y et al. Cell biology of mammalian spermiogenesis. In Desjardins C and Ewing L, eds. Cell and
Molecular Biology of the Testis. New York: Oxford University Press, 1993.)

spermatogonia but also extending into late spermatids enabling diploid cells to form haploid cells. Meiosis serves
(Figure 1.8a and b). important cellular activities such as DNA repair, genetic
Three members of the TM9SF family of proteins of exchange, and synapsis of homologous chromosomes,
unknown function were noted in the isolated germ cell with many genes implicated in this process.8,10,72–82
Golgi fraction, with TM9SF3 localized.51 In yeast, a In addition to changes in nuclear events, pachytene
related protein to TM9SF3, emp70 (endomembrane pro- spermatocytes undergo a dramatic increase in size dur-
tein 70), has been considered as endocytic and involved ing meiosis coincident with major changes to the size and
in eisosome regulation (a subdomain of the plasma mem- structure of their spherical Golgi apparatus.83,84 An exam-
brane marked for endocytosis).70 In mammalian cells, ple of the Golgi apparatus of spermatocytes as seen in the
emp70 has been considered to be involved in endosome EM is illustrated in Figure 1.9a.
to Golgi trafficking.71 In our study, 51 TM9SF3 is a Golgi
integral membrane protein uncovered for the first time as MG160 AND GBF1
a Golgi marker not only to germ cells (Figure 1.8a and c), While the three proteins localized to the Golgi apparatus
but also with a ubiquitous expression in many other cell of spermatogonia continue to be expressed in spermato-
types, such as Sertoli and Leydig cells (Figure 1.8c), epi- cytes, additional proteins first appear during spermato-
didymal epithelial cells, 51 and neurons of the brain (not cyte development, such as MG160 (Golgin160/MEA-2/
shown). Golga3) and GBF1 (GTPase involved in Golgi vesicular
traffic), with the latter extending into early spermiogenesis
DEFINING GOLGI PROTEINS LOCALIZED TO (Figure 1.10a).
SPERMATOCYTES: MG160, GBF1, GL54D, AND GIANTIN MG160 is a coiled-coil protein and part of the Golgin
Meiosis is unique to germ cells and involves one round of family.85 It is normally expressed in all somatic cells and
DNA replication followed by two consecutive cell divisions is a universal Golgi marker.86,87 The protein is known
8  Golgi apparatus regulation of differentiation

the null mutant may be speculated to be due to an essen-


tial and nonredundant role during meiosis. MG160 has
II 1 2 been implicated in tethering Golgi mini stacks together,91
3
Di and in this role it may be implicated in the doubling in
4 size of the Golgi apparatus of spermatocytes and its even-
P(XII) Mitosis 5 tual partitioning into four Golgi apparatuses with each
(A–B)
6
one destined for the four resulting haploid spermatids.83
P(VII)
Meiosis Several of the Golgi markers used in our study51 readily
P(I) (PL–II) 7 visualized the segregated Golgi fragments during meiosis
Acrosome (Figure 1.8c). This was also visualized by EM, where a dis-
Z
(1–7) 8 tinct Golgi apparatus was noted at opposite poles of the
L nucleus of a spermatocyte at stage XIV of the cycle during
Postacrosome
PL
(8–18) 9 its meiotic division (Figure 1.9b).
Hermes biogenesis GBF1 (Golgi-specific brefeldin A-resistance guanine
B (19) nucleotide exchange factor) regulates vesicular traffick-
In ing for active secretion.92 The expression of GBF1 during
A spermatogenesis marked the first evidence for a differen-
10 tial expression of this protein in any cell differentiation
pathway. GBF1 expression begins at preleptotene sper-
11 matocytes and extends into early spermiogenesis (Figure
1.10a and c). Hence, the burst of GBF1 expression in sper-
12 matocytes and early spermatids likely reflects their high
19 18 17 16 15 14 13
secretory activity. In fact, forerunners of the proacrosomic
granules first appear in zygotene primary spermatocytes
Figure 1.7 Thirty representative germ cell types are and extend into late pachytene spermatocytes,74,93–95 with
depicted as they undergo differentiation during spermato- acrosome formation advancing through the first half of
genesis. Illustrated are the mitotic spermatogonia types A, spermiogenesis.10,14
intermediate (In) and B; spermatocytes undergoing meiosis:
preleptotene (PL), leptotene (L), zygotene (Z), pachytene (P),
GL54D AND GIANTIN
diplotene (Di), and secondary spermatocytes (II). Spermatids The most abundant protein of our isolated testis germ cell
evolve through 19 steps of spermiogenesis that can be sub- Golgi fraction was a protein designated from the annota-
divided into acrosome formation (steps 1–7), postacrosome tion of UniProtKB/Swiss-Prot, as glycosyltransferase 54
events (steps 8–18) and Hermes body (cytoplasmic drop- domain-containing protein (GL54D), an MGAT4 family
let) formation (step 19). (Reproduced with permission from member also known as GNT1IP-S.96 GL54D, an integral
Mol Biol Cell: Au, CE et al., Mol Biol Cell. 2015; 26(22):4015–4032 membrane glycoprotein like TM9SF3, is a Golgi marker.
DOI:10.1091/mbc.) GL54D was uncovered for the first time at the endogenous
protein level by the mass spec characterization of our iso-
lated testis Golgi fraction.51 However, whereas TM9SF3 is
ubiquitous to all cells as a universal Golgi marker, GL54D
to interact with Golgi membranes through RAB6 and is germ cell specific, revealing a differential expression
Arf1.88 In the testis, MG160 expression demarcates the profile during spermatogenesis. GL54D is first detected
boundary between spermatogonial renewal and spermio- in pachytene spermatocytes at stage VII of the cycle with
genesis (i.e., spermatocyte differentiation) with expres- expression extending up to step 16 spermatids (Figure
sion exclusive to spermatocytes (Figure 1.10b). MG160 1.11a, b, and c). GL54D is a type II, N-linked integral mem-
has long been known to be a prominent gene product of brane terminally glycosylated glycoprotein largely facing
male germ cells89; however, during spermatogenesis it the lumina of Golgi cisternae and with sole expression in
is differentially expressed. A null mutation in the gene the testis, where it is expressed as the most abundant Golgi
abolishes protein expression in mice with a phenotype protein of germ cells.
of disrupted germ cell development during late meiosis, Giantin (GOLGB1) was the most abundant protein of
coinciding precisely with its localization to meiotic sper- the isolated testis Golgi apparatus fraction in the tethering
matocytes. Sperm development is markedly decreased docking and fusion category and at far higher concentra-
in these mice and the resultant few sperm are infertile. tions than in liver Golgi fractions.51 Like MG160, giantin is
When the gene is truncated in a transgenic mouse model a Golgin involved in the tethering and vesicular trafficking
exactly as for the MG160 null mutant, pachytene sper- functions of the Golgi apparatus.97 During spermatogen-
matocytes are depleted.90 esis, giantin has been revealed in pachytene spermatocytes
Although MG160 (membrane protein of the Golgi but its precise expression pattern during spermatogenesis
apparatus) is expressed and localized to Golgi apparatus has not been deduced.98–100 Nevertheless, the coincidence
in all somatic cells,86,87 the male germ cell specific effect of between giantin, a Golgi resident protein on the cytosolic
GRASP55 and GPP34  9

(a) (b)

II 1 2
3 P Lu
Di S
4
P(XII) Mitosis 5
(A–B) 10 µm
P(VII) 6
Meiosis
(PL–II) (c)
P(I) 7 S
Acrosome
Z (1–7) L
8
L Postacrosome P
(8–18)
PL 9
Hermes biogenesis P
(19)
B
10 µm
In
A
10

11
TM9SF3; TMED4/p25
12 TMED7/p27
19 18 17 16 15 14 13

Figure 1.8  (a) Wheel depicting the expression profile of TMED7/p27, TMED4/p25, and TM9SF3. All three proteins span most of
spermatogenesis from spermatogonia until step 16 spermatids, with the exception of TMED7/p27 that reappears at step 19 sperma-
tids. (b) LM-IHC of a seminiferous tubule reactive for TMED7/p27 at stage VII of the cycle. Reactivity is noted in the spherical Golgi
apparatus of spermatocytes (P), early round spermatids (small arrowheads), late step 19 spermatids (curved arrows), and Sertoli
cells (S). (c) LM-IHC of tubules at stages XIV and XIII of the cycle reactive for TM9SF3. Golgi reactions are noted for pachytene sper-
matocytes (P), Sertoli cells (S), Leydig cells (L), and late spermatids (large arrowheads). At stage XIV, spermatocytes undergo meiosis
and reveal multiple Golgi bodies (boxes).

side and rims of Golgi cisternae,85 and GL54D, a luminal study were traditional Golgi markers, but others had
Golgi protein, and with both being expressed in pachy- unexpected Golgi localizations.51 EM images of acro-
tene spermatocytes, suggests that a coordination exists some formation by the Golgi apparatus are illustrated in
between these two proteins in protein modifications Figure 1.9c–e.
within the Golgi apparatus. In addition to TMED7/p27, TMED4/p25, TM9SF3,
GBF1, and GL54D, all with widespread distributions, five
DEFINING GOLGI LOCALIZED PROTEINS IN EARLY additional proteins with Golgi characteristics (Grasp55,
SPERMATIDS DURING ACROSOME FORMATION GPP34, Man2α1, ManIIX, and LAMAN) revealed onset of
The acrosome is a lysosomal structure that is formed by Golgi expression in early spermatids during acrosome for-
the Golgi apparatus during early spermiogenesis with an mation (Figure 1.12a). Examples are illustrated for GRASP
unprecedented specificity for adherence to the nuclear 55 (Figure 1.12b) and GPP34 (Figure 1.12c).
surface of spermatids. As such it follows the contours
and modifications that occur to the shape of the head GRASP55 AND GPP34
and nucleus of spermatids during late steps of spermio- GRASP55 (Golgi reassembly associated protein) has been
genesis.13 At the time of fertilization, sperm undergo the implicated in the stacking of Golgi cisternae.102–104 Its
acrosome reaction, an event unique in the life of a sperma- expression first appears after meiosis with Golgi immuno-
tozoon. Acrosomal contents released during fertilization reactivity to steps 1–7 spermatids, and with no localization
promote sperm penetration through the oocyte’s invest- to Sertoli or Leydig cells of the testis, all of which reveal
ments, leading to sperm-egg interaction and fusion.101 stacked Golgi cisternae.105,106 Alternative functions to those
Acrosome formation is an intense period of protein of cisternae stacking have been proposed for GRASP55,
synthesis marked by the expression of a large number such as unconventional protein secretion and Golgi rib-
of proteins localized to the Golgi apparatus as it forms bon formation.107 Unlike typical lysosomes, the acrosome
the acrosome. Some of the proteins uncovered in our has to bind to the nuclear surface of spermatids and this
10  Golgi apparatus regulation of differentiation

UNEXPECTED ER RESIDENT PROTEINS WITH GOLGI/


N
ACROSOME LOCALIZATIONS: SAPRETICULIN, PDILT,
AND UBXD8
G
N Remarkably, three normally ER-located proteins
G
were also Golgi localized during acrosome formation
S (Figures 1.12a and 1.13a).51 They included UBX domain
S (b)
containing 8 protein (UBXD8/FAF2), sapreticulin
(Figure 1.13b), and protein disulphide isomerase-like
(a) S protein of the testis (PDILT) (Figure 1.13c).
Sapreticulin is a saposin B domain containing ER
A
S
luminal protein with the canonical ER targeting KDEL
N tail.107 The only other protein known to have saposin
(d) domains is prosaposin, normally targeted through the
S secretory pathway to lysosomes where it is converted by
selective proteolysis to individual saposins A, B, C, and
S S D that are needed to solubilize different glycolipids for
degradation. Intriguingly, prosaposin is not expressed
in male germ cells.110 Hence, sapreticulin remains as the
A
only saposin domain expressing protein in spermatids.
N
(c) (e) N During acrosome formation, sulfoglycolipids are highly
expressed and localized to the Golgi apparatus.111 The
major sulfoglycolipid is seminolipid112 and it is pre-
Figure 1.9  EM of the Golgi apparatus of different germ
dicted to be the potential client of sapreticulin. Hence, a
cells. (a) Golgi apparatus of a mid pachytene spermatocyte. N:
speculated role for sapreticulin is in sulfoglyolipid orga-
nucleus; S: stacks of Golgi cisternae. (b) Spermatocyte under-
nization of the Golgi of spermatids during acrosome
going meiotic division revealing a Golgi apparatus (G) at
formation.
opposite poles of the nucleus (N). (c) Stack of Golgi saccules
The testis-specific PDILT performs a specialized
(S) of step 1 spermatid shows several proacrosomic granules
chaperone function by liaising with partner proteins
(arrowheads) next to the nucleus (N). (d) Forming acrosome
in disulfide-dependent complexes within germ cells
(A) at step 4 spermatid is applied to the nucleus (N). (e) Step 7
of the testis.113 The PDI family member erp44 (endo-
spermatid shows a Golgi stack (S) and expanded acrosome (A)
plasmic reticulum resident protein) is now accepted to
applied to the nucleus (N). Original magnifications: 9a: 22,500×;
be Golgi localized in most cell types but whose local-
9b: 38,000×; 9c–e: 48,750×.
ization changes to ER depending on its cargo.114,115
A known PDILT client in germ cells is the acrosome
located membrane protein ADAM3 (a disintegrin and
may involve unconventional secretion pathways to accom- metalloprotease).113,116 However, after localizing to the
modate this novel function for the acrosome. Of note is the Golgi and acrosome from steps 1–7 (solid brown line,
fact that of all the five Golgi proteins indicated above, only Figure  1.13a), PDILT relocalizes to the cytoplasm (ER)
GRASP55 does not become expressed again during later where it is present from steps 8–19 (stippled brown line,
steps of spermiogenesis and in the forming Hermes body, Figure 1.13c).113,117 Hence, it is the PDILT client proteins
where Golgi cisternae do not form a stack (Figure 1.12a). that likely dictate its localization to specific organelles of
GPP34, now known as GOLPH3 (Golgi phophopro- the secretory pathway.
tein), has a well-defined role in the regulation of Golgi The ubiquitin-like domain containing protein UBXD8
ribbon structure,108 and this property may also be shared (FAF2), an ER associated degradating protein, is promi-
with GRASP55.107 During steps 1–7 spermatids, the Golgi nent in the cytoplasm (ER) of spermatocytes (prelepto-
apparatus is crescent in shape with the TGNs exposed to tene to diplotene, stippled blue line, Figure 1.13a) but
the nuclear surface of early spermatids. This would allow relocalizes to the Golgi during acrosome formation
easy access of the proacrosomic granules to the nuclear (solid blue line, Figure 1.13a). In the ER of other cell
surface to which they bind. Defining the shape of the Golgi types, a function for UBXD8 in quality control to remove
ribbon may be in part the role of GOLPH3 and GRASP55, newly synthesized misfolded secretory proteins has been
but this would be limited to its expression profile during a proposed as well as in lipid droplet formation118 and
defined time point during spermatogenesis. GOLPH3 is not ER-associated protein degradation (ERAD).119,120 Its role
expressed from steps 8–18 but returns at step 19 spermatids in the Golgi and acrosome of early spermatids remains
to be included in the forming Hermes body (Figure 1.12a). to be determined.
Other unexpected proteins with Golgi/acrosome localizations  11

(a) (b)
L P

1 PL
II 2 P
3
Di S
4 S
P(XII) Mitosis 5
(A–B)
P(VII) 6 10 µm
Meiosis
(PL–II) (c)
P(I) 7
Acrosome Lu
Z (1–7) 8
L Postacrosome A
(8–18) P
9 A
PL Hermes biogenesis A
(19) P
B
10 µm
In
A
10

11
GBF1
12
19 18 17 16 15 14 13 MG160

Figure 1.10  (a) Wheel depicting the expression profile of GBF1 and MG160. While both begin expression in PL spermatocytes,
MG160 is only expressed during spermatocyte development while GBF1 extends up to step 7 spermatids and then is absent until
it reappears at step 19. (b) LM-IHC of seminiferous tubules reactive for MG160. Reactive germ cells include the Golgi apparatus of
preleptotene (PL) and pachytene (P) spermatocytes. Sertoli (S) and Leydig (L) cells also show a Golgi reaction. (c) LM-IHC of GBF1 at
stage VI with Golgi reaction in pachytene spermatocytes (P) and Golgi (arrowheads) and acrosome (A) reactions in round spermatids.

OTHER UNEXPECTED PROTEINS WITH GOLGI/ somatic counterpart Hsc70,125 HSP70.2 may participate in
ACROSOME LOCALIZATIONS clathrin-coated vesicle trafficking known to be prominent
Several other proteins were unexpectedly localized to during acrosome formation.26
the Golgi apparatus and acrosome of steps 1–7 sperma- Using a different antibody obtained from Eddy,126
tids (Figures 1.12a and 1.13a). They include Annexin A6, HSP70.2 localized to spermatocytes and elongating sper-
HSP70.2 (Figure 1.13d), and FAM3C (Figure 1.13e). matids, including the sperm flagellum. The HSP70.2
Annexin A6 is a calcium-dependent membrane binding family of chaperone proteins may facilitate the intracel-
protein linked to membrane and cytoskeletal organization lular transport of proteins in elongating spermatids and
and cholesterol homeostasis.121 It may serve a function in likely supports plasma membrane remodeling and remov-
membrane trafficking122 that is highly active in acrosome ing surplus cytoplasm to maintain proper sperm head
formation. shapes.125–129
HSP70.2 (HspA2) is part of the heat shock protein A family with sequence similarity 3, member C
(HSP)70 family of molecular chaperones needed to sta- (FAM3C) (Figure 1.13e) is a protein of poorly under-
bilize otherwise labile proteins.123 The HSP70 family of stood function that is linked to GPP34 (GOLPH3) during
chaperones usually masks exposed hydrophobic residues Golgi extension and vesicular release130 and together with
on client proteins, preventing their aggregation and insol- GRASP55 and GOLPH3 may be required for maintaining
ubility. Our recent study51 represents the first localization the shape of the crescentic Golgi ribbon during acrosome
of the germ cell cytosolic chaperone HSP70.2 to the acro- formation.
some and Golgi apparatus of steps 1–7 spermatids (Figure Taken together, the recruitment of chaperones
1.13d), which we found to be antibody-specific.124 As for its (HSP70.2), protein-folding enzymes (PDILT), and ERAD
12  Golgi apparatus regulation of differentiation

(a) (b) Lu

II 1 2
3
Di
4
P(XII) Mitosis 5
(A–B)
P(VII) 6 10 µm
Meiosis
P(I) (PL–II)
7 (c)
Acrosome
Z (1–7) 8
L Postacrosome
(8–18) 9
PL Lu
Hermes biogenesis
B (19)

In 10 µm
A
10

11

12 GL54D
19 18 17 16 15 14 13

Figure 1.11  (a) Wheel depicting the expression profile of GL54D, which spans the Golgi apparatus from pachytene spermato-
cytes at stage VII until step 16 spermatids. (b) LM-IHC of seminiferous tubules reactive for GL54D at stage XIV. The upper tubule shows
Golgi reactions for pachytene spermatocytes (P), step 1 (small arrowheads), and step 14 (large arrowheads) spermatids. (c) LM-IF of
GL54D with reactions in Golgi of round (small arrowheads) and late (large arrowheads) spermatids. Lu, lumen.

quality control proteins (UBXD8) to the Golgi apparatus SEGREGATION OF GOLGI LOCALIZED PROTEINS
must reflect a requirement to assure the fidelity of sort- TO THE ACROSOME
ing and transport of proteins during the enormous bur- During acrosome formation (steps 1–7), 16 proteins
den of protein synthesis and trafficking during acrosome localized to the Golgi apparatus (Figure 1.14). Of the
biogenesis. 16, 12 (i.e., GRASP55, GBF1, ManIIX, Annexin A6,
GPP34, Man2α1, HSP70.2, Fam3C, PDILT, sapreticu-
Man2α1, ManIIX, AND LAMAN lin, UBXD8, and LAMAN) localized to both the Golgi
Man2α1 and ManIIX are both well-known Golgi manno- apparatus and developing acrosome. However, four
sidases. Man2α1 is a universal Golgi marker localizing to Golgi proteins (i.e., GL54D, TM9SF3, TMED4/p25, and
germ and Sertoli cells.100,132 ManIIX is a male germ cell spe- TMED7/p27) reacted only for the Golgi apparatus with
cific mannosidase partially overlapping with Man2α1 in its no evidence of reactivity for the acrosome (Figure 1.15).
specificity from steps 1–7 spermatids where both the Golgi Hence, a segregation of proteins exists during acrosome
and acrosome are reactive (Figure 1.12a). These sequence- formation whereby some proteins are excluded from the
related proteins likely have related functions in alpha man- acrosome but are present in the Golgi apparatus involved
nosidase trimming of high mannose containing N-linked in its formation.
oligosaccharides during acrosome formation.133–135 Acrosome formation is often explained by the matura-
LAMAN, also known as Man2β1, a major glycoprotein, tion model for Golgi biogenesis and traffic13; however, the
was also found in the Golgi apparatus and acrosome of segregation of the four Golgi markers is not consistent
steps 1–7 spermatids (Figure 1.14). LAMAN is a ubiqui- with this model. Alternative models to maturation have
tous lysosomal alpha mannosidase that cleaves alpha man- been proposed for Golgi traffic,137–140 and based on our
nosidase linkages in glycans.136 Its functional significance observations the spermatid Golgi is well suited to examine
in acrosome formation and spermatogenesis is unknown. the validity of these various models.
Golgi role in chromatoid body and MVB formation  13

(a) (b)
Lu
A
II 1 2
3 A
Di
4
A
P(XII) Mitosis 5
(A–B) 10 µm
P(VII) Meiosis 6
(PL–II) (c)
P(I) 7 Lu
Acrosome
Z A
(1–7) 8 A
L Postacrosome
(8–18)
PL 9
Hermes biogenesis A
B (19)
10 µm
In
A
10

11 GRASP55

12 ManIIX
19 14 13 Annexin A6; Man2 1; Fam3C; GPP34;
18 17 16 15
Sapreticulin; HSP70.2; LAMAN

Figure 1.12  (a) Wheel depicting the expression profiles of several Golgi localized proteins. Spanning the Golgi apparatus
from steps 1-7 spermatids are GRASP55, ManIIX, Man2α1, Annexin A6, Fam3C, GPP34, sapreticulin, HSP70.2, and LAMAN. While
GRASP55 is limited to steps 1–7 spermatids, ManIIX reappears from steps 15–19 while the others only reappear at step 19 spermatids.
(b) and (c) LM-IHC of GRASP55 (b) and GPP34 (c) at stage V showing Golgi (small arrowheads) and acrosome (a) reactions of the step
5 spermatids.

GOLGI ROLE IN CHROMATOID BODY AND MVB processing of the Golgi-derived components to the develop-
FORMATION ing acrosome. The imino sugar N-butyldeoxynojirimycin
During early spermiogenesis, the chromatoid body is (NB-DNJ) is an inhibitor of the N-linked oligosaccharide
closely associated with the Golgi apparatus. It is seen processing enzymes alpha-glucosidase I and II, with the
as a spongy-looking mass of electron dense granulo- potential to inhibit both glucosidases and glucosyltrans-
filamentous material with areas of low electron density ferases. Treatment with NB-DNJ results in accumulation
infiltrated with numerous medium-sized vesicles.14,141–143 of MVBs in the vicinity of the nucleus and presence of pro-
In late spermiogenesis, the chromatoid body migrates acrosomic granules that do not fuse together to form an
toward the centrioles occupying the opposite pole of the acrosome. Thus, acrosomal membrane proteins and cyto-
nucleus alongside the annulus of the flagellum.142,144 The solic acrosome-associated proteins become redirected to
vesicles are CMPase- and NADPase-positive, suggesting MVBs in affected testes.149,150
a contributing role from the Golgi apparatus.24 VPS26A/ Additionally, a role for the endocytic apparatus of early
VPS35-containing retromer vesicles have also been iden- spermatids has been defined in acrosome formation.
tified within the chromatoid body.145 Some of our Golgi Ubiquitin-specific peptidase 8 (USP8), a deubiquitinating
localized proteins during acrosome formation may also enzyme that works as a regulator of endosomal sorting and
have an impact on vesicles associated with the chroma- vesicle morphology, is highly expressed in male germ cells.
toid body. USP8 interacts with the spermatid protein endosomal-
Spermatid development is also marked by the presence sorting complex required for transport-0 (ESCRT-0) and
of endocytic structures (i.e., pits, endosomes, multivesicu- microtubule structures with both contributing to acrosome
lar bodies [MVBs], and lysosomes).146,147 Another proposed formation. USP8 links, via its MIT domain, the developing
role of the spermatid Golgi apparatus is to contribute to acrosome and vesicles to microtubules, which monitor both
MVB formation.24,148 Conversely, MVBs play a role in the acrosome assembly and shaping.151
14  Golgi apparatus regulation of differentiation

(a) (b) (c)


Lu
A Lu
II 1 2 * *
3
A A
Di 4 A A
P(XII) Mitosis 5
(A–B) 10 µm 10 µm
P(VII) Meiosis 6

P(I) (PL–II) (d) (e)


7
Acrosome A
Z (1–7) 8 Lu
L Postacrosome Lu
(8–18) A
PL 9 T
Hermes biogenesis
A A
B (19)
10 µm 10 µm
In
A
10
Annexin A6; Man2 1; Fam3C; GPP34;
Sapreticulin; HSP70.2; LAMAN
11
UBXD8
12
13 PDILT
19 18 17 16 15 14

Figure 1.13  (a) Wheel depicting the expression profiles of several Golgi localized proteins. All proteins listed span the Golgi
apparatus of steps 1–7 spermatids. In addition, UBXD8 also reveals a cytoplasmic reaction in spermatocytes during meiosis while
PDILT is cytoplasmic from steps 8–19. Annexin A6, Man2α1, Fam3C, GPP34, sapreticulin, HSP70.2, and LAMAN reappear at step 19
spermatids. (b), (c), (e) LM-IHC of sapreticulin (b), PDILT (c), and Fam3C (e) at stages V of the cycle revealing Golgi (small arrowheads)
and acrosome (a) reactions of the step 5 spermatids. In (c), the cytoplasmic lobes of the late spermatids show a cytoplasmic reaction
(white asterisks). (d) LM-IF of HSP70.2 at stage VII of the cycle showing a Golgi (small arrowhead) and acrosome (A) reaction at step 7
spermatids. The tails (T) of the late spermatids in the lumen (Lu) also reveal reactivity.

VPS54, the vacuolar-sorting protein responsible for early Early evidence of abnormalities of acrosome forma-
endocytic retrograde transport, is also detected in male germ tion was noted for the blind-sterile (bs) autosomal reces-
cells and follows the intracellular route of USP8/ESCRT-0 sive mutation in mouse that results in male sterility.
during acrosome formation. Spontaneous recessive mutant This is caused by mutations in the Tbc1d20 gene, which
mice lacking Vps54 are infertile. The mutant mouse sperma- encodes a GTPase-activating protein specific for RAB1
tids do not develop an acrosome, as the PAS-positive proac- and RAB2 small GTPase families.155 Sterile bs/bs males
rosomal granules are prevented from coalescing to form the exhibit reduced testis weights and few epididymal sperm
acrosome.152 Thus, as the role of proteins related to endocy- that when present reveal morphologically aberrant head
tosis in spermatids is mounting, some of the proteins exam- shapes. The most striking effect of the bs mutation is the
ined in our study51 may also be related to the early endosome failure of acrosome formation during spermiogenesis with
pathway and roles related to acrosome biogenesis. disorganized proacrosomic granules being detected and
no evidence of a single acrosomal cap or fully developed
DATA FROM OTHER SOURCES REVEALING PROTEINS acrosome.156
REQUIRED FOR ACROSOME FORMATION The GTPase-activating protein, AGFG1, Arf-GAP
An important role of the Golgi apparatus of early sperma- domain and FG repeats-containing protein 1 (also
tids is formation of proacrosomic granules that are capable known as HRB), specific for ARF proteins, shows a simi-
of fusing together as they elaborate the developing acro- lar phenotype when absent.157 HRB, an Asn-Pro-Phe
some. The list of Golgi proteins critical for this process motif-containing protein, associates with the cytosolic
and involved in globozoospermia (abnormal head and no surface of proacrosomic transport vesicles. Although
acrosome) is growing as evidenced from studies includ- such vesicles form in spermatids that lack HRB, the
ing mutant and/or deficient mouse models (see reviews in vesicles are unable to fuse, blocking early acrosome
Foster and Gerton153 and Niu et al.154). development.157,158
Data from other sources revealing proteins required for acrosome formation  15

GBF1 GL54D
GRASP55 TM9SF3; TMED4/p25 GRASP55
TMED7/p27 GPP34
ManIIX
UBXD8 GBF1
Annexin A6; Man2α1; Fam3C; GPP34;
Sapreticulin; HSP70.2; LAMAN PDILT Man2α1
ManIIX TM9SF3
HSP70.2 GL54D
Fam3C TMED7/p27
Sapreticulin TMED4/p25
Annexin A6
1 LAMAN
II 2 PDILT
3
Di UBXD8
4
P(XII) Mitosis 5
(A–B)
P(VII) Meiosis 6

P(I) (PL–II) 7 CE
Acrosome ER
Z (1–7) W
8 S T
L Postacrosome
(8–18) ER
9
PL Hermes biogenesis
(19) A S
B TGN
In W
A N
10

11
Figure 1.15  Schematic representation of stacks (S) of Golgi
12 apparatus of the step 7 spermatid adjacent to the nucleus (N).
19 18 17 16 15 14 13 Proteins localized to both the Golgi and acrosome are enclosed
by the red bracket. Proteins restricted to Golgi apparatus are
enclosed by the green bracket. The trans Golgi network (TGN),
cis Golgi element (CE), endoplasmic reticulum (ER), and wells
Figure 1.14 Wheel depicting the expression profiles (W) of spermatid Golgi apparatus are indicated. (Reproduced
(solid lines) of Golgi localized proteins during the course of with permission from Mol Biol Cell: Au, CE et al., Mol Biol Cell.
spermatogenesis. Dashed lines represent the cytoplasmic 2015; 26(22):4015–4032 DOI:10.1091/mbc.)
localizations of UBXD8 and PDILT. While some proteins show
widespread distributions, many localize to steps 1–7 sperma-
tids during their involvement in the formation of the acro-
some. Some proteins are expressed early in spermatogenesis Protein interacting with C kinase 1 (PICK1), a periph-
but then are repressed in later spermatids only to return in late eral membrane protein involved in protein trafficking, is
step 19 spermatids. highly expressed in round spermatids, where it localizes
to Golgi-derived proacrosomic granules. It cooperates
with other proteins such as Golgi GOPC and CK2alpha’
in acrosome biogenesis. Male mice deficient in PICK1 are
The Golgi-associated PDZ- and coiled-coil motif- infertile and show fragmentation of acrosomes in the early
containing protein (GOPC) localizes to the trans-Golgi steps of spermiogenesis.160
region in early spermatids, and male mice in which GOPC Sirt1, a member of the sirtuin family of proteins, has
is disrupted are infertile. The primary defect is fragmenta- important roles in numerous biological processes. Germ
tion of acrosomes in early round spermatids and abnormal cell specific Sirt1-knockout mice revealed that spermio-
vesicles that fail to fuse and form the acrosome.159 genesis was disrupted due to a defect in acrosome biogen-
Autophagy-related gene 7 (Atg7) is homologous to the esis, resulting in a phenotype similar to that observed in
ubiquitin-activating enzyme E1 (Uba1) and encodes the human globozoospermia. Sirt1 depletion resulted in the
E1-like enzyme that is essential to the ubiquitin conju- failure of microtubule-associated protein 1 light chain 3
gation system. Germ cell specific Atg7-knockout mice (MAP1LC3/LC3) and Atg7 to be recruited to Golgi appa-
are infertile due to a defect in acrosome biogenesis. Atg7 ratus-derived vesicles, and as a consequence resulted in
partially regulates GOPC during acrosome biogenesis. the failure of GOPC and PICK1 recruitment to nucleus-
Proacrosomic vesicles fail to fuse into a single acrosomal associated acrosomic vesicles. Thus Sirt1 has a novel physi-
vesicle during the Golgi phase of early spermatids.160 ological function in acrosome biogenesis.161
16  Golgi apparatus regulation of differentiation

Islet cell auto antigen 1-like protein (ICA1L), which reveal complex mechanisms and interacting proteins for
has sequence similarities to ICA69 (also known as ICA1), acrosome formation.167
has been identified as a BAR-domain binding partner Galnt3, N-acetylgalactosaminyltransferase 3, is a mem-
(Bin, amphiphysin, Rvs) of PICK1 that is crucial for acro- ber of the GalNAc transferase gene family involved in the
some formation. ICA1L and PICK1 are highly expressed initiation of mucin-type O-glycosylation. Galnt3-deficient
in spermatids and traffic together at different steps of mice are infertile. Galnt3 protein localizes in the cis-medial
spermiogenesis. ICA1L-knockout mice reveal that PICK1 cisternae of the Golgi stacks of spermatocytes and sper-
expression is reduced by 80% in the testes of these mice. matids in seminiferous tubules. Spermatozoa in Galnt3-
Sperm from ICA1L-knockout mice have abnormalities deficient mice are rare and immotile, and reveal deformed
in the acrosome, nucleus, and flagellar mitochondrial heads. During acrosome formation, Golgi-derived proac-
sheath formation. Both total and motile sperm numbers rosomal vesicles of various sizes do not coalesce to form
are reduced and about half of the remaining sperm have a single acrosomal vesicle. Overall, the data reveal that
the characteristics of globozoospermia. These defects deficiency of Galnt3 results in a severe reduction of mucin-
ultimately result in reduced fertility of male ICA1L- type O–glycans in spermatids and causes impaired acro-
knockout mice. ICA69/ICA1L-double knockout male mice some formation, leading to oligoasthenoteratozoospermia
are sterile.163 (abnormal low number of sperm and with low motility).168
CCDC136 (coiled-coil domain containing 136), a Polypeptide N-acetylgalactosaminyltransferase-like  pro­­
novel testis-specific protein, also localizes to the acro- tein 5 (GALNTL5) belongs to the pp-GalNAc-T gene fam-
some of round and elongating spermatids. Ccdc136- ily. GALNTL5, catalyzes the first step in mucin-type
deficient males are infertile due to disrupted acrosome O-glycosylation of peptides in the Golgi apparatus.
formation. The expression levels of proteins SPACA1 Heterozygous mutation of Galntl5 causes infertility in male
(sperm acrosome membrane-associated protein 1, see mice because of immotile sperm. Mouse GALNTL5 local-
below) and PICK1 involved in acrosome formation are izes in the cytoplasm of round spermatids in the region
also significantly downregulated in the testes of Ccdc136- of the acrosome, and in Galntl5-deficient mutant mice an
deficient mice.164 abnormal distribution of protein loading into acrosomes is
The Fads2 gene encodes an enzyme enriched in testicular observed along with other abnormalities.169
membrane phospholipids that is required for the endog- Several other Golgi proteins are associated with acro-
enous synthesis of an omega-3 fatty acid, docosahexaenoic some development and interact with GOPC or HRB but
acid (DHA).86 In Fads2-deficient spermatids, proacrosomic a phenotype for their absence has not as yet been defined.
vesicles fail to fuse and form a well-developed acrosome. They include the Golgi protein SPATA16 protein, a tes-
Furthermore, syntaxin 2 reveals no extensive colocal- tis-specific protein that belongs to the tetratricopeptide
ization with acrosin in these mice. Syntaxins are a large repeat-like superfamily (also known as NYD-SP12)170,171
protein family implicated in the targeting and fusion of and family with sequence similarity 170, member B
intracellular transport vesicles. Absence of acrosomes in (Fam170b).172
Fads2-deficient round spermatids and misplaced syntaxin An interesting twist is the protein MFSD14A, a sol-
2 suggests an essential role of DHA in proper delivery ute carrier protein expressed in Sertoli cells. Its absence
of membrane proteins required for proacrosomic vesicle is associated with transport of a solute, suggested to be
fusion.165 mannose, from the bloodstream that is required for sper-
Sec23 is a component of the coat protein complex II miogenesis. Small vesicles that stain for glycoproteins are
(COPII) which promotes the formation of transport vesi- abundantly found in early spermatids of mutant mice
cles from the endoplasmic reticulum. Mutations affecting model suggesting a defect in vesicular trafficking from the
SEC23IP, a phospholipaseA1-like protein that interacts Golgi and/or fusion with the developing acrosome.173
with Sec23, also cause male subfertility by interfering
with acrosome biogenesis and leading to round-headed PROTEINS INVOLVED IN ACROSOME BINDING
spermatozoa.166 TO THE NUCLEAR SURFACE
SMAP2, an ArfGTPase-activating protein detected in In addition to a role in acrosome formation, the early sper-
the TGNs of pachytene spermatocytes up to early round matid has developed a mechanism whereby the forming
spermatids, binds to both clathrin, a protein that plays acrosome binds to the nuclear surface opposite the trans
a major role in the formation of coated vesicles, and the face of the Golgi apparatus.
clathrin assembly protein (CALM). In SMAP2-deficient Sperm acrosome associated 1 (Spaca1) is a member of
male mice, the diameter of proacrosomic vesicles budding the globozoospermia-related genes. Disruption of Spaca1
from the TGNs increases, TGNs are distorted, and acro- leads to the disappearance of the acrosome-anchoring
some formation is severely impaired. Moreover, a link is scaffold, the acroplaxome, and decreased expression of
established between SMAP2, CALM, and syntaxin2 in Ccdc-136 and PICK1. This coincides with the failure of
clathrin-coated vesicle formation derived from the TGN acrosomal expansion during spermiogenesis resulting in
and subsequent acrosome formation. Together the data the degeneration and disappearance of the acrosome.174
Defining Golgi proteins in spermatids  17

Dpy19l2, a transmembrane protein, is expressed in sper-


matids with a specific localization restricted to the inner
nuclear membrane facing the acrosome. In the absence of S
Dpy19l2 the acrosome fails to be linked to the nucleus lead- *
ing to a disruption of vesicular trafficking from the Golgi
apparatus.175,176
The TATA element modulatory factor (TMF), a well- S
known Golgi associated protein, was highly abundant in
our isolated testis Golgi fraction.51 Also known as TMF/ *
(a) * (b)
ARA160, its absence in male mice led to a specific devia-
tion of the trans face of the Golgi (TGNs) away from
the nucleus of the developing early round spermatids.
Concomitantly, proacrosomic vesicles derived from the
TGNs of KO mice lacked targeting properties and did not
tether to the spermatid nuclear membrane acroplaxome.
A proposed function in round spermatids is its association
with microtubules for Golgi positioning as a requirement
for acrosome formation.176 (c) (d)

DEFINING GOLGI PROTEINS IN SPERMATIDS Figure 1.16  EM images of Golgi apparatus of late sperma-
tids. (a) Golgi apparatus of a step 14 spermatid lies freely in the
Post acrosome period: Differential expression
cytoplasmic lobe. It is compact, spherical, and composed of
of SORTILIN and ManIIX
tightly apposed flattened cisternae forming stacks (S) with ER
After acrosome formation, the Golgi apparatus becomes (asterisks) close by. (b) A step 16 spermatid reveals a small iso-
spherical and positions itself in the cytoplasmic lobe of lated Golgi stack of cisternae (S) with autophagosomes (curved
late spermatids. An EM illustration of a step 12 spermatid arrows) containing cisternae nearby. (c) Step 17 spermatid
Golgi is seen in Figure 1.16a. Coincident with the move- reveals an aggregate of small flattened cisternae and associ-
ment of the Golgi apparatus into the cytoplasmic lobe at ated vesicular profiles with a few isolated cisternae (arrows) at
step 8 spermatids, Golgi-located immunogenicity of 12 a distance. (d) Forming Hermes body of a step 19 spermatid
proteins (i.e., GRASP55, GBF1, ManIIX, Annexin A6, reveals small flattened cisternae (arrows) that do not form a
GPP34, Man2α1, HSP70.2, Fam3C, PDILT, sapreticulin, stack but at times show close and random focal associations.
UBXD8, and LAMAN) was below detection. In contrast, Original magnifications: 16a–d: 48,750×.
four Golgi proteins (i.e., GL54D, TM9SF3, TMED4/p25,
and TMED7/p27) maintained reactivity into late spermio-
genesis, thus defining the identity of the Golgi apparatus The Golgi is responsible for glycosylation180 and this
in late spermatids. occurs during acrosome formation with expression
Golgi migration away from the acrosome also corre- of Man2α1 and ManIIX. These sequence-related pro-
sponded precisely with the onset of expression of sorti- teins likely have related functions in alpha mannosidase
lin in the Golgi apparatus at step 8 and continuing until trimming of high mannose containing N-linked oli-
step 19 spermatids (Figure 1.17a and examples in Figure gosaccharides.133 The lack of detection of both of these
1.17b, c). Sortilin transports lysosomal enzymes from the proteins from steps 8–14 would suggest a diminution in
Golgi apparatus to MVBs and lysosomes.178 At the end of Golgi mannosidase activity that in turn would be linked
acrosome formation the TGNs appear to be fully utilized to the well-known major changes in N-linked glycosyl-
and undergo fragmentation into vesicles and tubules24,28,29, ation of proteins accounting for the high mannose and
46,179 and are no longer evident in step 8 spermatids and complex glycoproteins differentially expressed during
beyond. In early spermatids, trafficking from the TGNs spermatogenesis.32,96
to the developing acrosome appears to utilize mannose 6 However, from steps 15–19, ManIIX expression returns
phosphate receptors as their RNA transcripts are promi- (Figure 1.17a and d), likely heralding a return of complex
nent.180 However, from step 8 onward, the expression of sugar modification reactions on N-linked glycoproteins
sortilin suggests its role in trafficking that would involve with its expression being essential for spermatogenesis
Golgi cisternae other than the TGNs. and male fertility.182,183 Terminal sugar incorporation in
Of note is the absence of MVBs and lysosomes from the Golgi apparatus has been documented in late sper-
steps 9–10 spermatids onward.14,33 Hence, sortilin in the matids for glycoprotein synthesis destined for the plasma
Golgi apparatus of late spermatids would not be involved membrane.184
in lysosomal targeting but rather likely functions in During the last steps of spermiogenesis (steps 15–19),
plasma membrane remodeling. there is the appearance of tubulobulbar complexes.185
18  Golgi apparatus regulation of differentiation

(a) (b)
Lu

II 1 2
3 S
Di
4 Rb Rb
S
P(XII) Mitosis 5
(A–B) 10 µm
P(VII) 6
Meiosis (c)
(PL–II)
P(I) 7
Acrosome S Rb
Z (1–7) Lu
8
L Postacrosome Rb
(8–18)
9
PL Hermes biogenesis S
10 µm
B (19)
In (d)
A A
10
Rb
A

11 Rb
Lu
10 µm
12
19 18 17 16 15 14 13

Sortilin ManIIX

Figure 1.17  (a) Wheel depicting the expression profiles of ManIIX and sortilin. ManIIX spans the Golgi apparatus of steps 1–7
spermatids and steps 15–19 while sortilin is restricted to steps 8–19. (b) LM-IHC of sortilin in a tubule at stage VI of the cycle. Reactions
are noted in the Golgi apparatus (large arrowheads) of the cytoplasmic lobes of late spermatids. Sertoli cells (S) are also reactive, as
are small spherical bodies near the base of the tubule corresponding to residual bodies (Rb). (c) LM-IHC of sortilin at stage VIII shows
reactions in Golgi of Sertoli cells (S) and step 8 spermatids (arrowheads) and residual bodies (Rb). (d) LM-IHC of ManIIX. Right tubule
at stage VI shows acrosome (A) and Golgi reaction at step 6 spermatids (small arrowheads) and late spermatids (large arrowheads);
left lower tubule at late stage VIII with reactions in residual bodies (Rb).

Such complexes are long (2–3 μm), narrow (50 nm) tubu- as unstacked entities congregate into a focal bulbous
lar projections of the spermatid’s plasma membrane that dilation of cytoplasm at the neck region of the step 19
invaginate into Sertoli cells. They disappear just before the spermatid flagellum (Figure 16d), which we have termed
release of spermatids into the lumen of the seminiferous the Hermes body, previously named the cytoplasmic
tubule at step 19.186 The Golgi apparatus may serve to pro- droplet.35
vide plasma membrane proteins needed for formation of ManIIX selectively reappears at step 15, coinciding
these membranous complexes, and this may involve a role closely with Golgi fragmentation and with a marked
for ManIIX and sortilin, both highly expressed during late expression of β-COP. 35,186 These findings suggest a
spermiogenesis. marked retrograde transport of COPI vesicles from the
Golgi to the ER,187 which may be linked to Golgi frag-
FRAGMENTATION OF THE GOLGI APPARATUS mentation and cisternal unstacking.188 Nevertheless,
IN LATE SPERMATIDS the expression of ManIIX and sortilin suggests that the
After step 15, the Golgi apparatus undergoes fragmen- Golgi is still functional during these very late steps of
tation, vacuolation, and autophagy (Figure 1.16b), 29,34 spermiogenesis. In addition, the Golgi apparatus after
suggesting that components of the Golgi ribbon frag- fragmentation appears to be destined to end up in two
ment into individual stacks between steps 16–19 (Figure different locations. The individual Golgi cisternae end
1.16b), with Golgi cisternae separating from the stack up in the forming Hermes body at step 19 spermatids
and becoming liberated as distinct entities floating in for functions related to epididymal sperm. 35 Other com-
the cytoplasm (Figure 1.16c). At step 19, the cisternae ponents of the Golgi ribbon appear in residual bodies as
Fate of Golgi localized proteins destined for the forming Hermes body at step 19 spermatids  19

small but yet discrete entities that are especially reactive are defined as step 19 spermatids, and it may be advanta-
for sortilin (Figure 1.17c) and ManIIX (Figure 1.17d). geous to define those at stage VII as 19a and at stage VIII
What these entities correspond to awaits future inves- as 19b, as illustrated by Clermont in Figure 1.4, which
tigation by EM. was never published by him but which we used in an arti-
cle describing his life in research.16
FATE OF GOLGI LOCALIZED PROTEINS DESTINED FOR Interestingly, a resurgence of protein expression
THE FORMING HERMES BODY AT STEP 19 SPERMATIDS highlights the step 19 spermatids. Reactivity returns
At the last step of spermiogenesis, two prominent for TMED7/p27 that persisted until step 16, as well as
structures appear in relation to step 19 spermatids for GBF1, Annexin A6, GPP34, Man2α1, LAMAN,
(Figures 1.4 and 1.5). The larger structure adjacent to the HSP70.2, PDILT, sapreticulin, and FAM3C, which were
head of the spermatid constitutes the residual body into shut down at step 8. ManIIX becomes absent from steps
which the majority of unused organelles congregate and 8–14 but then reappears from steps 15–19 spermatids
which is phagocytosed and degraded by Sertoli cells.189,190 (Figure 1.18a). All of these proteins localized to the
The other smaller structure appears as a bulge of cyto- forming Hermes body. In addition, two additional Golgi
plasm positioned at the neck (connecting piece) region proteins became expressed for the first time at step 19
of the flagellum; it defines the cytoplasmic droplet191 (i.e., TMED2/p24 [Figure 1.18a] and carboxypeptidase
that is retained by sperm as they traverse the epididy- D, a serine exopeptidase that releases C-terminal amino
mis14,34,192–194 and that has been renamed recently as the acids and which is concentrated in the Golgi apparatus),
Hermes body.35 where it functions by cycling to and from the plasma
Step 19 spermatids span two stages of the cycle (i.e., membrane195 (Figure 1.18a and b).
stages VII and VIII) with a duration of 58 hours for VII Although a stacked Golgi apparatus and ribbon are
and 30 hours for VIII (Figure 1.3).3 However, it is only at no longer detectable at step 19, these proteins define the
stage VIII of the cycle that the forming Hermes body seg- flattened membranous elements that congregate into the
regates itself from the residual body and is seen as a dis- forming Hermes body.35 Interestingly, sortilin is also
tinct entity. At both of these stages, the late spermatids expressed at step 19 spermatids, but unlike the 13 proteins

(a) (b)
Rb
Rb

II 1 2
3
Di Lu
4
P(XII) Mitosis 5 10 µm
(A–B)
P(VII) 6
Meiosis
P(I) (PL–II)
7
Acrosome
Z (1–7) 8
L Postacrosome
(8–18)
PL 9
Hermes biogenesis
B (19)
In
A
10 Carboxypeptidase D; TMED2/p24
GBF1
11 ManIIX
Annexin A6; Man2 1; Fam3C; GPP34;
12 Sapreticulin; HSP70.2; LAMAN
19 18 17 16 15 14 13 TMED7/p27
PDILT

Figure 1.18  (a) Wheel depicting several Golgi localized proteins (solid lines) showing different expression profiles during sper-
matogenesis. The dashed line for PDILT shows its cytoplasmic localization during late spermiogenesis. (b) LM-IHC of carboxypepti-
dase D shows reaction in Hermes bodies (curved arrows) and residual bodies (Rb) of step 19 spermatids.
20  Golgi apparatus regulation of differentiation

listed above, sortilin ends up exclusively in residual bod- (Figure 1.17d). In addition, the data also revealed that a
ies at step 19 spermatids and is not a constituent of the segregation of both of these proteins was taking place,
Hermes body. as sortilin ended up exclusively in the residual body
An important function of step 19 spermatids is while ManIIX expression was also noted in the forming
in spermatid individualization into spermatozoa by Hermes body at step 19 (Figure 1.18a). These proteins
disruption  of the syncytium formed by intercellular not only serve as controls for each other but also point
bridges.196–198 It is attractive to propose the internal to key dramatic differences.
membranes (Golgi cisternae and vesicles) of the form-
ing Hermes body as the source of new plasma mem- PROTEINS OF UNKNOWN FUNCTION
brane to seal off each individual spermatid resulting in In the present work, we describe two abundant proteins
spermatozoa. of unknown function as new Golgi markers (GL54D and
The forming Hermes body of step 19 spermatids is TM9SF3). Moreover, over 200 other proteins of unknown
maintained in epididymal sperm. The protein composi- function were uncovered in the isolated testis Golgi frac-
tion of the Hermes body of epididymal sperm has been tion, each of which may now be studied in detail to eluci-
analyzed in detail by proteomics and localization stud- date their mechanisms during germ cell differentiation.51
ies.34 The data suggest that the flattened cisternal mem-
branes show marked differences in their Golgi protein DO PATTERNS OF DIFFERENTIAL EXPRESSION
composition from the Golgi apparatus of germ cells OF GOLGI LOCALIZED PROTEIN EXPRESSION
evolving during spermatogenesis with some Golgi local- COINCIDE WITH NON-GOLGI PROTEINS DURING
ized proteins not entering the Hermes body (i.e., GL54D, SPERMATOGENESIS
TMED4/p25, sortilin, UBXD8, TM9SF3, and GRASP55). The data on all 20 Golgi localized proteins revealed a
In this way, the flattened cisternae of the step 19 sper- marked differential expression pattern as germ cells
matids appear to have different molecular constituents evolved during spermatogenesis (Figure 1.19). Hence, a
and this may be analogous to that postulated for yeast- proteomics-based strategy of the isolated testis Golgi frac-
flattened cisternae.199,200 In addition, these cisternae may tion enabled the identification and selection of 20 proteins
function independent of a stacked configuration as sug- whose differential Golgi localization was mapped during
gested for Golgi cisternae in other systems201 and with- spermatogenesis. Figure 1.20 depicts proteins expressed
out the conventional setup of a continuous Golgi ribbon. during the period of mitosis, meiosis, acrosome, postacro-
Thus, the Hermes body of epididymal sperm formed at some, and Hermes body formation.
step 19 spermatids appears to have evolved a system of In an earlier study we hypothesized that the flattened
unique flattened membranous cisternae unlike the con- cisternae of the Hermes body at step 19 spermatids were of
ventional stacked Golgi apparatus to carry out specific Golgi identity.34 One of the objectives of our study on iso-
functions for sperm maturation. 35 lation of the Golgi apparatus of germ cells51 was to attempt
to validate this hypothesis. In a companion study, we iso-
FATE OF GOLGI LOCALIZED PROTEINS DESTINED lated the Hermes body of epididymal sperm and charac-
FOR RESIDUAL BODIES AT STEP 19 SPERMATIDS terized its protein composition by proteomics. Based on
Before being released from the seminiferous epithelium, their abundance, several selected non-Golgi proteins were
the step 19 spermatid rids itself of organelles not uti- characterized and localized to the Hermes body of epidid-
lized during spermatid differentiation. Defined as the ymal sperm.35 A detailed morphological analysis was also
residual body (Figures 1.4 and 1.5), this excess cytoplas- performed of their differential expression during sper-
mic mass contains mitochondria not used during for- matogenesis. In this study, a comparison of the differential
mation of the midpiece of the flagellum, lipid droplets, expression between the 20 Golgi localized proteins will
and ER cisternae including remnants of the annulate be made with the non-Golgi proteins35 in order to define
lamellae and radial body.9,14,94,189,202,203 The residual body possible functional correlations between these two sets of
is embraced by processes of Sertoli cells and phagocy- proteins during spermatogenesis.
tosed within the Sertoli cell cytoplasm, where it under-
goes degradation with participation of its lysosomal MATCHING PROTEIN EXPRESSION PROFILES
system.189 OF GOLGI AND NON-GOLGI PROTEINS DURING
Intriguingly, in our study, all proteins found in the SPERMATOGENESIS
step 19 forming Hermes body were also noted in resid- The most abundant protein of the isolated Hermes body
ual bodies. This suggests that specific components of fraction was glucose transporter 3 (GLUT-3). 35 Its expres-
the Golgi ribbon segregate themselves into these two sion was first detected in pachytene spermatocytes at
distinct structures. While the reaction for Golgi pro- stage VII of the cycle exactly coincident with the expres-
teins in residual bodies was usually diffuse, in only two sion of GL54D, the most abundant Golgi protein of germ
cases were the reactions seen as discrete clumps of reac- cells (Figure 1.21a). 51 The reaction product delineated the
tion products (i.e., sortilin) (Figure 1.17c) and ManIIX entire plasma membrane of early germ cells (solid green
Coincidental temporal expression of non-Golgi and Golgi proteins: Relationship with ER of spermatids  21

Annexin A6; Man2 1; Fam3C; GPP34; all the way up to step 19 spermatids (Figure 1.22a and
Carboxypeptidate D; TMED2/p24 Sapreticulin; HSP70.2; LAMAN
GRASP55 GL54D b). Golgi proteins with widespread distribution include
GBF1 TM9SF3; TMED4/p25 TMED7/p27 (Figure 1.22c), TM9SF3, and TMED4/p25, all
Sortilin TMED7/p27 of which may serve as the client for MCT1 (Figure 1.22a).
ManIIX UBXD8 The enhanced expression of a number of proteins dur-
MG160 PDILT
ing meiosis (Figure 1.23a) was remarkable. From pachy-
tene spermatocytes (stage I) until the end of meiosis, there
was prominent immunoreactivity noted for calnexin, a
chaperone participating in the folding and quality con-
trol of client proteins (Figure 1.23b) and binding immu-
II 1 2 noglobulin protein (BIP), an ER HSP70 family member
3
Di also known as glucose-regulated protein 78 or HSPA5.
4
Vesicle-associated membrane protein 3 (VAMP3), the
P(XII) Mitosis 5
GTP-binding protein RAB14 (Figure  1.23c), clathrin
(A–B)
P(VII)
Meiosis 6 heavy chain (CHC), elongation factor EF1α, the ubiq-
P(I) (PL–II) 7 uitin-directed AAA-ATPase P97, proteasome α 1-7, the
Acrosome GTPase activating protein, sec23, a coat component of
Z (1–7) COPII vesicles, and ubiquitin were also highly expressed
8
L Postacrosome during this time point in meiosis (Figure 1.23a). All of
(8–18) 9
PL
Hermes biogenesis
these proteins mark in large part to the period of MG160
B (19) expression to these cells (Figure 1.23a). Another coin-
In
cident pattern is that seen for UBXD8 as a cytoplasmic
A localized protein (Figure 1.23d) and MG160 (Figure
10 1.23a) both expressed in spermatocytes and both reveal
overlapping expression during meiosis. The meiotic
11 period represents a dramatic size increase of the Golgi
12
apparatus during spermatocyte maturation due to an
19 18 17 16 15 14 13 accumulation of trans Golgi elements83 and correspond-
ing to a significant increase in size of the Golgi apparatus
(0.5–1.0 to 2–3 μm in diameter) to produce four haploid
daughter cells as a consequence of meiosis.209 These pro-
teins may be related to this event.
Figure 1.19  Wheel depicting widespread Golgi distribu- Another similar distribution pattern (Figure 1.24a)
tions (solid lines) during spermatogenesis. Cytoplasmic local- exists for the renewed onset of expression of ManIIX from
izations for PDILT and UBXD8 are depicted by dashed lines. steps 15–19 (Figure 1.24b) with the coincident expression
of atlastin 3 (functioning in tubular ER network forma-
tion) (Figures 1.24c and d), β-COP, ERP57, and RAB14 at
these same steps of differentiation. A rationale for these
line, Figure 1.21a) with reactivity becoming spotty from coincidences of expression patterns is still unknown but
steps 8–18 spermatids (dashed green line, Figure 1.21a; indicates a partnership between non-Golgi and Golgi
and Figures 1.21b and c). Curiously, the wave of sorti- proteins.
lin expression in the Golgi apparatus at step 8 coincided
with a marked diminishment in surface located GLUT-3 COINCIDENTAL TEMPORAL EXPRESSION OF
of the plasma membrane of late spermatids (Figure NON-GOLGI AND GOLGI PROTEINS: RELATIONSHIP
1.21a). While sortilin expression is known to regulate WITH ER OF SPERMATIDS
GLUT-4 trafficking in insulin-responsive cells, 204–208 its Extensive membrane trafficking exists between the ER
role in GLUT-3 sorting and abundance is unclear but is and Golgi especially for the secretory transport of newly
predicted from the present study. Sortilin also shows a synthesized proteins, their posttranslational modifica-
coincident expression pattern with PDILT, as the latter tions including N and O glycosylation, and the quality
turns from being Golgi localized (steps 1–7) to a cyto- control of folding of newly synthesized secretory pro-
plasmic ER localization from steps 8–19 (Figure 1.21a). teins. Of the numerous Golgi proteins identified in our
Monocarboxylate transporters (MCTs) utilize proton previous work (50), only three Golgi proteins (Figure
gradients to move molecules with one carboxylate group 1.25a) (i.e., TMED7/p27, TM9SF3 [Figure 1.25b], and
such as lactate or pyruvate across the plasma membrane. TMED4/p25) showed widespread distributions across
MCT1 showed a widespread immunoreactivity decorat- spermatogenesis coincident in part with that for BIP
ing the plasma membranes of preleptotene spermatocytes (Figure 1.25c) and calnexin.
22  Golgi apparatus regulation of differentiation

Ax
Aa

TMED7/p27
TM9SF3 Ax G
TMED4/p25 Aa
C ICB TMED7/p27
UBXD8 DL
CB TM9SF3
GL54D
Testis TMED4/p25
GBF1
ICB M GL54D
Annexin A6
ManIIX
Man2a1
Sortilin
Fam3C CA
GPP34 G
AS N
Sapreticulin
PDILT Acrosome
AS
HSP70.2 (steps 1–7)
LAMAN Postacrosome
ManIIX (steps 8–18)
Grasp55

ER
S
TMED7/p27
TM9SF3 ER
TMED7/p27
TMED4/p25 GBF1
UBXD8 Annexin A6
Meiosis
GL54D Golgi Man2a1
spermatocytes (PI-II)
MG160 1318 proteins An Fam3C
GBF1 GPP34
Sapreticulin
PDILT
HSP70.2
M LAMAN
ManIIX
HB
Carboxypeptidate D
TMED7/p27 TMED2/p24
RB
TM9SF3
TMED4/p25 Ad
UBXD8
Mitosis
spermatogonia Hermes biogenesis
(step 19)

Figure 1.20  Schematic representation of key events in germ cell differentiation, including mitosis of spermatogonia, meiosis of
spermatocytes from preleptotene (PL) to secondary (II) spermatocytes, acrosome formation from steps 1–7 spermatids, postacro-
some events from steps 8–18, and Hermes body formation at step 19 spermatids. The Golgi apparatus of adult rat germ cells was
isolated and a proteomic analysis by tandem mass spectrometry identified 1318 Golgi proteins. LM-IHC identified several Golgi
proteins belonging to different classes of germ cells and appearing at different time points during germ cell differentiation. The pro-
teins highlighted in red first localized to spermatogonia, those in blue first appeared in spermatocytes, those in green to spermatids
(steps 1–7), those in orange to late spermatids, and those in purple to step 19 spermatids. Thus, the different Golgi localized proteins
reveal differential expression profiles during germ cell differentiation. Note that as each group of proteins, indicated by a specific
color, appeared during development, some maintained expression at later steps of development while others were cut short. Some
expressed in early spermatids (steps 1–7) lost expression from steps 8–18 only to reappear in step 19 spermatids. A few proteins only
localized to step 19 spermatids.
Coincidental temporal expression of non-Golgi and Golgi proteins: Relationship with ER of spermatids  23

(a) (b)

1 T
II 2
3 Lu
Di
4
P(XII) Mitosis 5
P P
(A–B) 10 µm
P(VII) 6
Meiosis
(PL–II) (c) Lu
P(I) 7
Acrosome PM
PM
Z (1–7) 8
L Postacrosome
(8–18) PM
PL 9
Hermes biogenesis
B (19)
10 µm
In
A
10

11
Sortilin
12 GL54D
19 18 17 16 15 14 13 PDILT
GLUT-3

Figure 1.21  (a) Wheel depicting the expression profiles of GL54D, sortilin, GLUT-3, and PDILT. Both GL54D and GLUT-3 dem-
onstrate a precise onset of expression in pachytene spermatocytes at stage VII of the cycle. GL54D is Golgi localized, while GLUT-3
displays an intense reaction on the plasma membrane of germ cells. After step 7, GLUT-3 becomes spotty and remains as such until
step 19 where it shows intense reactivity in the forming Hermes body. Coincident with the spotty reaction of GLUT-3 commencing at
step 8 is the onset of expression of sortilin and the cytoplasmic localization of PDILT. (b) LM-IF of a tubule at stage VII showing intense
reactivity of GLUT-3 on the plasma membrane of pachytene spermatocytes (P), early round spermatids positioned in the center of
the tubule (arrowheads), and the tails (T) of late spermatids in the lumen (Lu). (c) LM-IHC of GLUT-3 at stage II-III reveals plasma mem-
brane (PM) reaction of round spermatids and tails of late spermatids in the lumen (Lu).

Spermiogenesis marks the presence of several specific These modified forms of ER appearing at late steps of sper-
events in the life history of ER.210,211,212 During acrosome miogenesis coincide at a time point when PDILT (its cyto-
formation, ER cisternae are intimately associated with plasmic expression), the chaperone ERP57 (ER protein 57),
various components of the Golgi apparatus9,27,28,213,214 UDP-glucose: glycoprotein glucosyltransferase (UGGT), a
(Figure 1.6). Of the many ER proteins noted in our study,51 key player in the quality control ER mechanisms, glucose-
only BIP and calnexin overlapped with Golgi proteins regulated protein 170 (GRP170) and a component of the ER
involved in acrosome formation and for which a func- chaperone network, and atlastins 1 and 3 predominate (Figure
tional correlation may be deduced. During the later part 1.26a). As the annulate lamellae reveal pores of fixed size
of spermiogenesis, the ER forms annulate lamellae and and shape in their stacked flattened cisternae that resemble
the radial body. Annulate lamellae are focal gatherings nuclear pores, then the presence of RanBP5, a nuclear import
of short flattened membranous cisternae forming a stack factor that spans steps 12–17 spermatids (Figure 1.26a and
where individual cisternae are separated by a fair dis- b) suggests that some cytosolic spermatid proteins contain
tance and characterized by numerous porelike structures a nuclear localization signal that are transported by RanBP5
having a similar appearance to the pore complex of the to the annulate lamellae for some as-yet-unknown function.
nuclear envelope.14,202,213–222 The radial body appears as an The granular-like localizations revealed with the antibodies
aggregate of collapsed ER cisternae in the form of a spoke to these proteins in the cytoplasmic lobe of late spermatids
wheel. It has been suggested that the radial body is a site probably correspond to annulate lamellae and radial bod-
where ER regresses.214,223 ies and suggest roles for these proteins in their structural
24  Golgi apparatus regulation of differentiation

(a) (b)
P
RS
II 1 2
3 P
Di RS
4 Lu
P(XII) Mitosis 5
(A–B) 10 µm
P(VII) 6
Meiosis
(PL–II) (c)
P(I) 7 Lu
Acrosome
Z (1–7) 8
L Postacrosome
(8–18) 9
PL
Hermes biogenesis
B (19) P
P 10 µm
In
A

10

11
12 MCT1
19 18 17 16 15 14 13 TM9SF3; TMED4/p25
TMED7/p27

Figure 1.22  (a) Wheel depicting the expression profiles of MTC1, TMED7/p27, TM9SF3, and TMED4/p25. A widespread distri-
bution is noted for all four of these proteins. (b) LM-IHC of MTC1 in a tubule at stage VII of the cycle showing reactions over the
plasma membrane of pachytene spermatocytes (P), early round spermatids (RS), and step 19 spermatids (curved arrows). (c) LM-IF
of TMED7/p27 at stage V with reactions in Golgi of round (small arrowheads) and late (large arrowheads) spermatids and pachytene
(P) spermatocytes.

integrity and functions (e.g., GRP170, Figure 1.26c). Lactate syntaxin family with a role in mediating endosomal traf-
dehydrogenase C (LDHC) (Figure 1.26d) was also seen to ficking), α-tubulin, Vps13c (member of the Vps13 family
correlate in its cytoplasmic localization to that of PDILT, for of vacuolar protein sorting-associated proteins) (Figure
which an explanation is as yet unknown. 1.27b), and MCT2 (Figure 1.27c). Transferrin and albu-
The pan-germ cell ER markers calnexin and BIP were min also localize to step 19 spermatids as secretory prod-
exclusively restricted to ER and localized from pachytene ucts of Sertoli cells that adsorb to their surface. Several
spermatocytes to step 19 spermatids. These two ER mark- glutathione S transferase (GST) isoforms have also been
ers were never localized to the acrosome or Golgi apparatus noted to be expressed only at step 19 spermatids, 224 as
but did reveal cytoplasmic concentrations as well as focal was also seen for two Golgi proteins (i.e., carboxypepti-
distinct entities at late steps of spermiogenesis presum- dase D and TMED2/p24 [see above]).
ably corresponding to annulate lamellae/radial body com- Many non-Golgi and Golgi proteins while expressed
plexes. These proteins thus provide a control for UBXD8 earlier during spermatogenesis are repressed for some
and PDILT described above that relocated between the ER time only to become reexpressed again at step 19 (Figure
and Golgi during spermatogenesis. 1.28). However, some Golgi proteins, along with RanBP5,
although expressed earlier in spermatogenesis do not end
OVERLAPPING EXPRESSION PROFILES OF NON-GOLGI up in step 19 spermatids (Figure 1.29). This suggests an
AND GOLGI PROTEINS AT STEP 19 SPERMATIDS active but specific biosynthetic and membrane trafficking
As seen in Figure 1.27a, protein expression is extensive in machinery for step 19 spermatids during which time the
step 19 spermatids. Some non-Golgi proteins are solely Hermes body is being formed, as we have speculated.35,51
expressed at step 19 spermatids, such as annexin A1, AP1, It is well established that many genes required for the
and AP2 (activator protein transcription factors), atlastin development and/or function of spermatozoa are tran-
2, malectin (membrane-anchored ER protein), peroxire- scribed much earlier during germ cell differentiation, with
doxin 1, peroxiredoxin 6, syntaxin 13 (member of the an arrest of transcription by step 8 spermatids,30,225 then
Conclusion 25

(a) (b)
Lu
*
* *
II 1 2
3 RS
Di RS
4
Mitosis P
P(XII) 5 P 10 µm
(A–B)
P(VII) Meiosis 6 (c)
P(I) (PL–II)
7
Acrosome
Z Lu
(1–7) 8
Postacrosome *
L
(8–18)
PL 9 *
Hermes biogenesis
(19) * 10 µm
B
In (d)
A
* Lu
10
A
11 *

12 * A
*
19 18 17 16 15 14 13 10 µm

MG160 Calnexin; BIP EF1α; VAMP3; p97; Proteasome α 1-7; Sec23; Ubiquitin
CHC RAB14 UBXD8

Figure 1.23  (a) Wheel depicting the expression profiles of several proteins. Golgi localized MG160 and the cytoplasmic localiza-
tion of UBXD8 match each other precisely from PL to the end of meiosis. Precise onset of expression in pachytene spermatocytes
at stage I of the cycle occurs for calnexin and BIP as well as for EF1α, VAMP3, p97, proteasome α 1-7, Sec 23, RAB14, and CHC; these
proteins overlap in their expression profiles until the end of meiosis. Their distribution profiles at later steps of germ cell differentia-
tion vary according to a given protein. (b) LM-IF of calnexin in a tubule at stage II–III of the cycle. Cytoplasmic localizations are noted
in pachytene spermatocytes (P), early round spermatids (RS) and in cytoplasmic lobes of the late spermatids (asterisks). (c) LM-IHC of
RAB14 shows intense cytoplasmic reaction in pachytene spermatocytes (asterisks) and in Hermes bodies (curved arrows). (d) LM-IHC
of UBXD8 with Golgi (arrowheads) and acrosome (A) reaction in round spermatids and cytoplasmic reaction (asterisks) in pachytene
spermatocytes.

translationally repressed, and finally translated several CONCLUSION


days after mRNA production due to a complex interplay For the first time we have identified the differential
of RNA-binding proteins and non-coding RNA.226–230 As expression pattern of proteins and molecular markers
a result, the differential expression of the Golgi and non- related to the Golgi apparatus during germ cell devel-
Golgi proteins after step 8 must necessarily reflect transla- opment as well as non-Golgi proteins. The end point of
tional regulation. Transcriptional regulation of changes in Hermes body formation in step 19 spermatids was also
the structure and function of organelles of the secretory characterized by a unique set of markers. The differen-
pathway is well established in somatic cells.231 Therefore, the tiation specific expression of new and established Golgi
translation-controlled pathway for organelle modification markers during spermatogenesis may now help define a
and morphogenesis of germ cells232 must be prominent even new model system to answer the unresolved questions
at step 19, an unexplored area of investigation. in Golgi apparatus structure and function.233 Since the
A detailed morphological analysis of the differen- resource also includes unexpected waves of changes
tial expression profile of the non-Golgi proteins dur- in expression patterns of proteins of the plasma mem-
ing spermatogenesis can be seen in Figure 1.30. A brane, endosomes, ER, and cytosol then this also enables
comparison of the differential expression between the a context to test the current models of Golgi appara-
non-Golgi proteins and Golgi localized is illustrated in tus structure and trafficking thus far unresolved by
Figure 1.31. experimentation.
26  Golgi apparatus regulation of differentiation

(a) (b)
Lu

II 1 2
3
Di
4

P(XII) Mitosis 5 10 µm
(A–B)
P(VII) 6
Meiosis (c)
(PL–II)
P(I) 7
Acrosome
Z (1–7)
8 *
L Postacrosome Lu
(8–18)
*
9
PL
Hermes biogenesis
(19) 10 µm
B
*
In (d)
A

10 Rb
Rb

11

Rb Lu
12
13
19 18 17 16 15 14 10 µm

ManIIX Atlastin-3; COP; ERP57 RAB14

Figure 1.24  (a) Wheel depicting the expression profile of the Golgi localized protein ManIIX with several non-Golgi proteins.
Remarkable similarities are noted for these proteins from steps 15–19. (b) LM-IF at stage VII of ManIIX with Golgi reactions in round
(small arrowheads) and late (large arrowheads) spermatids. (c) LM-IHC of atlastin 3 at stage VI shows cytoplasmic reaction in late
spermatids (asterisks). (d) LM-IHC of atlastin 3 at stage VII reveals reactions in residual bodies (Rb) and Hermes body (curved arrows)
of step 19 spermatids.
Conclusion 27

(a) (b)

Lu

1 2
II
3
Di
4 P
P
P(XII) Mitosis 10 µm
5
(A–B)
P(VII) 6
Meiosis (c)
(PL–II)
P(I)
7
Acrosome
Z (1–7) ER
8 Lu

L Postacrosome
(8–18)
PL 9
Hermes biogenesis ER
(19) 10 µm
B

In
A

10

11

12
13
19 18 17 16 15 14

TM9SF3; TMED4/p25 TMED7/p27 Calnexin; BIP

Figure 1.25  (a) Wheel depicting the expression profiles of Golgi localized proteins (TMED7/p27, TM9SF3, TMED4/p25) with non-
Golgi proteins, calnexin, and BIP. While not identical, there is considerable overlap between these two sets of proteins. (b) LM-IF of
TM9SF3 reveals Golgi reactions in pachytene spermatocytes (P), round (small arrowheads), and late (large arrowheads) spermatids.
(c) LM-IHC of BIP reveals cytoplasmic reaction in round spermatids (arrows) and granular ER reactions in late spermatids.
28  Golgi apparatus regulation of differentiation

(a) (b)
Lu *

II 1 2
3 *
*
Di
4
P(XII) *
Mitosis 5
(A–B)
P(VII) Meiosis 6 10 µm

P(I) (PL–II) (c)


7
Acrosome Lu
Z (1–7) 8 ER
L Postacrosome ER
(8–18)
9
PL Hermes biogenesis
B (19)
10 µm
In
A (d)
10 Lu
* *
11 *

12
13
19 18 17 16 15 14
10 µm

UGGT Atlastin-1; GRP170; Hexokinase 1 Atlastin-3; βCOP; ERP57


RanBP5 LDHC PDILT

Figure 1.26  (a) Wheel depicting the expression profiles of several proteins. LDH and the cytoplasmic localization of PDILT over-
lap each other precisely from steps 8–19. Several proteins show somewhat overlapping profiles in late spermatids. RanBP5 also
shows localization in late spermatids (steps 12–16). (b) LM-IHC of RanBP5 in a tubule at stage II–III of the cycle. Reactivity is noted in
the cytoplasm of the late spermatids (asterisks). (c) LM-IHC of GRP170 in the cytoplasmic lobe of a late spermatid shows granular ER
reactions (arrows). (d) LM-IHC of LDHC showing cytoplasmic reaction in cytoplasm of late spermatid (asterisks).

(a) (b)
Lu
II 1 2
3
Di
4 S S
P(XII) 5
Mitosis
P(VII) (A–B) 6 10 µm
Meiosis
P(I) (PL–II) 7 (c)
Z Acrosome Lu
8
(1–7)
L
Postacrosome
(8–18) 9
PL
Hermes biogenesis
B
(19)
In 10 µm
A
10

11 Annexin A1; AP1; AP2


Atlastin-2; Malectin; Peroxiredoxin 1;
12 Peroxiredoxin 6; Syntaxin 13;
13
19 18 17 16 15 14 Transferrin; Albumin; α Tubulin;
MCT2; Vps13c

Figure 1.27  (a) Wheel depicting the expression profile of a number of proteins that are solely expressed at step 19 spermatids.
Albumin and transferrin are secreted products of Sertoli cells that adsorb to the surface of step 19 spermatids. (b) and (c) LM-IHC
of Vps13c (b) and MCT2 (c) in a tubule at stage VII of the cycle. In (b), Sertoli cells (S) are reactive and in (b) and (c) the small dilated
Hermes bodies (curved arrows) attached to the neck region of the step 19 spermatids are also reactive.
Conclusion 29

Carboxypeptidase D; TMED2/p24 Annexin A1; AP1; AP2; Atlastin-2; EF1α; VAMP3; p97; Ubiquitin;
Malectin; Peroxiredoxin 1; Peroxiredoxin 6; Proteasome α 1-7; Sec23
Sortilin Syntaxin 13; Transferrin; Albumin; CHC
GBF1 α Tubulin; MCT2; Vps13c RAB14
UGGT
Annexin A6; Man2α1; Fam3C; GPP34; Calnexin; BIP
Sapreticulin; HSP70.2; LAMAN Atlastin-1; GRP170; Hexokinase 1
MCT1
Atlastin-3; βCOP; ERP57
ManIIX
PDILT
LDHC
TMED7/p27 GLUT-3

II 1 2
3
Di
4
P(XII) 5
Mitosis
P(VII) (A–B)
6
Meiosis
P(I) (PL–II) 7

Z Acrosome
(1–7) 8
L
Postacrosome
PL (8–18) 9
Hermes biogenesis
B (19)
In
A
10

11

12
14 13
19 18 17 16 15

Figure 1.28  Wheel depicting Golgi and non-Golgi proteins that end up in step 19 spermatids. All of these proteins localize to the
Hermes body with the exception of sortilin, which ends up exclusively in residual bodies.
30  Golgi apparatus regulation of differentiation

GRASP55 TM9SF3; TMED4/p25 Annexin A1; AP1; AP2; Atlastin-2; Calnexin; BIP
Malectin; Peroxiredoxin 1; Peroxiredoxin 6;
MG160 RanBP5 Syntaxin 13; Transferrin; Albumin; EF1α; VAMP3; p97;
α Tubulin; MCT2; Vps13c Proteasome α 1-7; Sec23; Ubiquitin
GL54D UBXD8 UGGT CHC
Atlastin-1; GRP170; Hexokinase 1 RAB14
Atlastin-3; βCOP; ERP57
UBXD8
RanBP5
LDHC PDILT

MCT1 GLUT-3
II 1 2
3
Di
4
P(XII) Mitosis 5
(A–B) II 1 2
P(VII) 6 3
Meiosis Di
(PL–II) 4
P(I) 7
P(XII) Mitosis 5
Acrosome
Z (1–7) (A–B)
8 P(VII) 6
Meiosis
L Postacrosome
P(I) (PL–II)
(8–18) 7
PL 9 Acrosome
Hermes biogenesis Z (1–7) 8
(19)
B L Postacrosome
(8–18) 9
In PL
A Hermes biogenesis
B (19)
10
In
A
11 10

12 11
14 13
19 18 17 16 15
12
19 18 17 16 15 14 13

Figure 1.29  Wheel depicting Golgi proteins that do not


end up in the Hermes body of step 19 spermatids.

Figure 1.30  Wheel summarizing the expression profile


of all the non-Golgi proteins in germ cells during the entire
course of spermatogenesis. A varied distribution profile is
noted for each protein. UBXD8 and PDILT are represented
showing their cytoplasmic localizations in germ cells but not
their Golgi localizations.
Conclusion 31

Carboxypeptidase D; TMED2/p24 TM9SF3; TMED4/p25 LDHC


GRASP55 TMED7/p27 Calnexin; BIP
GBF1 MCT1 EF1α; VAMP3; p97; Ubiquitin;
Annexin A1; AP1; AP2; Atlastin-2; Proteasome α 1-7; Sec23
Sortilin Malectin; Peroxiredoxin 1; Peroxiredoxin 6;
Syntaxin 13; Transferrin; Albumin; CHC
ManIIX α Tubulin; MCT2; Vps13c RAB14
MG160 UGGT
UBXD8
Annexin A6; Man2α1; Fam3C; GPP34; Atlastin-1; GRP170; Hexokinase 1
Sapreticulin; HSP70.2; LAMAN PDILT
Atlastin-3; βCOP; ERP57
GL54D RanBP5 GLUT-3

II 1 2
3
Di
4
P(XII) Mitosis 5
(A-B)
P(VII) 6
Meiosis
P(I) (PL-II)
7
Acrosome
Z (1-7) 8
L Postacrosome
(8-18)
PL 9
Hermes biogenesis
B (19)
In
A
10

11

12
13
19 18 17 16 15 14

Figure 1.31  Wheel summarizing the expression profiles of all the Golgi (solid lines) and non-Golgi (dashed lines) proteins in
germ cells during spermatogenesis. A varied profile is noted for each given protein. UBXD8 and PDILT are represented showing both
their Golgi and cytoplasmic localizations.
32  Golgi apparatus regulation of differentiation

ACKNOWLEDGMENTS 11. Dym M, Clermont Y. Role of spermatogonia in


We thank the Facility for Electron Microscopy Research at the repair of the seminiferous epithelium fol-
McGill University (FEMR) (http://www.medicine.mcgill​ lowing x-irradiation of the rat testis. Am J Anat.
.ca/femr/) for EM services (Drs. H. Vali and Kelly Sears) 1970;128(3):265–282.
and the excellent tissue thin sections prepared by Jeannie 12. Clermont Y, Harvey SC. Duration of the cycle of the
Mui. The EM images were taken with a Technai 12 TEM seminiferous epithelium of normal, hypophysec-
(FEI, Hillsboro, OR), coupled to or connected to an AMT tomized and hypophysectomized-hormone treated
XR80C CCD camera (Advanced Microscopy Techniques albino rats. Endocrinology. 1965;76:80–89.
Corp, Woburn, MA). Daniel Friedman, Kristyn Malcolm, 13. Russell LD, Ettlin RA, SinhaHikim AP, Clegg ED.
Zariyat Mannan, Rebecca Richard, Carl Dahlen, Nadiya Histological and Histopathological Evaluation of the
Goswami, Abigail Belasen, Maria-Teresa Eyzaguirre, Testis. Clearwater, FL: Cache River Press; 1990:286.
Aurore Fonderflick, Sohee Kang, Dru Perkins, Andrea 14. Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing
Prince, Jason Lee, Raja Talla, Rachel Adilman, and Adam the wave, cycle, life history, and genes/proteins
Parent contributed by assistance in LM-IHC. We are espe- expressed by testicular germ cells. Part 2: Changes in
cially grateful to the investigators indicated in Table S2 of spermatid organelles associated with development of
Au et al.50 who generously provided antibodies, advice, spermatozoa. Microsc Res Tech. 2010b;73(4):279–319.
and guided us to commercial antibodies when available. 15. Gao Y, Xiao X, Lui WY, Lee WM, Mruk D, Cheng CY.
Supported by CIHR grant MOP 5605. Cell polarity proteins and spermatogenesis. Semin
Note: This chapter is dedicated to Yves Clermont and Cell Dev Biol. 2016;59:62–70.
Charles Philippe Leblond, who formulated the stem cell 16. Hess RA, Hermo L, Robaire B. Lessons learned in
renewal theory for male germ cell differentiation: http:// andrology: Yves Clermont, an interview by Lonnie
www.mcgill.ca​/anatomy/stem-cell-renewal-theory. D. Russell. Andrology. 2015;3(6):1015–1021.
17. Loveland KL, Major AT, Butler R, Young JC, Jans
REFERENCES DA, Miyamoto Y. Putting things in place for fer-
1. Valentine JW, Collins AG, Meyer CP. Morphological tilization: Discovering roles for importin proteins
complexity increase in metazoans. Paleobiology. in cell fate and spermatogenesis. Asian J Androl.
1994;20(2):131–42. 2015;17(4):537–544.
2. Jorpes JE, Werner B, Aberg B. The fuchsin-sulfurous 18. Griswold MD. Spermatogenesis: The commitment to
acid test after periodate oxidation of heparin and meiosis. Physiol Rev. 2016;96(1):1–17.
allied polysaccharides. J Biol Chem. 1948;176(1):​277–282. 19. Chocu S, Calvel P, Rolland AD, Pineau C.
3. Leblond CP, Clermont Y. Spermiogenesis of rat, Spermatogenesis in mammals: Proteomic insights.
mouse, hamster and guinea pig as revealed by the Syst Biol Reprod Med. 2012;58(4):179–190.
periodic acid-fuchsin sulfurous acid technique. 20. Yu Z, Guo R, Ge Y et al. Gene expression profiles in
Am J Anat. 1952;90(2):167–215. different stages of mouse spermatogenic cells during
4. Clermont Y, Leblond CP. Renewal of spermatogonia spermatogenesis. Biol Reprod. 2003;69(1):37–47.
in the rat. Am J Anat. 1953;93(3):475–501. 21. Bose R, Manku G, Culty M, Wing SS. Ubiquitin-
5. Roosen-Runge EC. Kinetics of spermatogenesis in proteasome system in spermatogenesis. Adv Exp Med
mammals. Ann N Y Acad Sci. 1952;55(4):574–584. Biol. 2014;759:181–213.
6. Clermont Y. Kinetics of spermatogenesis in mam- 22. O’Donnell L. Mechanisms of spermiogenesis
mals: Seminiferous epithelium cycle and spermato- and spermiation and how they are disturbed.
gonial renewal. Physiol Rev. 1972;52(1):198–236. Spermatogenesis. 2014;4(2):e979623.
7. Steinberger E, Steinberger A. Spermatogenic func- 23. Fok KL, Chen H, Ruan YC, Chan HC. Novel reg-
tion of the testis. In: Greep RO, Astwood EB, editors. ulators of spermatogenesis. Semin Cell Dev Biol.
Handbook of Physiology. Baltimore, MD: Waverley 2014;29:31–42.
Press; 1975:325–345. 24. Thorne-Tjomsland G, Clermont Y, Hermo L.
8. De Kretser D, Kerr J. The cytology of the testis. In Contribution of the Golgi apparatus components to
Knobil E, Neill J, Ewing L, Greenwald G, Markert C, the formation of the acrosomic system and chroma-
Pfaff D, eds. The Physiology of Reproduction. New York: toid body in rat spermatids. Anat Rec. 1988;221(2):​
Raven Press; 1988:837–932. 591–598.
9. Clermont Y, Hermo L, Oko R. Cell biology of mam- 25. Breucker H, Schafer E, Holstein AF. Morphogenesis
malian spermiogenesis. In Desjardins C, Ewing L, and fate of the residual body in human spermiogen-
editors. Cell and Molecular Biology of the Testis. New esis. Cell Tissue Res. 1985;240(2):303–309.
York: Oxford University Press; 1993:332–376. 26. Griffiths G, Warren G, Stuhlfauth I, Jockusch BM.
10. Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing The role of clathrin-coated vesicles in acrosome for-
the wave, cycle, life history, and genes/proteins mation. Eur J Cell Biol. 1981;26(1):52–60.
expressed by testicular germ cells. Part 1: Background 27. Hermo L, Clermont Y, Rambourg A. Endoplasmic
to spermatogenesis, spermatogonia, and spermato- reticulum-Golgi apparatus relationships in the rat
cytes. Microsc Res Tech. 2010a;73(4):241–278. spermatid. Anat Rec. 1979;193(2):243–255.
References 33

28. Hermo L, Rambourg A, Clermont Y. Three- 44. Ayala I, Colanzi A. Alterations of Golgi organization
dimensional architecture of the cortical region of in Alzheimer’s disease: A cause or a consequence?
the Golgi apparatus in rat spermatids. Am J Anat. Tissue Cell. 2016;49(2):133–140.
1980;157(4):357–373. 45. Burgos MH, Gutierrez LS. The Golgi complex
29. Susi FR, Leblond CP, Clermont Y. Changes in the of the early spermatid in guinea pig. Anat Rec.
Golgi apparatus during spermiogenesis in the rat. 1986;216(2):139–145.
Am J Anat. 1971;130(3):251–267. 46. Mollenhauer HH, Morre DJ, Vanderwoude WJ.
30. Monesi V. Synthetic activities during spermatogen- Endoplasmic reticulum-Golgi apparatus associa-
esis in the mouse RNA and protein. Exp Cell Res. tions in maize root tips. Mikroskopie. 1976;31(9–10):​
1965;39(1):197–224. 257–272.
31. Hecht N, Behr R, Hild A, Bergmann M, Weidner W, 47. Martinez-Menarguez JA, Geuze HJ, Ballesta J.
Steger K. 2009. The common marmoset (Callithrix Identification of two types of beta-COP vesicles in
jacchus) as a model for histone and protamine the Golgi complex of rat spermatids. Eur J Cell Biol.
expression during human spermatogenesis. Hum 1996;71(2):137–143.
Reprod. 24:536–545. 48. Clermont Y, Lalli M, Rambourg A. Ultrastructural
32. Batista F, Lu L, Williams SA, Stanley P. Complex localization of nicotinamide adenine dinucleotide
N-glycans are essential, but core 1 and 2 mucin phosphatase (NADPase), thiamine pyrophosphatase
O-glycans, O-fucose glycans, and NOTCH1 are (TPPase), and cytidine monophosphatase (CMPase)
dispensable, for mammalian spermatogenesis. Biol in the Golgi apparatus of early spermatids of the rat.
Reprod. 2012;86(6):179. Anat Rec. 1981;201(4):613–622.
33. Clermont Y, Tang XM. Glycoprotein synthesis in the 49. Smith CE, Hermo L, Fazel A, Lalli MF, Bergeron JJ.
Golgi apparatus of spermatids during spermiogen- Ultrastructural distribution of NADPase within the
esis of the rat. Anat Rec. 1985;213(1):33–43. Golgi apparatus and lysosomes of mammalian cells.
34. Oko R, Hermo L, Chan PT, Fazel A, Bergeron JJ. The Prog Histochem Cytochem. 1990;21(3):1–120.
cytoplasmic droplet of rat epididymal spermatozoa 50. Au CE, Bell AW, Gilchrist A, Hiding J, Nilsson T,
contains saccular elements with Golgi characteris- Bergeron JJ. Organellar proteomics to create the cell
tics. J Cell Biol. 1993;123(4):809–821. map. Curr Opin Cell Biol. 2007;19(4):376–385.
35. Au CE, Hermo L, Byrne E et al. Compartmentalization 51. Au CE, Hermo L, Byrne E et al. Expression, sort-
of membrane trafficking, glucose transport, glycoly- ing, and segregation of Golgi proteins during
sis, actin, tubulin and the proteasome in the cyto- germ cell differentiation in the testis. Mol Biol Cell.
plasmic droplet/Hermes body of epididymal sperm. 2015;26(22):4015–4032.
Open Biol. 2015;5(8):150080. 52. Regaud C. Etudes sur la structure des tubes semi-
36. Clermont Y, Haguenau F. [Electron microscopy niferes et sur la spermatogenese chez les mammife-
of Golgi’s zone of rat spermatids]. C R Hebd Seances res. Arch Anat Micro. 1901(4):101–156.
Acad Sci. 1955;241(11):708–710. 53. Brinster RL. Germline stem cell transplantation and
37. Clermont Y, Rambourg A, Hermo L. transgenesis. Science. 2002;296(5576):2174–2176.
Connections  between the various elements of 54. de Rooij DG. Proliferation and differentiation of
the cis- and mid-compartments of the Golgi appa- spermatogonial stem cells. Reproduction. 2001;121(3):​
ratus of early rat spermatids. Anat Rec. 1994;240(4):​ 347–354.
469–480. 55. Oatley JM, Brinster RL. Regulation of spermatogo-
38. Rambourg A, Clermont Y. Three-dimensional elec- nial stem cell self-renewal in mammals. Annu Rev Cell
tron microscopy: Structure of the Golgi apparatus. Dev Biol. 2008;24:263–286.
Eur J Cell Biol. 1990;51(2):189–200. 56. Meistrich ML, van Beek MEAB. Spermatogonial
39. Jarvela T, Linstedt AD. Isoform-specific tethering stem cells: Assessing their survival and ability to
links the Golgi ribbon to maintain compartmental- produce differentiated cells. In Chapin RE, Heindel,
ization. Mol Biol Cell. 2014;25(1):133–144. J.J., eds. Methods in Toxicology: Male Reproductive
40. Rios RM. The centrosome-Golgi apparatus nexus. Toxicology. San Diego: Academic Press; 1993:​
Philos Trans R Soc Lond B Biol Sci. 2014;369(1650): 106–123.
20130462. 57. Yoshida S, Takakura A, Ohbo K et al. Neurogenin3
41. Feinstein TN, Linstedt AD. GRASP55 regulates Golgi delineates the earliest stages of spermatogenesis in
ribbon formation. Mol Biol Cell. 2008;19(7):​2696–2707. the mouse testis. Dev Biol. 2004;269(2):447–458.
42. Vinke FP, Grieve AG, Rabouille C. The multiple 58. Nakagawa T, Nabeshima Y, Yoshida S. Functional
facets of the Golgi reassembly stacking proteins. identification of the actual and potential stem cell
Biochem J. 2011;433(3):423–433. compartments in mouse spermatogenesis. Dev Cell.
43. Lavieu G, Dunlop MH, Lerich A, Zheng H, Bottanelli 2007;12(2):195–206.
F, Rothman JE. The Golgi ribbon structure facilitates 59. Yoshida S, Hiyoshi K, Ichinose T et al. Effect of
anterograde transport of large cargoes. Mol Biol Cell. nanoparticles on the male reproductive system of
2014;25(19):3028–3036. mice. Int J Androl. 2009;32(4):337–342.
34  Golgi apparatus regulation of differentiation

60. Mecklenburg JM, Hermann BP. Mechanisms regu- 76. Kleene KC. A possible meiotic function of the pecu-
lating spermatogonial differentiation. Results Probl liar patterns of gene expression in mammalian sper-
Cell Differ. 2016;58:253–287. matogenic cells. Mech Dev. 2001;106(1–2):3–23.
61. Boitani C, Di Persio S, Esposito V, Vicini E. 77. Liebe B, Petukhova G, Barchi M et al. Mutations that
Spermatogonial cells: Mouse, monkey and man affect meiosis in male mice influence the dynamics
comparison. Semin Cell Dev Biol. 2016;59:79–88. of the mid-preleptotene and bouquet stages. Exp Cell
62. Komeya M, Ogawa T. Spermatogonial stem Res. 2006;312(19):3768–3781.
cells: Progress and prospects. Asian J Androl. 78. Moens PB. Molecular perspectives of chromosome
2015;17(5):771–775. pairing at meiosis. Bioessays. 1994;16(2):101–106.
63. Satpute-Krishnan P, Ajinkya M, Bhat S, Itakura E, 79. Scherthan H. Knockout mice provide novel insights
Hegde RS, Lippincott-Schwartz J. ER stress-induced into meiotic chromosome and telomere dynamics.
clearance of misfolded GPI-anchored proteins via Cytogenet Genome Res. 2003;103(3–4):235–244.
the secretory pathway. Cell. 2014;158(3):522–533. 80. van der Laan R, Uringa EJ, Wassenaar E et al.
64. Liaunardy-Jopeace A, Bryant CE, Gay NJ. The COP Ubiquitin ligase Rad18Sc localizes to the XY
II adaptor protein TMED7 is required to initiate and body and to other chromosomal regions that are
mediate the delivery of TLR4 to the plasma mem- unpaired and transcriptionally silenced during
brane. Sci Signal. 2014;7(336):ra70. male meiotic prophase. J Cell Sci. 2004;117(Pt 21):​
65. Dominguez M, Dejgaard K, Fullekrug J et al. gp25L/ 5023–5033.
emp24/p24 protein family members of the cis-Golgi 81. Mikolcevic P, Isoda M, Shibuya H et al. Essential
network bind both COP I and II coatomer. J Cell Biol. role of the Cdk2 activator RingoA in meiotic telo-
1998;140(4):751–765. mere tethering to the nuclear envelope. Nat Commun.
66. Pastor-Cantizano N, Montesinos JC, Bernat- 2016;7:11084.
Silvestre C, Marcote MJ, Aniento F. p24 family 82. Griswold MD, Hogarth CA, Bowles J, Koopman P.
proteins: Key players in the regulation of traf- Initiating meiosis: The case for retinoic acid. Biol
ficking along the secretory pathway. Protoplasma. Reprod. 2012;86(2):35.
2016;253(4):967–985. 83. Suarez-Quian CA, An Q, Jelesoff N, Dym M.
67. Jerome-Majewska LA, Achkar T, Luo L, Lupu F, The Golgi apparatus of rat pachytene spermato-
Lacy E. The trafficking protein Tmed2/p24beta(1) cytes during spermatogenesis. Anat Rec. 1991;
is required for morphogenesis of the mouse embryo 229(1):16–26.
and placenta. Dev Biol. 2010;341(1):154–166. 84. Oke BO, Suarez-Quian CA. Partitioning of the
68. Strating JR, Martens GJ. The p24 family and selective Golgi apparatus in rat primary and secondary sper-
transport processes at the ER-Golgi interface. Biol matocytes during meiosis. Anat Rec. 1992; 233(2):​
Cell. 2009;101(9):495–509. 245–256.
69. Zakariyah A, Hou W, Slim R, Jerome-Majewska L. 85. Munro S. The Golgin coiled-coil proteins of the Golgi
TMED2/p24beta1 is expressed in all gestational apparatus. Cold Spring Harb Perspect Biol. 2011;3(6):
stages of human placentas and in choriocarcinoma a005256.
cell lines. Placenta. 2012;33(3):214–219. 86. Bell AW, Ward MA, Blackstock WP et al. Proteomics
70. Aguilar PS, Frohlich F, Rehman M et al. A plasma-­ characterization of abundant Golgi membrane pro-
membrane E-MAP reveals links of the eisosome with teins. J Biol Chem. 2001;276(7):5152–5165.
sphingolipid metabolism and endosomal trafficking. 87. Dahan S, Ahluwalia JP, Wong L, Posner BI,
Nat Struct Mol Biol. 2010;17(7):901–908. Bergeron JJ. Concentration of intracellular hepatic
71. Schimmoller F, Diaz E, Muhlbauer B, Pfeffer SR. apolipoprotein E in Golgi apparatus saccular dis-
Characterization of a 76 kDa endosomal, multispan- tensions and endosomes. J Cell Biol. 1994;127(6 Pt 2):​
ning membrane protein that is highly conserved 1859–1869.
throughout evolution. Gene. 1998;216(2):311–318. 88. Witkos TM, Lowe M. The Golgin family of coiled-
72. Baarends WM, van der Laan R, Grootegoed JA. DNA coil tethering proteins. Front Cell Dev Biol. 2015;​
repair mechanisms and gametogenesis. Reproduction. 3:86.
2001;121(1):31–39. 89. Kondo M, Sutou S. Cloning and molecular character-
73. Costa Y, Cooke HJ. Dissecting the mammalian ization of cDNA encoding a mouse male-enhanced
synaptonemal complex using targeted mutations. antigen-2 (Mea-2): A putative family of the Golgi
Chromosome Res. 2007;15(5):579–589. autoantigen. DNA Seq. 1997;7(2):71–82.
74. Escalier D. Impact of genetic engineering on the 90. Bentson LF, Agbor VA, Agbor LN et al. New point
understanding of spermatogenesis. Hum Reprod mutation in Golga3 causes multiple defects in sper-
Update. 2001;7(2):191–210. matogenesis. Andrology. 2013;1(3):440–450.
75. Handel MA, Cobb J, Eaker S. What are the sper- 91. Cheung PY, Pfeffer SR. Transport vesicle tethering
matocyte’s requirements for successful meiotic divi- at the trans Golgi network: Coiled coil proteins in
sion? J Exp Zool. 1999;285(3):243–250. action. Front Cell Dev Biol. 2016;4:18.
References 35

92. Wright J, Kahn RA, Sztul E. Regulating the large 106. Christensen AK. Leydig cells. In: Hamilton DW, Greep
Sec7 ARF guanine nucleotide exchange factors: The RO, editors. Handbook of Physiology. 5. Washington,
when, where and how of activation. Cell Mol Life Sci. DC: American Physiological Society; 1975:57–94.
2014;71(18):3419–3438. 107. Rabouille C, Linstedt AD. GRASP: A multitasking
93. Anakwe OO, Gerton GL. Acrosome biogenesis tether. Front Cell Dev Biol. 2016;4:1.
begins during meiosis: Evidence from the synthe- 108. Buschman MD, Xing M, Field SJ. The GOLPH3
sis and distribution of an acrosomal glycoprotein, pathway regulates Golgi shape and function and
acrogranin, during guinea pig spermatogenesis. Biol is activated by DNA damage. Front Neurosci. 2015;​
Reprod. 1990;42(2):317–328. 9:362.
94. Fawcett DW. Gametogenesis in the male: Prospects 109. Gilchrist A, Au CE, Hiding J et al. Quantitative
for its control. Symp Soc Dev Biol. 1975(33):25–53. proteomics analysis of the secretory pathway. Cell.
95. Nicander L, Ploen L. Fine structure of spermatogo- 2006;127(6):1265–1281.
nia and primary spermatocytes in rabbits. Z Zellforsch 110. Hoskins DD, Brandt H, Acott TS. Initiation of sperm
Mikrosk Anat. 1969;99(2):221–234. motility in the mammalian epididymis. Fed Proc.
96. Huang HH, Stanley P. A testis-specific regulator of 1978;37(11):2534–2542.
complex and hybrid N-glycan synthesis. J Cell Biol. 111. Fujiwara Y, Ogonuki N, Inoue K et al. t-SNARE
2010;190(5):893–910. Syntaxin2 (STX2) is implicated in intracellu-
97. Wong M, Munro S. Membrane trafficking. The lar transport of sulfoglycolipids during meiotic
specificity of vesicle traffic to the Golgi is encoded prophase in mouse spermatogenesis. Biol Reprod.
in the Golgin coiled-coil proteins. Science. 2013;88(6):141.
2014;346(6209):1256898. 112. Zhang Y, Hayashi Y, Cheng X et al. Testis-specific
98. Moreno RD, Ramalho-Santos J, Chan EK, Wessel sulfoglycolipid, seminolipid, is essential for germ
GM, Schatten G. The Golgi apparatus segregates cell function in spermatogenesis. Glycobiology.
from the lysosomal/acrosomal vesicle during rhe- 2005;15(6):​6 49–654.
sus spermiogenesis: Structural alterations. Dev Biol. 113. Tokuhiro K, Ikawa M, Benham AM, Okabe M.
2000a;219(2):334–349. Protein disulfide isomerase homolog PDILT is
99. Moreno RD, Ramalho-Santos J, Sutovsky P, Chan required for quality control of sperm membrane pro-
EK, Schatten G. Vesicular traffic and Golgi appara- tein ADAM3 and male fertility [corrected]. Proc Natl
tus dynamics during mammalian spermatogenesis: Acad Sci U S A. 2012;109(10):3850–3855.
Implications for acrosome architecture. Biol Reprod. 114. Gannon J, Bergeron JJ, Nilsson T. Golgi and related
2000b;63(1):89–98. vesicle proteomics: Simplify to identify. Cold Spring
100. Ramalho-Santos J, Moreno RD, Wessel GM, Chan EK, Harb Perspect Biol. 2011;3(12).
Schatten G. Membrane trafficking machinery compo- 115. Smirle J, Au CE, Jain M, Dejgaard K, Nilsson T,
nents associated with the mammalian acrosome dur- Bergeron J. Cell biology of the endoplasmic reticu-
ing spermiogenesis. Exp Cell Res. 2001;267(1):45–60. lum and the Golgi apparatus through proteomics.
101. Hermo L, Pelletier RM, Cyr DG, Smith CE. Cold Spring Harb Perspect Biol. 2013;5(1):a015073.
Surfing the wave, cycle, life history, and genes/ 116. Linder B, Bammer S, Heinlein UA. Delayed transla-
proteins expressed by testicular germ cells. Part tion and posttranslational processing of cyritestin,
3: Developmental changes in spermatid flagellum an integral transmembrane protein of the mouse
and cytoplasmic droplet and interaction of sperm acrosome. Exp Cell Res. 1995;221(1):66–72.
with the zona pellucida and egg plasma membrane. 117. van Lith M, Karala AR, Bown D et al. A developmen-
Microsc Res Tech. 2010c;73(4):320–363. tally regulated chaperone complex for the endoplas-
102. Zhang X, Wang Y. GRASPs in Golgi structure and mic reticulum of male haploid germ cells. Mol Biol
function. Front Cell Dev Biol. 2015;3:84. Cell. 2007;18(8):2795–2804.
103. Seemann J, Jokitalo E, Pypaert M, Warren G. 118. Olzmann JA, Richter CM, Kopito RR. Spatial regu-
Matrix proteins can generate the higher order lation of UBXD8 and p97/VCP controls ATGL-
architecture of the Golgi apparatus. Nature. mediated lipid droplet turnover. Proc Natl Acad Sci
2000;407(6807):1022–1026. U S A. 2013;110(4):1345–1350.
104. Shorter J, Watson R, Giannakou ME, Clarke M, 119. Xu Y, Liu Y, Lee JG, Ye Y. A ubiquitin-like domain
Warren G, Barr FA. GRASP55, a second mam- recruits an oligomeric chaperone to a retrotranslo-
malian GRASP protein involved in the stacking cation complex in endoplasmic reticulum-associated
of Golgi cisternae in a cell-free system. EMBO J. degradation. J Biol Chem. 2013;288(25):18068–18076.
1999;18(18):4949–4960. 120. Xia Y, Yan LH, Huang B, Liu M, Liu X, Huang
105. Rambourg A, Clermont Y, Marraud A. Three- C. Pathogenic mutation of UBQLN2 impairs its
dimensional structure of the osmium-impregnated interaction with UBXD8 and disrupts endoplas-
Golgi apparatus as seen in the high voltage electron mic reticulum-associated protein degradation.
microscope. Am J Anat. 1974;140(1):27–45. J Neurochem. 2014;129(1):99–106.
36  Golgi apparatus regulation of differentiation

121. Garcia-Melero A, Reverter M, Hoque M et al. 136. Sun H, Wolfe JH. Recent progress in lysosomal
Annexin A6 and late endosomal cholesterol modu- alpha-mannosidase and its deficiency. Exp Mol Med.
late integrin recycling and cell migration. J Biol Chem. 2001;33(1):1–7.
2016;291(3):1320–1335. 137. Dmitrieff S, Sens P. Transient domain formation in
122. Pons M, Grewal T, Rius E, Schnitgerhans T, Jackle S, membrane-bound organelles undergoing maturation.
Enrich C. Evidence for the involvement of annexin Phys Rev E Stat Nonlin Soft Matter Phys. 2013;88(6):062704.
6 in the trafficking between the endocytic compart- 138. Lavieu G, Zheng H, Rothman JE. Stapled Golgi cis-
ment and lysosomes. Exp Cell Res. 2001;269(1):13–22. ternae remain in place as cargo passes through the
123. Mack KL, Shorter J. Engineering and evolution of stack. Elife. 2013;2:e00558.
molecular chaperones and protein disaggregases 139. Patterson GH, Hirschberg K, Polishchuk RS, Gerlich
with enhanced activity. Front Mol Biosci. 2016;3:8. D, Phair RD, Lippincott-Schwartz J. Transport
124. Mamelak D, Lingwood C. Expression and sulfo- through the Golgi apparatus by rapid partition-
galactolipid binding specificity of the recombi- ing within a two-phase membrane system. Cell.
nant testis-specific cognate heat shock protein 70. 2008;133(6):1055–1067.
Glycoconj J. 1997;14(6):715–722. 140. Dancourt J, Zheng H, Bottanelli F et al. Small car-
125. Bocking T, Aguet F, Harrison SC, Kirchhausen T. goes pass through synthetically glued Golgi stacks.
Single-molecule analysis of a molecular disassem- FEBS Lett. 2016;590(12):1675–1686.
blase reveals the mechanism of Hsc70-driven clath- 141. Eddy EM. Cytochemical observations on the chro-
rin uncoating. Nat Struct Mol Biol. 2011;18(3):​295–301. matoid body of the male germ cells. Biol Reprod.
126. Eddy EM. Role of heat shock protein HSP70-2 in 1970;2(1):114–128.
spermatogenesis. Rev Reprod. 1999;4(1):23–30. 142. Fawcett DW, Eddy EM, Phillips DM. Observations
127. Dix DJ, Allen JW, Collins BW et al. Targeted gene on the fine structure and relationships of the chro-
disruption of Hsp70-2 results in failed meiosis, germ matoid body in mammalian spermatogenesis. Biol
cell apoptosis, and male infertility. Proc Natl Acad Sci Reprod. 1970;2(1):129–153.
U S A. 1996;93(8):3264–3268. 143. Susi FR, Clermont Y. Fine structural modifications
128. Huszar G, Stone K, Dix D, Vigue L. Putative creatine of the rat chromatoid body during spermiogenesis.
kinase M-isoform in human sperm is identified as Am J Anat. 1970;129(2):177–191.
the 70-kilodalton heat shock protein HspA2. Biol 144. Russell L, Frank B. Ultrastructural characterization
Reprod. 2000;63(3):925–932. of nuage in spermatocytes of the rat testis. Anat Rec.
129. Huszar G, Ozenci CC, Cayli S, Zavaczki Z, Hansch E, 1978;190(1):79–97.
Vigue L. Hyaluronic acid binding by human sperm 145. Da Ros M, Hirvonen N, Olotu O, Toppari J, Kotaja
indicates cellular maturity, viability, and unreacted N. Retromer vesicles interact with RNA granules
acrosomal status. Fertil Steril. 2003;79 Suppl 3:1616–1624. in haploid male germ cells. Mol Cell Endocrinol.
130. Rosario MO, Perkins SL, O’Brien DA, Allen RL, Eddy 2015;401:73–83.
EM. Identification of the gene for the developmentally 146. Segretain D. Endocytosis in spermatids during sper-
expressed 70 kDa heat-shock protein (P70) of mouse miogenesis of the mouse. Biol Cell. 1989;67(3):​289–298.
spermatogenic cells. Dev Biol. 1992;150(1):1–11. 147. Segretain D, Egloff M, Gerard N, Pineau C, Jegou B.
131. Halberg N, Sengelaub CA, Navrazhina K, Molina Receptor-mediated and absorptive endocytosis by
H, Uryu K, Tavazoie SF. PITPNC1 Recruits RAB1B male germ cells of different mammalian species. Cell
to the Golgi network to drive malignant secretion. Tissue Res. 1992;268(3):471–478.
Cancer Cell. 2016;29(3):339–353. 148. Tang XM, Lalli MF, Clermont Y. A cytochemi-
132. Igdoura SA, Herscovics A, Lal A, Moremen KW, cal study of the Golgi apparatus of the sperma-
Morales CR, Hermo L. Alpha-mannosidases tid during spermiogenesis in the rat. Am J Anat.
involved in N-glycan processing show cell specific- 1982;163(4):283–294.
ity and distinct subcompartmentalization within the 149. Oko R, Donald A, Xu W, van der Spoel AC. Fusion
Golgi apparatus of cells in the testis and epididymis. failure of dense-cored proacrosomal vesicles in an
Eur J Cell Biol. 1999;78(7):441–452. inducible mouse model of male infertility. Cell Tissue
133. Moremen KW. Golgi alpha-mannosidase II deficiency Res. 2011;346(1):119–134.
in vertebrate systems: Implications for asparagine-­ 150. van der Spoel AC, Mott R, Platt FM. Differential
linked oligosaccharide processing in mammals. sensitivity of mouse strains to an N-alkylated imino
Biochim Biophys Acta. 2002;1573(3):225–235. sugar: Glycosphingolipid metabolism and acrosome
134. Herscovics A. Importance of glycosidases in mam- formation. Pharmacogenomics. 2008;9(6):​717–731.
malian glycoprotein biosynthesis. Biochim Biophys 151. Berruti G, Ripolone M, Ceriani M. USP8, a regulator
Acta. 1999;1473(1):96–107. of endosomal sorting, is involved in mouse acrosome
135. Rose DR. Structure, mechanism and inhibition of biogenesis through interaction with the spermatid
Golgi alpha-mannosidase II. Curr Opin Struct Biol. ESCRT-0 complex and microtubules. Biol Reprod.
2012;22(5):558–562. 2010;82(5):930–939.
References 37

152. Paiardi C, Pasini ME, Gioria M, Berruti G. Failure 167. Funaki T, Kon S, Tanabe K et al. The Arf GAP
of acrosome formation and globozoospermia in SMAP2 is necessary for organized vesicle budding
the wobbler mouse, a Vps54 spontaneous recessive from the trans-Golgi network and subsequent acro-
mutant. Spermatogenesis. 2011;1(1):52–62. some formation in spermiogenesis. Mol Biol Cell.
153. Foster JA, Gerton GL. The acrosomal matrix. Adv 2013;24(17):2633–2644.
Anat Embryol Cell Biol. 2016;220:15–33. 168. Miyazaki T, Mori M, Yoshida CA et al. Galnt3 defi-
154. Niu CM, Guo JQ, Ma HT, Zheng Z, Zheng Y. ciency disrupts acrosome formation and leads to
[Sperm acrosome formation-associated genes in oligoasthenoteratozoospermia. Histochem Cell Biol.
mice: Advances in studies]. Zhonghua Nan Ke Xue. 2013;139(2):339–354.
2016;22(1):72–76. 169. Takasaki N, Tachibana K, Ogasawara S et al. A het-
155. Liegel RP, Handley MT, Ronchetti A et al. Loss-of- erozygous mutation of GALNTL5 affects male infer-
function mutations in TBC1D20 cause cataracts and tility with impairment of sperm motility. Proc Natl
male infertility in blind sterile mice and Warburg Acad Sci U S A. 2014;111(3):1120–1125.
micro syndrome in humans. Am J Hum Genet. 170. Xu M, Xiao J, Chen J et al. Identification and char-
2013;93(6):1001–1014. acterization of a novel human testis-specific Golgi
156. Sotomayor RE, Handel MA. Failure of acrosome protein, NYD-SP12. Mol Hum Reprod. 2003;9(1):9–17.
assembly in a male sterile mouse mutant. Biol Reprod. 171. Dam AH, Koscinski I, Kremer JA et al. Homozygous
1986;34(1):171–182. mutation in SPATA16 is associated with male infer-
157. Kang-Decker N, Mantchev GT, Juneja SC, tility in human globozoospermia. Am J Hum Genet.
McNiven MA, van Deursen JM. Lack of acro- 2007;81(4):813–820.
some formation in Hrb-deficient mice. Science. 172. Li Y, Lin S, Luo M et al. FAM170B, a novel acrosomal
2001;294(5546):1531–1533. protein involved in fertilization in mice. Mol Reprod
158. Kierszenbaum AL, Tres LL, Rivkin E, Kang-Decker Dev. 2015;82(10):787–796.
N, van Deursen JM. The acroplaxome is the dock- 173. Doran J, Walters C, Kyle V, Wooding P, Hammett-
ing site of Golgi-derived myosin Va/Rab27a/b- Burke R, Colledge WH. Mfsd14a (Hiat1) gene dis-
containing proacrosomal vesicles in wild-type ruption causes globozoospermia and infertility in
and Hrb mutant mouse spermatids. Biol Reprod. male mice. Reproduction. 2016;152(1):91–99.
2004;70(5):1400–1410. 174. Fujihara Y, Satouh Y, Inoue N, Isotani A, Ikawa
159. Yao R, Ito C, Natsume Y et al. Lack of acrosome M, Okabe M. SPACA1-deficient male mice are
formation in mice lacking a Golgi protein, GOPC. infertile with abnormally shaped sperm heads
Proc Natl Acad Sci U S A. 2002;99(17):11211–11216. reminiscent of globozoospermia. Development.
160. Wang H, Wan H, Li X et al. Atg7 is required for 2012;139(19):3583–3589.
acrosome biogenesis during spermatogenesis in 175. Pierre V, Martinez G, Coutton C et al. Absence
mice. Cell Res. 2014;24(7):852–869. of Dpy19l2, a new inner nuclear membrane pro-
161. Xiao N, Kam C, Shen C et al. PICK1 deficiency tein, causes globozoospermia in mice by prevent-
causes male infertility in mice by disrupting acro- ing the anchoring of the acrosome to the nucleus.
some formation. J Clin Invest. 2009;119(4):802–812. Development. 2012;139(16):2955–2965.
162. Liu C, Song Z, Wang L et al. Sirt1 regulates acrosome 176. Harbuz R, Zouari R, Pierre V et al. A recurrent dele-
biogenesis by modulating autophagic flux during tion of DPY19L2 causes infertility in man by block-
spermiogenesis in mice. Development. 2017;144(3): ing sperm head elongation and acrosome formation.
441–451. Am J Hum Genet. 2011;88(3):351–361.
163. He J, Xia M, Tsang WH, Chow KL, Xia J. ICA1L 177. Elkis Y, Bel S, Rahimi R et al. TMF/ARA160 gov-
forms BAR-domain complexes with PICK1 and is erns the dynamic spatial orientation of the Golgi
crucial for acrosome formation in spermiogenesis. apparatus during sperm development. PLoS One.
J Cell Sci. 2015;128(20):3822–3386. 2015;10(12):e0145277.
164. Geng Q, Ni L, Ouyang B, Hu Y, Zhao Y, Guo J. A 178. Braulke T, Bonifacino JS. Sorting of lysosomal pro-
novel testis-specific gene, Ccdc136, is required for teins. Biochim Biophys Acta. 2009;1793(4):605–614.
acrosome formation and fertilization in mice. Reprod 179. Ho HC, Tang CY, Suarez SS. Three-dimensional
Sci. 2016;23(10):1387–1396. structure of the Golgi apparatus in mouse sperma-
165. Roqueta-Rivera M, Abbott TL, Sivaguru M, tids: A scanning electron microscopic study. Anat
Hess  RA, Nakamura MT. Deficiency in the Rec. 1999;256(2):189–194.
omega-3 fatty acid pathway results in failure of acro- 180. O’Brien DA, Welch JE, Fulcher KD, Eddy EM.
some biogenesis in mice. Biol Reprod. 2011;85(4):​ Expression of mannose 6-phosphate receptor mes-
721–732. senger ribonucleic acids in mouse spermatogenic
166. Arimitsu N, Kogure T, Baba T et al. p125/Sec23- and Sertoli cells. Biol Reprod. 1994;50(2):429–435.
interacting protein (Sec23ip) is required for spermio- 181. Zhang X, Wang Y. Glycosylation quality control by the
genesis. FEBS Lett. 2011;585(14):2171–2176. Golgi structure. J Mol Biol. 2016;428(16):3183–3193.
38  Golgi apparatus regulation of differentiation

182. Fukuda MN, Akama TO. The in vivo role of 199. Losev E, Reinke CA, Jellen J, Strongin DE, Bevis BJ,
alpha-mannosidase IIx and its role in processing Glick BS. Golgi maturation visualized in living yeast.
of N-glycans in spermatogenesis. Cell Mol Life Sci. Nature. 2006;441(7096):1002–1006.
2003a;60(7):1351–1355. 200. Papanikou E, Glick BS. The yeast Golgi apparatus:
183. Fukuda MN, Akama TO. The role of N-glycans Insights and mysteries. FEBS Lett. 2009;583(23):​
in spermatogenesis. Cytogenet Genome Res. 3746–3751.
2003b;103(3–4):302–306. 201. Klumperman J. Architecture of the mammalian Golgi.
184. Tang XM. [Glycoprotein synthesis in spermatids of Cold Spring Harb Perspect Biol. 2011;3(7):a005181.
the rat—An EM radioautographic study]. Shi Yan 202. Kessel RG. The structure and function of annulate
Sheng Wu Xue Bao. 1985;18(2):157–169. lamellae: Porous cytoplasmic and intranuclear mem-
185. Russell L, Clermont Y. Anchoring device between branes. Int Rev Cytol. 1983;82:181–303.
Sertoli cells and late spermatids in rat seminiferous 203. Imreh G, Hallberg E. An integral membrane protein
tubules. Anat Rec. 1976;185(3):259–278. from the nuclear pore complex is also present in the
186. Guttman JA, Takai Y, Vogl AW. Evidence that tubu- annulate lamellae: Implications for annulate lamella
lobulbar complexes in the seminiferous epithelium formation. Exp Cell Res. 2000;259(1):180–190.
are involved with internalization of adhesion junc- 204. Hatakeyama H, Kanzaki M. Molecular basis
tions. Biol Reprod. 2004;71(2):548–559. of insulin-responsive GLUT4 trafficking sys-
187. Jackson LP. Structure and mechanism of COPI vesi- tems revealed by single molecule imaging. Traffic.
cle biogenesis. Curr Opin Cell Biol. 2014;29:67–73. 2011;12(12):1805–1820.
188. Wang Y, Wei JH, Bisel B, Tang D, Seemann J. Golgi 205. Huang G, Buckler-Pena D, Nauta T et al. Insulin
cisternal unstacking stimulates COPI vesicle budding responsiveness of glucose transporter 4 in 3T3-L1
and protein transport. PLoS One. 2008;3(2):e1647. cells depends on the presence of sortilin. Mol Biol Cell.
189. Morales C, Clermont Y, Hermo L. Nature and func- 2013;24(19):3115–3122.
tion of endocytosis in Sertoli cells of the rat. Am J 206. Kioumourtzoglou D, Pryor PR, Gould GW, Bryant
Anat. 1985;173(3):203–217. NJ. Alternative routes to the cell surface underpin
190. Tanii I, Yagura T, Inagaki N, Nakayama T, Imaizumi insulin-regulated membrane trafficking of GLUT4.
K, Yoshinaga K. Preferential localization of rat J Cell Sci. 2015;128(14):2423–2429.
GAPDS on the ribs of fibrous sheath of sperm fla- 207. Bogan JS. Regulation of glucose transporter trans-
gellum and its expression during flagellar formation. location in health and diabetes. Annu Rev Biochem.
Acta Histochem Cytochem. 2007;40(1):19–26. 2012;81:507–532.
191. Retzius G. Die Spermien der Huftiere. Niol Untersuch. 208. Hou JC, Pessin JE. Ins (endocytosis) and outs (exo-
1909;14:163–178. cytosis) of GLUT4 trafficking. Curr Opin Cell Biol.
192. Cooper TG. The epididymis, cytoplasmic droplets 2007;19(4):466–473.
and male fertility. Asian J Androl. 2011;13(1):​130–138. 209. Barbosa S, Pratte D, Schwarz H, Pipkorn R, Singer-
193. Fetic S, Yeung CH, Sonntag B, Nieschlag E, Cooper Kruger B. Oligomeric Dop1p is part of the endosomal
TG. Relationship of cytoplasmic droplets to motil- Neo1p-Ysl2p-Arl1p membrane remodeling complex.
ity, migration in mucus, and volume regulation of Traffic. 2010;11(8):1092–1106.
human spermatozoa. J Androl. 2006;27(2):294–301. 210. Hermo L, Pelletier RM, Cyr DG, Smith CE. 2010d.
194. Rengan AK, Agarwal A, van der Linde M, du Plessis Surfing the wave, cycle, life history, and genes/
SS. An investigation of excess residual cytoplasm proteins expressed by testicular germ cells. Part 4:
in human spermatozoa and its distinction from Intercellular bridges, mitochondria, nuclear enve-
the cytoplasmic droplet. Reprod Biol Endocrinol. lope, apoptosis, ubiquitination, membrane/voltage-­
2012;10:92. gated channels, methylation/acetylation, and
195. Bianco JA, Shafer RB. Radionuclide methods in the transcription factors. Microsc Res Tech. 73, 364–408.
assessment of left ventricular function. Am J Med Sci. 211. Hermo L, Oko R, Morales CR. Secretion and endocy-
1979;277(3):244–254. tosis in the male reproductive tract: A role in sperm
196. Dym M, Fawcett DW. The blood-testis barrier in the rat maturation. Int Rev Cytol. 1994;154:106–189.
and the physiological compartmentation of the semi- 212. Toshimori K, Yoshinaga K, Tanii I, Wakayama T,
niferous epithelium. Biol Reprod. 1970;3(3):308–326. Saxena DK, Ohoka T. Protein expression and cell
197. Fabian L, Brill JA. Drosophila spermiogenesis: Big organelle behavior in spermatogenic cells. Kaibogaku
things come from little packages. Spermatogenesis. Zasshi. 2001;76(3):267–279.
2012;2(3):197–212. 213. Thorne-Tjomsland G, Clermont Y, Tang XM.
198. Greenbaum MP, Iwamori N, Agno JE, Matzuk MM. Glucose-6-phosphatase activity of endoplasmic
Mouse TEX14 is required for embryonic germ cell reticulum and Golgi apparatus in spermatocytes and
intercellular bridges but not female fertility. Biol spermatids of the rat: An electron microscopic cyto-
Reprod. 2009;80(3):449–457. chemical study. Biol Cell. 1991;71(1–2):33–41.
References 39

214. Clermont Y, Rambourg A. Evolution of the endoplas- 225. Kierszenbaum AL, Tres LL. Structural and tran-
mic reticulum during rat spermiogenesis. Am J Anat. scriptional features of the mouse spermatid genome.
1978;151(2):191–211. J Cell Biol. 1975;65(2):258–270.
215. Franke WW, Scheer U, Krohne G, Jarasch ED. The 226. Kleene KC. Connecting cis-elements and trans-
nuclear envelope and the architecture of the nuclear factors with mechanisms of developmental regula-
periphery. J Cell Biol. 1981;91(3 Pt 2):39s–50s. tion of mRNA translation in meiotic and haploid
216. Kessel RG. Origin, differentiation, distribution and mammalian spermatogenic cells. Reproduction.
possible functional role of annulate lamellae dur- 2013;146(1):R1–19.
ing spermatogenesis in Drosophila melanogaster. 227. Hecht NB. Regulation of ‘haploid expressed genes’ in
J Ultrastruct Res. 1981;75(1):72–96. male germ cells. J Reprod Fertil. 1990;88(2):​679–693.
217. Kessel RG. The annulate lamellae—From obscurity 228. Reynolds N, Collier B, Maratou K et al. Dazl binds in
to spotlight. Electron Microsc Rev. 1989;2(2):257–348. vivo to specific transcripts and can regulate the pre-
218. Maul GG. Nuclear pore complexes. Elimination meiotic translation of Mvh in germ cells. Hum Mol
and reconstruction during mitosis. J Cell Biol. Genet. 2005;14(24):3899–3909.
1977;74(2):492–500. 229. Grellscheid SN, Dalgliesh C, Rozanska A et al.
219. Merisko EM. Annulate lamellae: An organelle in Molecular design of a splicing switch responsive to
search of a function. Tissue Cell. 1989;21(3):343–354. the RNA binding protein Tra2beta. Nucleic Acids Res.
220. Morozova KN, Gubanova NV, Kiseleva EV. 2011;39(18):8092–8104.
[Structural organization and possible functional role 230. Vourekas A, Zheng Q, Alexiou P et al. Mili and Miwi
of annulate lamellae containing cytoplasmic pores]. target RNA repertoire reveals piRNA biogenesis and
Tsitologiia. 2005;47(8):667–678. function of Miwi in spermiogenesis. Nat Struct Mol
221. Soderstrom KO. Annulate lamellae in the sperma- Biol. 2012;19(8):773–781.
tids of the rat. Z Mikrosk Anat Forsch. 1981;95(5):​ 231. Schroder M, Kaufman RJ. ER stress and the unfolded
845–854. protein response. Mutat Res. 2005;569(1–2):29–63.
222. Bird DJ, Seiler MW. 1986. Annulate lamellae and 232. Chappell VA, Busada JT, Keiper BD, Geyer CB.
single-pore complexes in human spermatogonia. Translational activation of developmental messenger
J Submicrosc Cytol. 18:823–828. RNAs during neonatal mouse testis development.
223. Dym M, Cavicchia JC. Functional morphology of the Biol Reprod. 2013;89(3):61.
testis. Biol Reprod. 1978;18(1):1–15. 233. Emr S, Glick BS, Linstedt AD et al. Journeys through
224. Papp S, Robaire B, Hermo L. Immunocytochemical the Golgi—Taking stock in a new era. J Cell Biol.
localization of the Ya, Yc, Yb1, and Yb2 subunits 2009;187(4):449–453.
of glutathione S-transferases in the testis and epi-
didymis of adult rats. Microsc Res Tech. 1995;30(1):​
1–23.
Androgen regulation
of spermatogenesis
2
WILLIAM H. WALKER

SPERMATOGENESIS TESTOSTERONE FUNCTIONS


Male fertility is dependent on the continuous process of Testosterone is required to maintain spermatogenesis.1,2
spermatogenesis, the production of sperm. Spermatogenesis Testosterone deprivation studies performed in rodents
occurs within the seminiferous tubules of the testis with established that the steroid hormone is required for
somatic Sertoli cells surrounding and supporting the germ cells to progress beyond meiosis and for the release
developing germ cells (Figure 2.1). Peritubular myoid cells of mature spermatids (reviewed in Sharpe1). Decreased
form webs that encircle the seminiferous tubules while intratesticular testosterone levels or elimination of
providing additional factors supporting spermatogen- androgen receptor (AR, NR3C4), the receptor for tes-
esis and also contract to force sperm through the tubules tosterone and other androgens, disrupts four major pro-
toward the rete testis and the epididymis. In the intersti- cesses in the testis that are required for spermatogenesis
tial region between seminiferous tubules, Leydig cells pro- and male fertility. First, the BTB cannot be maintained
duce testosterone and blood vessels provide nutrients and in the absence of testosterone signaling, resulting in the
growth factors from serum. loss of meiotic and postmeiotic germ cells. 3,4 Second,
Spermatogenesis initiates with spermatogonial stem meiosis cannot be completed. Third, round spermatids
cells located along the interior basement membrane of are prematurely released as they are differentiating into
the seminiferous tubule dividing to produce B spermato- elongated spermatids. 5–7 Fourth, mature spermatozoa
gonia (nonhuman primates and men) or undifferentiated cannot be released, resulting in the phagocytosis of the
spermatogonia (rodents). The spermatogonia undergo germ cells by Sertoli cells.7 Although these physiologi-
a series of mitotic divisions with incomplete cytokinesis cal effects of testosterone have been known for decades,
to form chains of developing germ cells along the base- it is only during the past few years that there has been
ment membrane. In the presence of retinoic acid signal- some better understanding of the molecular mecha-
ing, a final spermatogonial mitosis produces preleptotene nisms by which testosterone acts.
spermatocytes that are committed to undergo meiosis.
The preleptotene spermatocytes detach from the basement ANDROGEN RECEPTOR FUNCTIONAL DOMAINS
membrane and “pass through” the blood-testis barrier The effects of testosterone and other androgens are medi-
(BTB), which is required for spermatogenesis. The BTB is ated by AR, a 110 kDa protein member of the steroid hor-
a collection of adhesion junctions formed between adja- mone receptor family encoded by a single gene on the
cent supporting Sertoli cells near the basement membrane X chromosome. AR has numerous well-­characterized
that separates the basal portion of the tubule containing structurally and functionally distinct domains (reviewed
spermatogonia from the apical (luminal) portion that con- in Heinlein and Chang8 and Lonergan and Tindall9)
tains meiotic and post-meiotic germ cells. The BTB is dis- including the poorly conserved N-terminal domain
solved above the preleptotene spermatocyte and reformed (NTD) encoded by exon 1 that contains a proline-
below as the germ cell moves toward the lumen of the rich region (PRR) required for interactions with Src
tubule. After passing through the BTB, meiosis continues kinase10,11 (Figure  2.2). There is also a highly conserved
through the lengthy leptotene, zygotene, pachytene, and DNA-binding domain (DBD) including two zinc fingers
diplotene stages of prophase I followed by the rapid com- encoded by exons 2 and 3 that recognize specific DNA
pletion of meiosis I and II cell divisions that result in hap- sequences called androgen response elements (AREs).
loid round spermatids. The spermatids undergo extensive A ligand-binding domain (LBD) also contains a nuclear
differentiation (spermiogenesis) including condensation export sequence that is encoded by exons 4 through 8.
of the nucleus and DNA, termination of transcription, Another portion of exon 4 encodes a short hinge region
elimination of much of the cytoplasm, acrosome forma- that separates the LBD from the DBD and contains a
tion, cell elongation, and development of a flagellum. ligand-dependent nuclear localization signal (NLS).
Spermatogenesis is completed with the release of mature Domains encoding transcription activation functions
elongated spermatids as spermatozoa (spermiation) into are located in the N terminus (AF-1, AF5) and in the LBD
the lumen of the tubule (reviewed in Sharpe1). (AF-2).

40
Androgen receptor functional domains  41

Seminiferous tubule

PTM cell

Spermatogonial GDNF Pachytene Round


spermatocyte spermatid
stem cell
GDNF Elongated
spermatid
Leptotene Mature
spermatocyte spermatozoa

Testosterone Sertoli cell


Leydig cells Testo BTB
stero
ne

Testoste
rone

Spermatogonia

Figure 2.1  Somatic cells supporting spermatogenesis. Leydig cells in the interstitial space between seminiferous tubules pro-
vide testosterone to themselves, Sertoli, and pertitubular myoid (PTM) cells. The PTM and Sertoli cells produce glial cell-derived neu-
rotrophic factor (GDNF) to support the self-renewal of spermatogonial stem cells. The blood-testis barrier (BTB) is shown between
adjacent Sertoli cells. The progression of germ cell development from spermatogonial stem cell through mature spermatozoa is
shown.

AR gene

5′ Exon 1 2 3 4 5 6 7 8 3′

AR protein
NTD DBD LBD

1 PRR 537 625 669 919


Hinge
NLS
AF-1 AF-5 AF-2

Figure 2.2  Androgen receptor (AR) structure. The eight exons of human AR are shown. Exon 1 encodes the amino terminal
domain (NTD) that includes the proline rich region (PRR) that interacts with Src kinase. Exons 2 and 3 encode the DNA binding
domain (DBD) and exons 4 through 8 encode the hinge region and ligand binding domain (LBL). Within the hinge region is the
nuclear localization signal (NLS). Transcription activation domains 1, 2, and 5 (AF-1, AF-2, AF-5) are shown. Amino acid residues that
separate domains are shown below the AR protein.
42  Androgen regulation of spermatogenesis

TESTOSTERONE PRODUCTION with levels of testosterone (10–250 nM) that are similar to
The major sites of testosterone production in men are the or lower than those found in the testis (200–800 nM)1,13,14
Leydig cells located in the interstitial regions of the tes- activated the nonclassical pathway, including the phos-
tis. Testosterone is produced in response to luteinizing phorylation of ERK and CREB, and increased the tran-
hormone binding to its receptor on Leydig cells, which scription of CREB-regulated genes, including CREB itself,
causes the induction of the steroidigenic machinery that is LDH-A and EGR.24 Thus, the nonclassical pathway also
responsible for testosterone production and secretion. Due regulates gene expression downstream of kinase activa-
to the intratesticular location of the Leydig cells, testos- tion. Together, these findings indicate that the nonclassical
terone concentrations in the testes of men (340 to 2000 nM) pathway has rapid effects such that increased phosphor-
are  25- to 125-fold greater than that in serum (8.75 to ylation and activation of Src, ERK, and CREB can be
35  mM). Testosterone is similarly elevated in the rodent detected within 1 minute with gene expression regulated
testis and is the major androgen in the testis, exceeding the at later time points. Moreover, the pathway has prolonged
levels of dihydrotestosterone (DHT) by 15- to 40-fold.12–16 effects through increased protein phosphorylation that
The bioavailable levels of testosterone in the testis far can be sustained for at least 12 hours and via long-term
exceed that required for function, as the binding affinity of gene expression changes.24
AR for testosterone is approximately 1 nM and regulation Recently, the first in vivo assays of nonclassical testos-
of gene expression via AR binding requires 1 to 10 nM.17 terone signaling were performed for Sertoli cells within rat
The physiological importance of high testosterone levels in testes. Adult rats were treated with a GnRH antagonist to
the testis is not well understood, but it is known that high decrease intratesticular testosterone levels. After 7 hours,
testosterone levels are required to maintain spermatogen- testicular testosterone concentrations fell to 23% of con-
esis because sperm counts decrease at a logarithmic rate as trol and the levels of phosphorylated ERK kinase (p-ERK)
testosterone levels fall below 70 nM in the rodent.18 in stage VII seminiferous tubule cross sections decreased
to 34% of controls. Subsequent injections of testosterone
TESTOSTERONE SIGNALING PATHWAYS increased intratesticular testosterone to 125% of controls
Testosterone has been shown to act via two pathways: the and restored p-ERK levels in stage VII tubules to control
classical and the nonclassical.19,20 In the classical pathway, levels within 1 hour.25 The rapid testosterone-mediated
testosterone diffuses through the plasma membrane and increase in p-ERK levels is not likely to result from clas-
binds AR that is sequestered by heat shock proteins in the sical testosterone signaling because accumulation of tes-
cytoplasm. A conformational change in AR causes the tosterone in the testis would require some latent period
receptor to be released from the heat shock proteins. AR and testosterone-mediated gene expression and protein
then translocates to the nucleus where it binds to AREs production requires more than 45 minutes.21 These studies
in gene promoter regions, recruits coregulator proteins, provided the first evidence that nonclassical testosterone
and regulates gene transcription. Activation of the clas- signaling occurs in vivo.
sical pathway requires at least 30 to 45 minutes to detect Both the nonclassical and classical signaling pathways
the first increases in gene transcription, with protein-­ are required to maintain spermatogenesis. Adenovirus
mediated effects occurring even later.21 constructs expressing either a peptide that inhibits AR-Src
In the nonclassical pathway (also called nongenomic) kinase interaction (nonclassical inhibitor) or a modi-
characterized in Sertoli cells, testosterone stimulation ini- fied AR that acts as a dominant negative inhibitor of
tiates cellular events within seconds that can be observed AR-mediated transcription (classical inhibitor) were
within minutes. Thus far, two rapidly activated nonclas- delivered into the seminiferous tubules and Sertoli cells
sical pathways have been described in Sertoli cells. In the of mice. Both inhibitors disrupted the integrity of the
first pathway, testosterone stimulation of cultured Sertoli BTB. In addition, the nonclassical inhibitor resulted in
cells isolated from rats transiently increases AR localiza- the loss of spermatocytes in adult mice. Similar studies of
tion to the region near the plasma membrane and trig- prepubertal mice indicated that the nonclassical inhibitor
gers the direct association of the proline-rich region of blocked the progression of meiosis and the production of
AR (amino acids 352–359) and the SH3 domain of Src spermatocytes. In contrast, the major effect of the classical
tyrosine kinase.22,23 Studies employing Sertoli cells from inhibitor was the premature release of meiotic and post-
AR-deficient testicular feminized (tfm) rats or siRNA meiotic germ cells. The combined effects of both inhibitors
against AR in wild-type Sertoli cells determined that resulted in a synergistic disruption of spermatogenesis.
AR was essential to drive this nonclassical testosterone Together, these studies indicate that both signaling path-
pathway. Testosterone-mediated activation of Src causes ways contribute to the maintenance of spermatogenesis.25
the phosphorylation and stimulation of the EGF recep- A second rapid-acting nonclassical pathway has been
tor (EGFR).22 Stimulation of EGFR is required to acti- described in which testosterone causes depolarization of
vate the MAP kinase cascade (Raf, MEK, ERK) that in the Sertoli cell within 30 seconds due to the closing of
turn activates p90Rsk kinase to phosphorylate the CREB K+ATP channels via G protein-mediated activation of phos-
transcription factor.22 It is likely that there are numerous pholipase C. As a result, there is rapid influx of Ca 2+ and
other downstream targets of the kinases activated by the the activation of intracellular signaling factors (reviewed
nonclassical pathway. Stimulation of cultured Sertoli cells in Loss et al.26). This second nonclassical pathway has only
Does testosterone limit the number of gonocytes?  43

been described in immature Sertoli cells. It is not known Evidence supporting the idea that AR actions do not influ-
whether AR is required to activate the pathway or whether ence gonocyte development was found in mice having the
the pathway contributes to Sertoli cell function in mature androgen receptor knocked out in all cells (ARKO mice).
Sertoli cells. Other studies performed in the Sertoli cell In this model, gonocyte migration to the basement mem-
line 93RS2 that lacks AR showed that testosterone acted brane of seminiferous tubules was not altered. Also, gono-
via the Zn2+ transporter ZIP9 to activate ERK1/2, CREB cyte transformation into spermatogonia stem cells and
and ATF1, stimulate Claudin 1 and Claudin 5 expression, the expansion of spermatogonia was not dependent on
and enhance tight junction formation between Sertoli AR based on the finding that wild-type and ARKO mice
cells.27 Thus far, it has not been determined whether the had similar numbers of germ cells from E 17.5 through
ZIP9-mediated signaling pathway is relevant to Sertoli P10.42 The idea that testosterone signaling via AR does not
cells present in vivo that express AR. regulate gonocyte development was supported by another
study of 1-day-old mice in which gonocytes are the only
CELL- AND TEMPORAL-SPECIFIC EXPRESSION OF AR germ cells present. At this age, the number of germ cells,
In the adult testis, AR is expressed in peritubular myoid, Sertoli cells, and Leydig cells were not altered 1 day after
Leydig, Sertoli, arteriole smooth muscle, and vascu- birth in ARKO mice.43 A different study that assessed the
lar endothelial cells. In peritubular myoid cells AR is number of germ cells in 2-day-old mice found that the
expressed from the fetal period through adulthood in men number of gonocytes present in ARKO mice exceeded
and rodents.28–31 Fetal and adult Leydig cells express AR at those present in wild-type mice.44 However, the apparent
constant levels.32 Sertoli cells in rodents first express AR difference in ARKO versus wild-type germ cell numbers at
3 to 5 days after birth.33–36 In men, AR is expressed at low 1 and 2 days after birth may be accounted for by the meth-
levels in Sertoli cells beginning about 5 months after birth ods by which the cells were counted. Specifically, for the
and remains weak until four years of age.28–31 In the adult 2-day-old mice only, the relative nuclear volumes of germ
rodent, AR levels are low in Sertoli cells throughout most cells normalized to the nuclear volumes of Sertoli cells was
of the cycle of the seminiferous epithelium except during used to compare gonocyte numbers. Using this method,
stages VI–VIII when AR levels peak.34,37,38 AR expres- the apparent increase in germ cells could be accounted for
sion in men is also stage-specific with the highest levels by normalizing to the nuclear volume of Sertoli cells.
of AR expressed during stage III of the six stages.39 The In contrast to the ARKO mouse studies, the Habert
stage-specific peaks in AR expression that are observed group showed that tfm mice that are androgen-insensitive
in Sertoli cells of the rodent correspond with the initia- due to global mutation of the androgen receptor had 40%
tion of testosterone-dependent processes that are essential more gonocytes at E17.5. The increase in gonocytes was
for spermatogenesis, including the formation of special- associated with an increased percentage of gonocytes that
ized adhesion junctions with elongating spermatids and were proliferating at E15.5 but not at later stages of devel-
the release of mature sperm that occur in stage VIII. The opment. In addition, the number of gonocytes in testis
higher levels of AR also coincide with the appearance of explants from wild-type mice was decreased by treatment
preleptotene spermatocytes and the initiation of meiosis with DHT.45 These data argue that testosterone acts to
during stages VII–VIII. Thus, it is possible that testosterone- limit the number of gonocytes during fetal development
mediated paracrine signals from Sertoli cells to prelepto- by yet-to-be-defined mechanisms.
tene spermatocytes may be responsible for the production Previous studies showed that AR was not present in fetal
of factors that are required later to complete meiosis. germ cells.46 In contrast, immunoreactive AR was found
There is less information available regarding the poten- in gonocyte nuclei at E 15.5 and AR transcripts were pres-
tial importance of limiting AR levels and testosterone ent in gonocytes purified from E15.5 and E17.5 mouse tes-
signaling in Sertoli cells during much of the remaining tis. Furthermore, a functional assay of AR responsiveness
stages of cycle of the seminiferous epithelium. One study suggests that an AR is active in fetal gonocytes at these
of a transgenic mouse model in which high levels of AR times.45,47 These observations raise an important question:
were expressed in Sertoli cells from the promoter of the Does testosterone act through AR in fetal gonocytes to
androgen-binding protein gene caused reduced spermato- limit their proliferation, and if so, then why is there not an
genesis because Sertoli cells matured prematurely, halted increase in the number of gonocytes in ARKO mice?
proliferation, and the fewer Sertoli cells present could Although it is possible that AR could be expressed in
only support smaller populations of germ cells. However, gonocytes, and there are periodic reports of androgen
no information was provided about any direct effects of receptors being expressed in developing mammalian germ
increased AR expression on processes that were required cells,48–52 there is general consensus that AR is either not
for spermatogenesis in adults.40,41 present or not essential in germ cells after birth. Three
functional studies indicate that if expressed, AR is not
DOES TESTOSTERONE LIMIT THE NUMBER required in germ cells to complete spermatogenesis. First,
OF GONOCYTES? spermatogonial stem cells isolated from tfm mice were able
There is some controversy regarding whether testoster- to complete spermatogenesis after transplantation into
one regulates the migration and number of gonocyte (also wild-type mice.53 Second, spermatogenesis is not affected
known as prospermatogonia) germ cells in the fetal testis. in knockout mice in which AR expression is eliminated
44  Androgen regulation of spermatogenesis

beginning in early meiosis.54 Third, chimeric mice hav- Other studies of PTM-ARKO mice determined that
ing both AR-defective and wild-type germ cells produced although Sertoli cell numbers, maturation markers, and
pups from the AR defective germ cells.55 These findings the expression of AR were not affected, the expression of
reinforce the prevailing idea that germ cells respond to some AR-regulated genes in Sertoli cells was decreased
testosterone signaling only indirectly via paracrine and and the size of the seminiferous epithelium lumen was
justacrine signals from somatic cells. reduced. These last observations suggest that there are sig-
nals from the PTM cell to the Sertoli cell that control gene
TESTOSTERONE SUPPORTS THE EXPANSION expression and the production of seminiferous tubule
OF SPERMATOGONIA fluid that carries nutrients to germ cells and developing
The expansion of germ cells within the first days after sperm.57
birth is affected by androgen signaling because by PD5, It is interesting to note that the number of spermato-
when only spermatogonial stem cells and the first differ- gonia was similar in Sertoli cell AR knockout (SCARKO)
entiating spermatogonia are present, germ cell numbers and wild-type mice, suggesting that AR actions in Sertoli
were reduced in ARKO mice by approximately 60%.43 In cells do not have a large effect on the maintenance of sperm­
contrast, testosterone does not appear to alter the numbers togonial stem germ cells or their conversion to spermato-
of spermatogonia that are present later in the developing gonia. In Sertoli cells, there is no evidence yet that Gdnf
and adult mouse because testosterone treatment initiated expression is regulated by AR. Instead, follicle-stimulating
at day 21 and continuing for 6 weeks did not increase the hormone (FSH) is a major hormonal stimulator of Gdnf
number of spermatogonia in hpg mice that lack gonado- RNA expression in these cells.68,69
tropins and thus have low endogenous levels of testos- There is now evidence that testosterone signaling in
terone.56 These apparently contradictory results could be Sertoli cells can indirectly regulate gene expression in
explained by testosterone not having an effect on the pro- spermatogonia. The introduction of adenovirus vectors
liferation and survival of differentiating spermatogonia, expressing androgen receptor into the Sertoli cells of
whereas testosterone-mediated signals soon after birth SCARKO mice resulted in the activation of the ZBTB16
may be required to support the population of spermatogo- (PLZF) and c-Kit genes that in testis are only expressed in
nial stem cells that are expanding at that time. spermatogonia and support the expansion of undifferen-
Peritubular cell-specific AR knockout (PTM-ARKO) tiated and differentiated spermatogonia, respectively.25 It
mice display a progressive decrease in spermatogonia also was found that the nonclassical and classical pathways
and marked reductions in spermatocytes and spermatids provide different signals to spermatogonia because only
that cause the mice to become azoospermic.57 A poten- activation of the classical pathway permitted the reestab-
tial mediator of peritubular signals required to maintain lishment of ZBTB16 transcription, whereas activation of
spermatogonia is glial cell-derived neurotrophic factor classical or nonclassical signaling resulted in increased
(GDNF). GDNF acts through the GFRA1 and RET core- expression of the c-Kit gene.
ceptor complex to support the proliferation and mainte-
nance of spermatogonial stem cells and undifferentiated TESTOSTERONE AND AR ARE REQUIRED
spermatogonia.58–61 A relatively small window of optimal TO COMPLETE MEIOSIS
GDNF production is required to maintain spermato- There is significant loss of spermatocytes and sperma-
genesis as shown by the collapse of spermatogenesis due tids in rodents, especially in stages VII–VIII of the cycle
to global mutation of one GDNF allele or by constitu- of the seminiferous epithelium after reducing intrates-
tive expression of GDNF after the introduction of a Gdnf ticular testosterone levels by hypophysectomy or GnRH
transgene.62 GDNF is a growth factor that is expressed antagonist treatment as well as in hpg mice that lack
by Sertoli cells63–65 and peritubular cells.66,67 Testosterone gonadotropins. However, the loss of these germ cells can
was recently found to induce the production of GDNF be reversed under these conditions by testosterone treat-
by peritubular cells. In cultures of enriched peritubular ment (reviewed in O’Shaughnessy70). AR is required for
cells, testosterone stimulation increased the expression of testosterone support of meiosis because spermatocytes
GDNF mRNA and protein. Further studies showed that numbers are reduced and very few round spermatids are
testosterone stimulation of cocultures of peritubular cells produced in ARKO and tfm mice.43,71 Germ cell meiosis
and undifferentiated spermatogonia increased the pro- requires AR activity in Sertoli cells as shown by the pro-
liferation of the spermatogonia. Finally, the number and gressive loss of primary spermatocytes between stages
size of germ cell colonies observed after transplantation VI–VII in SCARKO mice.72 The stage-specific loss of
into recipient mice was increased after coculture of undif- germ cells in SCARKO mice matches the stages in which
ferentiated spermatogonia with peritubular cells in the AR expression levels normally peaks in Sertoli cells,
presence of testosterone.66,67 Together, these data raise the implicating AR-mediated signals from Sertoli cells as
possibility that in PTM-ARKO mice, GDNF production being essential for maintaining meiosis. In support of this
by peritubular cells is decreased due to the lack of testos- idea, SCARKO mice have meiosis arrested in the pachy-
terone signaling, resulting in insufficient GDNF signaling tene or diplotene stage of prophase I such that meiosis I
activity for stem cell self-renewal, which could explain the cell division does not occur and few haploid spermatids
progressive loss of spermatogonia in this mouse model. are produced.54,72 Together, these findings indicate that
Testosterone is required to maintain the blood-testis barrier  45

testosterone and AR in Sertoli cells are required for the from Sertoli cells was blocked. The unreleased elongated
pachytene spermatocytes to enter into meiotic divisions spermatids were observed to be degenerating between
that will produce round spermatids and for the survival Sertoli cells or were phagocytized by Sertoli cells and
of the spermatocytes. transported to the basement membrane for recycling.7,74,75
These hypomorph SCARKO mouse results match those
Meiotic processes and factors regulated by testosterone: In the
from earlier testosterone suppression models that caused
absence of testosterone signaling, male germ cells enter
the premature detachment of step 8 round spermatids.6,76
into meiosis I and progress through the leptotene and
The premature release of spermatids in the absence of
zygotene stages of meiosis to initiate the pairing of the
testosterone signaling may be explained by the changes in
chromosomes (synapsis) and the production of double-
the Sertoli-spermatid adhesion complex that occur dur-
strand DNA breaks that are required for later recombi-
ing the transition to elongated spermatids. During stage
nation events and the formation of chiasmata that are
8 when round spermatids initiate their elongation process,
needed for separation of the chromosomes. However,
the adhesion complexes between Sertoli cells and round
during the pachytene stage of meiosis, synapsis is not
spermatids are transformed from predominately desmo-
completed and double-strand breaks are not repaired,
some type connections to the specialized ectoplasmic
which inhibits recombination and desynapsis such that
specialization (ES) adherans junction that is unique to the
85% of spermatocytes had univalent chromosomes.73
testis.77 It is possible that the expression or localization of
These events result in the induction of cell cycle check-
proteins needed for the formation of the ES and elongated
point responses that lead to cell cycle delay or arrest
spermatid attachment may be dependent on AR signal-
and the apoptosis of the germ cells. The triggering of
ing in Sertoli cells because suppression of intratesticular
the checkpoint responses explains the halting of mei-
testosterone decreases the levels of proteins present at ES
osis during pachytene and the apoptosis of pachytene
junctions, including β1-integrin, p-FAK, and c-Src via the
spermatocytes.
ERK signaling pathway.78
One explanation for cell cycle arrest and apoptosis in
The paradox of how testosterone can be essential for
the absence of testosterone signaling is the loss of proteins
both the adhesion of elongating spermatids and the release
that are essential for the completion of meiosis. In sper-
of mature elongated spermatids (sperm) appears to be
matocytes of SCARKO mice, expression was decreased
resolved. The ES at the site of spermiation is disassembled
for RAD51 and DMC1 proteins that are required for the
and the components are relocated and recycled for use at
resolution of double-strand DNA breaks, recombina-
the site of new spermatid adhesion sites by 22 hours before
tion, and chromosome separation. There is also decreased
the final release of sperm during stage VII.79 Thus, the ES
expression of other proteins (TEX15, BRCA1, BRCA2, and
is not present when the sperm are released. Instead, sperm
PALB2) that regulate the loading of RAD51 and DMC1
adhesion to Sertoli cells is maintained via the tubulobul-
onto the sites of double-strand breaks.73 Thus far, it is not
bar and focal adhesion-related disengagement complexes.
known whether testosterone signals from Sertoli cells first
Testosterone signaling supports the disengagement of
act during the pachytene stage of meiosis on the RAD51,
sperm from these complexes.75 The O’Donnell group
DMRT, and associated proteins or earlier in leptotene and
provided evidence that only the disengagement complex
zygotene stages to permit correct chromosome synapsis,
remains associated with sperm that fail to be released in
which is essential for the later events in meiosis. The mech-
the absence of testosterone. This complex retains α6β1
anisms by which testosterone and AR-mediated signals
integrin and tyrosine-phosphorylated FAK from the ES.75
are delivered to the meiotic cells are not well understood.
Notably, FAK can be phosphorylated in response to non-
However, increased signaling by members of the EGF-
classical testosterone signaling and by Src kinase that is
EGFR family of growth factors in the testis was detected
activated by the same pathway. Furthermore, inhibition
in SCARKO mice and may be one mechanism by which
of Src kinase activity resulted in lower levels of sperm
decreased AR signaling may alter signals to germ cells.73
release.80 The idea that testosterone-mediated phosphory-
lation of components of the disengagement complex enable
TESTOSTERONE IS REQUIRED FOR SPERMATID sperm release also is consistent with findings that activa-
ATTACHMENT AND RELEASE OF SPERM tion of nonclassical signaling target ERK kinase occurs at
Most SCARKO mouse models do not provide informa- the time and location of sperm release in cultured semi-
tion about testosterone support of spermiogenesis because niferous tubules.81
spermatogenesis is arrested in meiosis. However, the
hypomorph SCARKO model from Braun’s group that TESTOSTERONE IS REQUIRED TO MAINTAIN
displays partial AR activity provided important informa- THE BLOOD-TESTIS BARRIER
tion about testosterone-regulated events during sperma- Testosterone is required for maintaining the BTB that
tid maturation. This model showed that progression of consists of numerous cellular junctions between Sertoli
round spermatids to elongated spermatids is sensitive to cells.3,4 The BTB is present near the basement membrane
the reduction in AR activity in Sertoli cells such that the of the seminiferous tubule and separates the basal com-
round spermatids are released prematurely. As well, ter- partment that has access to the circulatory system from
minal differentiation and release of elongated spermatids the luminal compartment that is dependent on the Sertoli
46  Androgen regulation of spermatogenesis

cell to provide nutrients and growth factors. All germ Jam3, and Claudin 3.3,4,91 However, presently it is not
cell development beyond the leptotene stage of meiosis known whether the expression of genes encoding BTB
occurs in the specialized environment beyond the BTB. proteins is enhanced by classical or nonclassical signaling
Spermatogenesis in rodents is halted after structural dis- in vivo.
ruption of the BTB.82,83 However, the extent to which BTB
disruptions affect fertility in men is less well understood. TESTIS GENE EXPRESSION IS REGULATED
Studies of cultured Sertoli cells showed that testosterone BY TESTOSTERONE
increased tight junction function as measured by transepi-
thelial resistance (TER).84–86 Testosterone also stimulated Altered testosterone signaling and SCARKO models:  Surveys
the expression of mRNA encoding a major tight junction of gene expression in various rodent models have been
protein N-cadherin and stimulated the localization of performed in the presence and absence of testosterone
Claudin 11 to Sertoli-Sertoli contact sites.86,87 Follow-up signaling. The various models include neonatal wild-
studies indicated that testosterone signaling increases type or adult hpg mice injected with testosterone pro-
steady-state levels of clatherin and clatherin association pionate for 4 to 24 hours,94,98,99 comparisons of normal
with tight junction proteins occludin and Claudin 11. In vs tfm mice,100 AR hypomorph mice,101 and four other
addition, testosterone facilitates the transcytosis and recy- SCARKO models.3,91,92,102 These studies of testosterone-
cling of tight junction proteins that are associated with the and AR-regulated gene expression have all employed
dissolution of the BTB as spermatocytes transit through isolation of mRNAs from the whole testis. One common
the barrier allowing the rapid reformation of the BTB that theme for nearly all of the models was the finding that
occurs below the spermatocytes.88,89 a greater percentage of genes were downregulated by
Claudin 3 is an adhesion protein that is induced by AR and testosterone. Nevertheless, these studies deter-
androgen and localizes to newly forming tight junctions at mined that testosterone signaling increased the expres-
the BTB.4 However, Claudin 3 deficient mice retain fertility, sion of genes encoding the BTB proteins Claudin 3 and
suggesting that other androgen-regulated junctional pro- Claudin 11,99 genes associated with vitamin A metabo-
teins are sufficient to maintain the BTB.90 Claudin 11 and lism, solute transport, or cytoskeletal function,100 as
occludin, major transmembrane components of the BTB, well as genes involved in metabolic processes and signal
were found to be downregulated in SCARKO mice.3,91,92 transduction.103 The SCARKO mouse models identi-
The mRNAs encoding Tjp1, Tjp2 iso1, Tjp2 iso2, and Tjp2 fied AR repressed genes associated with MAP kinase
iso3 that link the transmembrane tight junction proteins and serine endoprotease activity plus upregulated genes
to the cytoskeleton are also downregulated in SCARKO needed for signal transduction, cell adhesion, Ca2+ bind-
mice, suggesting the possibility that these genes are reg- ing, Ca2+-dependent phospholipid binding, and serine
ulated by testosterone. These results are consistent with protease inhibition.92 Also, in SCARKO models there
the finding that in SCARKO mice the BTB is formed but was differential expression of genes encoding proteases,
formation is delayed and the barrier is incomplete.3 Rats cell adhesion proteins, cytoskeletal elements, extra-
injected with flutamide to block AR actions had decreased cellular matrix components,3 tight junction proteins,
expression of the Cx43 gap junction mRNA and protein basement membrane components, tissue remodeling
and the loss of Cx43 localization to the BTB, whereas the factors, paracrine factors for the support of germ cells,
tight junction associated ZO-1 protein was mislocalized transport proteins,91 as well as cell adhesion, nuclear
away from the cell surface to the Sertoli cell cytoplasm.93 localization, and meiosis.102
DHT treatment of hpg mice restored the expression and RiboTag mouse models:  An important advance in the
localization of Claudin 3, Cx-43, and Claudin 11 to the identification of cell-specific changes in AR-regulated
BTB.94 mRNAs is the use of transgenic mice expressing the
Inhibition of either classical or nonclassical testoster- RiboTag, which is the ribosomal protein S6 having an
one signaling disrupted the BTB in vivo as injection of HA epitope tag. The expression of the RiboTag allows
mouse testis seminiferous tubules with adenovirus con- for immunoprecipitation with antisera directed against
structs expressing inhibitors of either pathway resulted the HA epitope that will isolate ribosomes and any
in decreased expression of Claudin 11 at the BTB and associated RNA. Conditional expression of the RiboTag
increased transit of a biotin tracer through the BTB and using a cell-specific cre recombinase allows the isolation
into the lumen of the tubules.25 The finding that the non- of the population of RNAs in the process of translation
classical testosterone signaling pathway is required for from individual cell types (the translome). The initial
maintaining the BTB is consistent with previous studies RiboTag strategy using a Sertoli cell-specific cre recom-
showing that ERK kinase activity and Src family kinases binase combined with microarray analysis did not
components of the nonclassical pathway are required for reveal an extensive list of AR-regulated mRNAs specific
maintaining the BTB.95–97 The finding that classical testos- to Sertoli cells perhaps due the short time period of tes-
terone signaling is required to maintain the BTB is sup- tosterone stimulation (4 hours).104
ported by previous studies showing that AR induces the A different RiboTag approach was used in which mRNA
expression of genes encoding BTB-associated proteins expression was compared in 10-day-old mice express-
including N-cadherin, Claudin 11, Occludin, Gelsolin, ing the RiboTag in an otherwise wild-type background
Summary 47

(SCRIBO mice) versus a SCARKO background to maintain Sertoli cell tight junctions and the BTB.105
(SCARIBO mice). Analysis of the ribosome-­associated Although AR-regulated genes contribute to the other
RNAs by RNA-seq yielded the most extensive list testosterone-mediated processes, few are known to
of AR-regulated genes in the testis to date. Without be essential. It is possible that the combined effects of
first isolating mRNAs from ribosomes of specific cell AR-regulated gene expression may be required to sup-
types, RNA-seq analysis identified 938 AR-regulated port specific stages of spermatogenesis. However, fur-
genes from all testis cells in 10-day-old mice with ther studies are required to identify the mechanisms by
slightly more genes being stimulated than inhibited which testosterone and AR support specific processes
by AR. About 20% (187) of the genes were differen- required for spermatogenesis.
tially expressed by twofold or more. The use of the
RiboTag strategy to isolate mRNAs associated with FUTURE CHALLENGES
the ribosomes of Sertoli cells identified 1090 mRNAs Cell-specific AR knockout mouse models have provided
that were differentially expressed in response to the important phenotypic information regarding the func-
loss of AR. The AR-regulated genes enriched in Sertoli tions of testosterone in the testis, including testosterone-
cells included those that encoded proteins associated mediated actions within targeted cells and effects on
with calcium ion, actin, or cytoskeletal protein bind- neighboring cells. The next goal will be to understand
ing, proteins active at the plasma membrane, adhesion how testosterone-mediated signals are transferred to other
functions including cell-cell junction and anchoring cells. For example, results from SCARKO mice suggest that
junctions, as well as proteins located at the stereocil- AR-mediated EGF signals originating from Sertoli cells
ium. AR was found to be more likely to repress (59%) must be limited to permit meiosis progression through
than activate (41%) Sertoli cell enriched gene expres- prophase I.73 However, it is not known how testosterone
sion. Of the 1090 Sertoli cell enriched AR-regulated acts via AR to decrease EGF signaling to spermatocytes or
genes found using the RiboTag strategy, 55% escaped which stage of meiosis must be protected from EGF. It is
detection in the less specific whole testis SCARKO vs likely that additional AR-regulated paracrine signals from
wild-type (SCARIBO vs SCRIBO) comparison and Sertoli and other cells remain to be identified. How these
38% were differently expressed by two-fold or more. signals act to support meiosis and other germ cell pro-
This result indicates that the RiboTag strategy has cesses will need to be determined. Once these questions
greater sensitivity than a total testis approach for assay- are addressed, we will then need to understand the intra-
ing differential gene expression. However, as with other cellular signaling events and cellular responses that occur
surveys of AR-regulated genes in the testis, few genes after the paracrine signals are received. After these issues
known to be essential for fertility were identified in the are understood, the information may be used to identify
list of AR-regulated genes. Nevertheless, several of the potential sources of defects causing male infertility or tar-
identified genes were known to contribute to processes gets for contraception strategies.
important for male fertility, including Tgfb2, which
is involved in SC barrier formation and maintenance; SUMMARY
Nr5a2, an activator of steroidogenic enzymes; and Testosterone-mediated alterations in the expression,
Timp1, an inhibitor of matrix metalloproteinases. It is phosphorylation, and localization of proteins associ-
possible that repeating the survey using mature mice ated with adhesion complexes in Sertoli cells are essen-
would reveal additional AR-regulated genes that are tial for the maintenance and continuous reestablishment
required for fertility. of the BTB, the adhesion of elongating spermatids, and
The RiboTag approach also found 445 mRNAs that are dif- the release of mature sperm. Additional, less well char-
ferentially expressed in the absence of AR in Sertoli cells acterized paracrine and juxtacrine signals from Sertoli
but are enriched in other cell types. These non-Sertoli cells are required for spermatocytes to complete meiosis.
cell mRNAs encoded proteins involved in binding to Although testosterone is essential for many processes that
adenyl ribonucleotides, adenyl nucleotides, nucleosides, are required to complete spermatogenesis, it is also useful
and ATP, which may suggest that androgen signaling in to consider critical spermatogenesis processes for which
SCs leads them to instruct other testicular cells to alter testosterone signaling is not required. At present, avail-
their nucleic acid and energy metabolism. The use of able evidence suggests that testosterone is not essential for
similar RiboTag strategies could be used in the future to (1) the survival and proliferation of spermatogonial stem
identify genes in germ cells at specific stages of develop- cells required to continually replenish germ cells, (2) the
ment that are regulated by testosterone and AR actions proliferation of differentiating spermatogonia needed
in Sertoli and other somatic cells of the testis. to expand the population of germ cells, (3) the entry
Of the major spermatogenesis processes regulated by into meiosis, and (4) the cellular restructuring required
testosterone, including spermatogonia proliferation, for differentiation of elongating spermatids. Decades of
BTB formation, meiosis, spermatid attachment, and research has brought us to a good understanding of the
spermiation, thus far AR-regulated gene expression spermatogenesis process that are and are not regulated by
can be shown to be directly linked to the production testosterone. We hope that future studies will provide a
of proteins (Claudin 11, Occludin) that are essential better understanding of the intracellular and extracellular
48  Androgen regulation of spermatogenesis

signaling strategies that are used by testosterone to sup- 15. Awoniyi CA, Sprando RL, Santulli R et al.
port spermatogenesis. Restoration of spermatogenesis by exogenously
administered testosterone in rats made azoospermic
REFERENCES by hypophysectomy or withdrawal of luteinizing
1. Sharpe RM. Regulation of spermatogenesis. In Knobil hormone alone. Endocrinology. 1990;127:177–184.
E, Neil JD (eds.). The Physiology of Reproduction. 16. Maddocks S, Hargreave TB, Reddie K et al.
New York: Raven Press;1994:1363–1434. Intratesticular hormone levels and the route of secre-
2. McLachlan R.I, O’Donnell L, Meachem SJ et al. tion of hormones from the testis of the rat, guinea pig,
Identification of specific sites of hormonal regula- monkey and human. Int J Androl. 1993;16:272–278.
tion in spermatogenesis in rats, monkeys, and man. 17. Tsai MJ, O’Malley BW. Molecular mechanisms of
Recent Prog Horm Res. 2002;57:149–179. action of steroid/thyroid receptor superfamily mem-
3. Willems A, Batlouni SR, Esnal A et al. Selective bers. Ann Rev Biochem. 1994;63:451–486.
ablation of the androgen receptor in mouse Sertoli 18. Zirkin BR, Santulli R, Awoniyi CA, Ewing LL.
cells affects Sertoli cell maturation, barrier for- Maintenance of advanced spermatogenic cells in the
mation and cytoskeletal development. PLoS One adult rat testis: Quantitative relationship to testoster-
2010;5:e14168. one concentration within the testis. Endocrinology.
4. Meng J, Holdcraft RW, Shima JE, Griswold MD, 1989;124:3043–3049.
Braun RE. Androgens regulate the permeability of 19. Walker WH. Molecular mechanisms of testosterone
the blood-testis barrier. Proc Natl Acad Sci U S A. action in spermatogenesis. Steroids. 2009;74:602–607.
2005;102:16696–16700. 20. Walker WH. Non-classical actions of testosterone
5. O’Donnell L, McLachlan RI, Wreford NG, Robertson and spermatogenesis. Philos Trans R Soc Lond B Biol
DM. Testosterone promotes the conversion of round Sci. 2010;365:1557–1569.
spermatids between stages VII and VIII of the 21. Shang Y, Myers M, Brown M. Formation of the
rat spermatogenic cycle. Endocrinology. 1994;135:​ androgen receptor transcription complex. Mol Cell.
2608–2614. 2002;9:601–610.
6. O’Donnell L, McLachlan RI, Wreford NG, de Kretser 22. Cheng J, Watkins SC, Walker WH. Testosterone acti-
DM, Robertson DM. Testosterone withdrawal pro- vates MAP kinase via Src kinase and the EGF receptor
motes stage-specific detachment of round sper- in Sertoli cells. Endocrinology. 2007;148:2066–2074.
matids from the rat seminiferous epithelium. Biol 23. Migliaccio A, Castoria G, Di Domenico M et al.
Reprod. 1996;55:895–901. Steroid-induced androgen receptor-oestradial recep-
7. Holdcraft RW, Braun RE. Androgen receptor func- tor β-Src complex triggers prostate cancer cell prolif-
tion is required in Sertoli cells for the terminal dif- eration. EMBO J. 2000;20:5406–5417.
ferentiation of haploid spermatids. Development. 24. Fix C, Jordan C, Cano P, Walker WH. Testosterone
2004;131:459–467. activates mitogen-activated protein kinase and the
8. Heinlein CA, Chang C. Androgen receptor in pros- cAMP response element binding protein transcrip-
tate cancer. Endocr Rev. 2004;25:276–308. tion factor in Sertoli cells. Proc Natl Acad Sci U S A.
9. Lonergan PE, Tindall DJ. Androgen receptor signal- 2004;101:10919–10924.
ing in prostate cancer development and progression. 25. Toocheck C, Clister T, Shupe J et al. Mouse sper-
J Carcinog. 2011;10:20. matogenesis requires classical and nonclassical tes-
10. Migliaccio A, Varricchio L, De Falco A et al. tosterone signaling. Biol Reprod. 2016;94:11.
Inhibition of the SH3 domain-mediated binding of 26. Loss ES, Jacobus AP, Wassermann GF. Rapid sig-
Src to the androgen receptor and its effect on tumor naling responses in Sertoli cell membranes induced
growth. Oncogene. 2007;26:6619–6629. by follicle stimulating hormone and testosterone:
11. Auricchio F, Migliaccio A, Castoria G. Sex-steroid Calcium inflow and electrophysiological changes.
hormones and EGF signalling in breast and prostate Life Sci. 2011;89:577–583.
cancer cells: Targeting the association of Src with ste- 27. Bulldan A, Dietze R, Shihan M, Scheiner-Bobis G. Non-
roid receptors. Steroids. 2008;73:880–884. classical testosterone signaling mediated through ZIP9
12. Hammond GL, Koivisto M, Kouvalainen K, Vihko stimulates claudin expression and tight junction for-
R. Serum steroids and pituitary hormones in infants mation in Sertoli cells. Cell Signal. 2016;28:1075–1085.
with particular reference to testicular activity. J Clin 28. Shapiro E, Huang H, Masch RJ et al. Immuno​local-
Endocrinol Metab. 1979;49:40–45. ization of androgen receptor and estrogen receptors
13. Comhaire FH, Vermeulen A. Testosterone concen- alpha and beta in human fetal testis and epididymis.
tration in the fluids of seminiferous tubules, the J Urol. 2005;174:1695–1698.
interstitium and the rete testis of the rat. J Endocrinol. 29. Berensztein EB, Baquedano MS, Gonzalez CR et al.
1976;70:229–235. Expression of aromatase, estrogen receptor alpha and
14. Turner TT, Jones CE, Howards SS et al. On the beta, androgen receptor, and cytochrome P-450scc
androgen microenvironment of maturing spermato- in the human early prepubertal testis. Pediatr Res.
zoa. Endocrinology. 1984;115:1925–1932. 2006;60:740–744.
References 49

30. Chemes HE, Rey RA, Nistal M et al. Physiological 44. Tan KA, De Gendt K, Atanassova N et al. The
androgen insensitivity of the fetal, neonatal, and role of androgens in Sertoli cell proliferation and
early infantile testis is explained by the ontogeny of functional maturation: Studies in mice with total
the androgen receptor expression in Sertoli cells. J or Sertoli cell-selective ablation of the androgen
Clin Endocrinol Metab. 2008;93:4408–4412. receptor. Endocrinology. 2005;146:2674–2683.
31. Boukari K, Meduri G, Brailly-Tabard S et al. Lack 45. Merlet J, Racine C, Moreau E, Moreno SG, Habert R.
of androgen receptor expression in Sertoli cells Male fetal germ cells are targets for androgens that
accounts for the absence of anti-Mullerian hormone physiologically inhibit their proliferation. Proc Natl
repression during early human testis development. Acad Sci U S A. 2007;104:3615–3620.
J Clin Endocrinol Metab. 2009;94:1818–1825. 46. Williams K, McKinnell C, Saunders PT et al. Neonatal
32. Rey RA, Musse M, Venara M, Chemes HE. Ontogeny exposure to potent and environmental oestrogens
of the androgen receptor expression in the fetal and and abnormalities of the male reproductive system
postnatal testis: Its relevance on Sertoli cell matura- in the rat: Evidence for importance of the androgen-
tion and the onset of adult spermatogenesis. Microsc oestrogen balance and assessment of the relevance to
Res Tech. 2009;72:787–795. man. Hum Reprod Update. 2001;7:236–247.
33. Buzek SW, Sanborn BM. Increase in testicular 47. Merlet J, Moreau E, Habert R, Racine C. Development
androgen receptor during sexual maturation in the of fetal testicular cells in androgen receptor deficient
rat. Biol Reprod. 1988;39:39–49. mice. Cell Cycle. 2007;6:2258–2262.
34. Bremner WJ, Millar MR, Sharpe RM, Saunders 48. Galena HJ, Pillai AK, Terner C. Progesterone and
PT. Immunohistochemical localization of andro- androgen receptors in non-flagellate germ cells of
gen receptors in the rat testis: Evidence for stage- the rat testis. J Endocrinol. 1974;63:223–237.
dependent expression and regulation by androgens. 49. Sanborn BM, Elkington JS, Steinberger E. Properties
Endocrinology. 1994;135:1227–1234. of rat testicular androgen binding proteins. Curr Top
35. Zhou X, Kudo A, Kawakami H, Hirano H. Mol Endocrinol. 1974;1:291–310.
Immunohistochemical localization of ­ androgen 50. Wright WW, Frankel AI. An androgen receptor in
receptor in mouse testicular germ cells ­during fetal and the nuclei of late spermatids in testes of male rats.
postnatal development. Anat Rec. 1996;​245:509–518. Endocrinology. 1980;107:314–318.
36. You L, Sar M. Androgen receptor expression in the 51. Frankel AI, Chapman JC, Wright WW. The equivo-
testes and epididymides of prenatal and postnatal cal presence of nuclear androgen binding proteins in
Sprague-Dawley rats. Endocrine. 1998;9:253–261. mammalian spermatids and spermatozoa. J Steroid
37. Vornberger W, Prins G, Musto N.A, Suarez-Quian Biochem. 1989;33:71–79.
CA. Androgen receptor distribution in rat testis: 52. Vornberger W, Prins G, Musto NA, Suarez-Quian
New implications for androgen regulation of sper- CA. Androgen receptor distribution in rat testis:
matogenesis. Endocrinology. 1994;134:2307–2316. New implications for androgen regulation of sper-
38. Shan L-X, KZhu L-J, Bardin CW, Hardy MP. matogenesis. Endocrinology. 1994;134:2307–2316.
Quantitative analysis of androgen receptor messen- 53. Johnston DS, Russell LD, Friel PJ, Griswold MD.
ger ribonucleic acid in developing Leydig cells and Murine germ cells do not require functional andro-
Sertoli cells by in situ hybridization. Endocrinology. gen receptors to complete spermatogenesis fol-
1995;136:3856–3862. lowing spermatogonial stem cell transplantation.
39. Suarez-Quian CA, Martinez-Garcia F, Nistal M, Endocrinology. 2001;142:2405–2408.
Regadera J. Androgen receptor distribution in adult 54. Tsai MY, Yeh SD, Wang RS et al. Differential effects
human testis. J Clin Endocrin Metab. 1999;84:350–358. of spermatogenesis and fertility in mice lacking
40. Hazra R, Corcoran L, Robson M et al. Temporal role of androgen receptor in individual testis cells. Proc Natl
Sertoli cell androgen receptor expression in spermato- Acad Sci U S A. 2006;103:18975–18980.
genic development. Mol Endocrinol. 2012;27(1):12–24. 55. Lyon MF, Glenister PH, Lamoreux ML. Normal
41. Hazra R, Upton D, Desai R et al. Elevated expres- spermatozoa from androgen-resistant germ cells of
sion of the Sertoli cell androgen receptor disrupts chimaeric mice and the role of androgen in sper-
male fertility. Am J Physiol Endocrinol Metab. matogenesis. Nature. 1975;258:620–622.
2016;311:E396–404. 56. Haywood M, Spaliviero J, Jimemez M, King NJ,
42. Li R, Vannitamby A, Meijer J, Southwell B, Hutson Handelsman DJ, Allan CM. Sertoli and germ cell
J. Postnatal germ cell development during mini- development in hypogonadal (hpg) mice express-
puberty in the mouse does not require androgen ing transgenic follicle-stimulating hormone alone
receptor: Implications for managing cryptorchidism. or in combination with testosterone. Endocrinology.
J Urol. 2015;193:1361–1367. 2003;144:509–517.
43. O’Shaughnessy PJ, Monteiro A, Abel M. Testicular 57. Welsh M, Saunders PT, Atanassova N, Sharpe RM,
development in mice lacking receptors for follicle Smith LB. Androgen action via testicular peritubu-
stimulating hormone and androgen. PLoS One. lar myoid cells is essential for male fertility. FASEB J.
2012;7:e35136. 2009;23:4218–4230.
50  Androgen regulation of spermatogenesis

58. Jijiwa M, Kawai K, Fukihara J et al. GDNF-mediated 72. De Gendt K, Swinnen JV, Saunders PT et al. A Sertoli
signaling via RET tyrosine 1062 is essential for main- cell-selective knockout of the androgen receptor
tenance of spermatogonial stem cells. Genes Cells. causes spermatogenic arrest in meiosis. Proc Natl
2008;13:365–374. Acad Sci U S A. 2004;101:1327–1332.
59. Buageaw A, Sukhwani M, Ben-Yehudah A et al. 73. Chen SR, Hao XX, Zhang Y et al. Androgen receptor
GDNF family receptor alpha1 phenotype of sper- in Sertoli cells regulates DNA double-strand break
matogonial stem cells in immature mouse testes. Biol repair and chromosomal synapsis of spermatocytes
Reprod. 2005;73:1011–1016. partially through intercellular EGF-EGFR signaling.
60. He Z, Jiang J, Kokkinaki M, Golestaneh N, Hofmann Oncotarget. 2016;7:18722–18735.
MC, Dym M. Gdnf upregulates c-Fos transcrip- 74. Saito K, O’Donnell L, McLachlan RI, Robertson DM
tion via the Ras/Erk1/2 pathway to promote mouse Spermiation failure is a major contributor to early sper-
spermatogonial stem cell proliferation. Stem Cells. matogenic suppression caused by hormone withdrawal
2008;26:266–278. in adult rats. Endocrinology. 2000;141:2779–2785.
61. Hofmann MC. Gdnf signaling pathways within the 75. Beardsley A, O’Donnell L. Characterization of nor-
mammalian spermatogonial stem cell niche. Mol mal spermiation and spermiation failure induced
Cell Endocrinol. 2008;288:95–103. by hormone suppression in adult rats. Biol Reprod.
62. Meng X, Lindahl M, Hyvonen ME et al. Regulation 2003;68:1299–1307.
of cell fate decision of undifferentiated spermatogonia 76. Cameron DF, Muffly KE, Nazian SJ. Reduced testos-
by GDNF. Science. 2000;287:​1489–1493. terone during puberty results in a midspermiogenic
63. Fouchecourt S, Godet M, Sabido O, Durand P. Glial lesion. Proc Soc Exp Biol Med. 1993;202:457–464.
cell-line-derived neurotropic factor and its receptors 77. Mruk D, Cheng C. Desmosomes in the testis: Moving
are expressed by germinal and somatic cells of the rat into an unchartered territory. Spermatogenesis.
testis. J Endocrinol. 2006;190:59–71. 2011;1:47–51.
64. Katoh-Semba R, Tsuzuki M, Miyazaki N et al. 78. Wong CH, Xia W, Lee NP et al. Regulation of ecto-
Distribution and immunohistochemical localization plasmic specialization dynamics in the seminiferous
of GDNF protein in selected neural and non-neural epithelium by focal adhesion-associated proteins in
tissues of rats during development and changes in testosterone-suppressed rat testes. Endocrinology.
unilateral 6-hydroxydopamine lesions. Neurosci Res. 2005;146:1192–1204.
2007;59:277–287. 79. O’Donnell, L, Nicholls PK, O’Bryan MK, McLachlan
65. Johnston DS, Olivas E, DiCandeloro P, Wright WW. RI, Stanton PG. Spermiation: The process of sperm
Stage-specific changes in GDNF expression by rat release. Spermatogenesis. 2011;1:14–35.
Sertoli cells: A possible regulator of the replication 80. Shupe J, Cheng J, Puri P, Kostereva N, Walker WH.
and differentiation of stem spermatogonia. Biol Regulation of Sertoli-germ cell adhesion and sperm
Reprod. 2011;85:763–769. release by FSH and nonclassical testosterone signal-
66. Chen LY, Brown PR, Willis WB, Eddy EM. Peritubular ing. Mol Endocrinol. 2011;25:238–252.
myoid cells participate in male mouse spermato- 81. Chapin RE, Wine RN, Harris MW, Borchers CH,
gonial stem cell maintenance. Endocrinology. 2014;​ Haseman JK. Structure and control of a cell-cell adhe-
155:4964–4974. sion complex associated with spermiation in rat sem-
67. Chen LY, Willis WD, Eddy EM. Targeting the Gdnf iniferous epithelium. J Androl. 2001;22:1030–1052.
gene in peritubular myoid cells disrupts undifferen- 82. Hew K.W, Heath GL, Jiwa AH, Welsh MJ. Cadmium
tiated spermatogonial cell development. Proc Natl in vivo causes disruption of tight junction-­associated
Acad Sci U S A. 2016;113:1829–1834. microfilaments in rat Sertoli cells. Biol Reprod.
68. Ding LJ, Yan GJ, Ge QY et al. FSH acts on the pro- 1993;49:840–849.
liferation of type A spermatogonia via Nur77 that 83. Wiebe JP, Kowalik A, Gallardi RL, Egeler O, Clubb
increases GDNF expression in the Sertoli cells. FEBS BH. Glycerol disrupts tight junction-associated
Lett. 2011;585:2437–2444. actin microfilaments, occludin, and microtubules in
69. Lamberti D, Vicini E. Promoter analysis of the gene Sertoli cells. J Androl. 2000;21:625–635.
encoding GDNF in murine Sertoli cells. Mol Cell 84. Janecki A, Jakubowiak A, Steinberger A. Regulation
Endocrinol. 2014;394:105–114. of  transepithelial electrical resistance in two-
70. O’Shaughnessy PJ. Hormonal control of germ cell compartment Sertoli cell cultures: In vitro model
development and spermatogenesis. Semin Cell Dev of the blood-testis barrier. Endocrinology. 1991;​
Biol. 2014;29:55–65. 129:1489–1496.
71. Johnston H, Baker PJ, Abel M et al. Regulation 85. Janecki A, Jakubowiak A, Steinberger A. Effects
of Sertoli cell number and activity by follicle- of cyclic AMP and phorbol ester on transepithe-
stimulating hormone and androgen during post- lial electrical resistance of Sertoli cell monolayers
natal development in the mouse. Endocrinology. in two-compartment culture. Mol Cell Endocrinol.
2004;145:318–329. 1991;82:61–69.
References 51

86. Kaitu’u-Lino TJ, Sluka P, Foo CF, Stanton PG. 96. Li MW, Xia W, Mruk DD et al. Tumor necrosis fac-
Claudin-11 expression and localisation is regu- tor {alpha} reversibly disrupts the blood-testis barrier
lated by androgens in rat Sertoli cells in vitro. and impairs Sertoli-germ cell adhesion in the semi-
Reproduction. 2007;133:1169–1179. niferous epithelium of adult rat testes. J Endocrinol.
87. Florin A, Maire M, Bozec A et al. Androgens and post- 2006;190:313–329.
meiotic germ cells regulate claudin-11 expression in 97. Li MW, Mruk DD, Lee WM, Cheng CY. Disruption
rat Sertoli cells. Endocrinology. 2005;146:1532–1540. of the blood-testis barrier integrity by bisphenol A
88. Su L, Mruk DD, Lee WM, Cheng CY. Differential in vitro: Is this a suitable model for studying blood-
effects of testosterone and TGF-beta3 on endo- testis barrier dynamics? Int J Biochem Cell Biol.
cytic vesicle-mediated protein trafficking  events 2009;41:2302–2314.
at the blood-testis barrier. Exp Cell Res. 98. Zhou Q, Shima JE, Nie R, Friel PJ, Griswold MD.
2010;316:2945–2960. Androgen-regulated transcripts in the neonatal
89. Yan HH, Mruk DD, Lee WM, Cheng CY. Blood-testis mouse testis as determined through microarray
barrier dynamics are regulated by testosterone and analysis. Biol Reprod. 2005;72:1010–1019.
cytokines via their differential effects on the kinetics 99. Sadate-Ngatchou PI, Pouchnik DJ, Griswold MD.
of protein endocytosis and recycling in Sertoli cells. Identification of testosterone-regulated genes in
FASEB J. 2008;22:1945–1959. testes of hypogonadal mice using oligonucleotide
90. Chakraborty P, Buaas FW, Sharma M et al. microarray. Mol Endocrinol. 2004;18:422–433.
Androgen-dependent Sertoli cell tight junction 100. O’Shaughnessy PJ, Abel M, Charlton HM, Hu
remodeling is mediated by multiple tight junction B, Johnston H, Baker PJ. Altered expression of
components. Mol Endocrinol. 2014;28:1055–1072. genes involved in regulation of vitamin A metabo-
91. Wang RS, Yeh S, Chen LM et al. Androgen recep- lism, solute transportation, and cytoskeletal func-
tor in Sertoli cell is essential for germ cell nursery tion in the androgen-insensitive tfm mouse testis.
and junctional complex formation in mouse testes. Endocrinology. 2007;148:2914–2924.
Endocrinology. 2006;147:5624–5633. 101. Eacker SM, Braun, RE. Androgen receptor in Leydig
92. Denolet E, De Gendt K, Allemeersch J et al. The effect cell function and development. In Payne AH, Hardy
of a Sertoli cell-selective knockout of the androgen MP, eds. Contemporary Endocrinology: The Leydig
receptor on testicular gene expression in prepubertal Cell in Health and Disease. Totowa, New Jersey:
mice. Mol Endocrinol. 2006;20:321–334. Humana Press; 2007:345–365.
93. Chojnacka K, Hejmej A, Zarzycka M et al. Flutamide 102. de Rooij DG, Okabe M, Nishimune Y. Arrest of sper-
induces alterations in the cell-cell junction ultra- matogonial differentiation in jsd/jsd, Sl17H/Sl17H, and
structure and reduces the expression of Cx43 at cryptorchid mice. Biol Reprod. 1999;61:​842–847.
the blood-testis barrier with no disturbance in the 103. Eacker SM, Shima JE, Connolly CM et al.
rat seminiferous tubule morphology. Reprod Biol Transcriptional profiling of androgen receptor
Endocrinol. 2016;14:14. (AR) mutants suggests instructive and permissive
94. McCabe MJ, Allan CM, Foo CF, Nicholls PK, roles of AR signaling in germ cell development. Mol
McTavish KJ, Stanton PG. Androgen initiates Sertoli Endocrinol. 2007;21:895–907.
cell tight junction formation in the hypogonadal 104. Sanz E, Evanoff R, Quintana A et al. RiboTag analysis
(hpg) mouse. Biol Reprod. 2012;87:38. of actively translated mRNAs in Sertoli and Leydig
95. Xiao X, Mruk DD, Lee WM, Cheng CY. c-Yes regu- cells in vivo. PLoS One. 2013;8:e66179.
lates cell adhesion at the blood-testis barrier and the 105. McCabe MJ, Foo CF, Dinger ME, Smooker PM, and
apical ectoplasmic specialization in the seminifer- Stanton PG. Claudin-11 and occludin are major con-
ous epithelium of rat testes. Int J Biochem Cell Biol. tributors to Sertoli cell tight junction function, in
2011;43:651–665. vitro. Asian J Androl. 2016;18:620–626.
Testicular immunoregulation
The role of Tyro3, Axl, and Mer receptor tyrosine
3
kinases and pattern recognition receptors
FEI WANG, QIAN JIANG, and DAISHU HAN

INTRODUCTION regions and a common highly conserved intracellular


Mammalian spermatogenesis is a unique male germ cell tyrosine kinase domain.5 Different subfamilies of RTKs
differentiation process occurring in the testis that requires are categorized based on their sequence identities and
a special microenvironment. From an immunological per- extracellular structural similarities, and the members
spective, the testis possesses a remarkable immunoprivi- of a subfamily often bind to common or similar ligands.
leged environment essential for the protection of germ TAM receptors belong to one subfamily of RTKs. Two
cells from detrimental immune responses and adopts an highly similar vitamin-K–dependent proteins, the prod-
effective innate local defense mechanism against micro- uct of growth-arrest–specific gene 6 (Gas6) and a negative
bial infection.1,2 Disruption of the testicular immune regulator of blood coagulation, protein S (ProS), are two
homeostasis may impair spermatogenesis and result in common ligands of TAM receptors.6 Gas6 and ProS are
male infertility. composed of N-terminal gamma-carboxylated glutamic
Spermatogenesis is completed during puberty, a long acid (GLA) followed by four epidermal growth-factor
time after the establishment of immune self-tolerance. (EGF)-like and C-terminal sex-hormone-binding globu-
Therefore, late-stage male germ cells synthesize a large lin (SHBG)-like domains (Figure 3.1). TAM receptors have
number of immunogenetic molecules that are strang- a similar structure, composed of two immunoglobulin
ers to the immune system. However, these germ cells are (Ig)-like domains in N-terminal and two fibronectin type
immunologically tolerated in the testis under physiologi- III (FNIII) domains in extracellular regions, followed by
cal conditions because of its immunoprivileged environ- a transmembrane domain and an intracellular protein
ment. Multiple mechanisms are involved in the regulation tyrosine kinase (TK) domain at C-terminal. The SHBG-
of this testicular immunoprivileged environment. The role like domains of ligands interact with the Ig-like domains
of Tyro3, Axl, and Mer (TAM) receptor tyrosine kinases in of TAM receptors, initiating signaling pathways through
regulating immune privilege in the testis has been investi- the activation of intracellular TK. Studies using  gene-
gated recently and a review of TAM functions in the testis knockout mice have provided direct insights into the
is provided in this chapter. physiological functions of TAM receptors. TAM signaling
Although the testis is an immunoprivileged organ, a pathways regulate several important biological processes,
broad spectrum of microorganisms, including viruses, including immune responses, phagocytosis of apoptotic
bacteria, and parasites, may infect the testis via circula- cells, and cell proliferation.7
tory dissemination or the ascending genitourinary tract.
Role of TAM receptors in regulating immune
However, microbial infection usually recovers without a
homeostasis
significant systemic immune response, suggesting that the
testis is well-equipped with an effective innate defense sys- Genetic mutations in TAM receptors result in chronic
tem against microbial infection. Various leukocytes can inflammatory and autoimmune diseases in mice, sug-
be found in the interstitial spaces of the testis, and these gesting that TAM signaling plays an important role in
immune cells may play roles in counteracting invading maintaining immune homeostasis.8 TAM signaling
microorganisms.3 Recent studies have demonstrated that contributes to immune homeostasis by regulating two
tissue-specific cells, including somatic and germ cells, are biological processes: the inhibition of innate immune
also equipped with innate immune machinery.4 Sertoli, responses and the promotion of phagocytic clearance of
Leydig, and male germ cells abundantly express vari- apoptotic cells.9
ous pattern recognition receptors (PRRs), which initiate Innate immune responses are initiated by micro-
innate immune responses in the testicular cells. This chap- bial pathogens through the activation of PRRs. Toll-
ter also describes PRR-initiated innate immune responses like receptors (TLRs) are the most well-characterized
in testicular cells and their effects on testicular function. PRRs10 and TLR signaling in innate immune cells, such
as dendritic cells (DCs) and macrophages, leads to the
TAM receptor tyrosine kinases production of numerous cytokines and chemokines,
Receptor tyrosine kinases (RTKs) belong to a superfam- recruiting and activating leukocytes to counteract
ily of transmembrane proteins with diverse extracellular invading microorganisms. The TLR-initiated innate

52
Introduction 53

N TAM receptors in the testis


GLA domain The role of TAM receptors in the testis was initially rec-
ognized using TAM triple knockout (TAM−/−) mice.14
Ligands: Although mutations of individual members or any com-
Gas6, ProS bination of two receptors do not affect mouse fertility,
EGF-like domains TAM−/− male mice are sterile because of spermatogenesis
degeneration. To investigate the mechanisms by which
N
SHBG-like domain TAM receptors regulate spermatogenesis, TAM receptor
and Gas6 expression in the mouse testis has been exam-
C
Ig domains ined.15 All three TAM receptors are expressed in Sertoli
cells. Axl and Mer, but not Tyro3, are expressed in Leydig
cells, and Gas6 is exclusively expressed in Leydig cells.
TAM receptors:
Tyro3, Axl, Mer FN III domains By contrast, neither TAM receptors nor their ligands are
expressed in male germ cells. The distribution of the TAM
system in the testis suggests that impaired spermatogen-
esis in TAM−/− mice is not germ cell autonomous, and the
Cell membrane degeneration of TAM−/− somatic cell functions contributes
to the impairment of spermatogenesis.
TK domain
Notably, TAM−/− male mice fulfill the first wave of
spermatogenesis, producing a few aberrant elongated
C sperm. Evident defective spermatogenesis initially appears
5 weeks postnatally, following the onset of sexual matura-
tion.16 As TAM−/− mice age, male germ cells are progres-
Immune Phagocytosis sively lost, beginning from elongated spermatids to round
Cell proliferation
responses of apototic cells spermatids, spermatocytes, and spermatogonia. TAM−/−
mice have a normal testosterone level, suggesting that
Figure 3.1  Schematic structure of TAM receptors and their defective spermatogenesis is attributable to impairment of
ligands. TAM receptors contain three members: Tyro3, Axl, and Sertoli cell functions.
Mer. TAM receptors are composed of two Ig-like domains at
N-terminal followed by two extracellular fibronectin type III Role of TAM receptors in the phagocytosis of apoptotic
(FN III) domains, a transmembrane domain, and an intracellu- germ cells
lar protein tyrosine kinase domain at C-terminal. The product Sertoli cells, the only somatic cells in the seminifer-
of growth arrest specific gene 6 (Gas6) and protein S (ProS) ous epithelium, play a critical role in spermatogenesis,
are ligands of TAM receptors. Gas6 and ProS share a common and one Sertoli cell function is the phagocytic removal
structure composed of an N-terminal gamma-carboxylated of apoptotic germ cells and residual bodies. More than
glutamic acid (GLA) domain, four epidermal growth factor 70% of male germ cells undergo apoptosis during sper-
(EGF)-like domains, and a C-terminal sex hormone binding matogenesis. Moreover, the most cytoplasmic portions of
globulin (SHBG)-like domain. The SHBG-like domain of ligand elongated spermatids form residual bodies and are shed
binds to the Ig domain of TAM receptors, thereby activating before sperm are released from the seminiferous epithe-
TAM receptors. TAM receptor activation initiates intracellu- lium. Timely phagocytic removal of apoptotic germ cells
lar signaling to regulate several important cellular processes and residual bodies is critical for maintaining seminifer-
such as proliferation, uptake of apoptotic cells, and immune ous epithelial homeostasis.17 The apoptotic germ cells and
responses. residual bodies are recycled after phagocytosis by Sertoli
cells as a source of energy, suggesting that apoptotic germ
cells and residual bodies are more than mere wastes.18
immune response also directs the adaptive immune Notably, the TAM/Gas6 system plays an important role
response against microbial infection.11 Although TLR in regulating the phagocytosis of apoptotic germ cells
activation in innate immune cells is critical in the by Sertoli cells.19 All three TAM receptors recognize and
host defense against microbial infection, TLR-initiated tether apoptotic cells through Gas6 in a redundant man-
innate immune responses must be tightly regulated, as ner, whereas Mer is responsible for triggering phagocy-
unrestrained TLR signaling generates a chronic inflam- totic signaling (Figure 3.2).
matory condition that may be harmful to the host itself. From an immunological perspective, phagocytic
Therefore, the negative regulation of TLR signaling is removal of apoptotic cells by phagocytes is a critical mech-
essential for immune homeostasis.12 TAM receptors are anism in the elimination of endogenous autoantigens for
important negative regulators of the innate immune the prevention of an autoimmune response.20 Impairment
response and play a critical role in maintaining immune of apoptotic cell clearance is associated with autoimmune
homeostasis.13 diseases.21 Late-stage male germ cells are formed after the
54  Testicular immunoregulation

Sperm

Residual body

Apoptotic germ cell


ATP
Breakdown

Recycle Lysosome

TAM
Residual body
Phagolysosome
DAMPs TRIF Phagosome
TLRs TAM
MyD88 TAM
SOCS1/3
Microbes
MAPKs NF-κB IRFs SOCS1/3 Apoptotic germ cell

AP-1

TAM
AP-1
P NF-κBP P
IRFs ???
TNF-α TAM Gas6
IL-1
IL-6 IFN-α/β
......

Figure 3.2  Functions of TAM receptors in Sertoli cells. Two major functions of TAM receptors in Sertoli cells have been defined.
(1) TAM receptors mediate phagocytosis of residual bodies and apoptotic germ cells by Sertoli cells. The phagosomes are fused
with lysosome to form phagolysosome. The residual bodies and apoptotic cells in the phagolysosome are degraded and recycled
as an energy source for Sertoli cells. (2) TAM receptors negatively regulate toll-like receptor (TLR) signaling pathways. TAM signaling
induces the expression of suppressor of cytokine signaling proteins (SOCS1/3), which inhibit TLR signaling pathways.

establishment of immune self-tolerance and synthesize male germ cells25,26 and HMGB1 is involved in testicu-
a large number of autoantigens that are immunogenetic. lar inflammation.27 The role of endogenous TLR ligands
Phagocytic removal of apoptotic germ cells and residual in testicular endogenous inflammation remains largely
bodies by Sertoli cells can prevent immunogenetic autoan- unknown.
tigens from inducing immune responses. Therefore, TAM
receptors may contribute to testicular immune homeosta- TAM inhibition of innate immune responses
sis by promoting the removal of germ cell autoantigens. in testicular cells
Moreover, damaged germ cells also induce the production TLR-initiated innate immune responses in the testis
of inflammatory cytokines in Sertoli cells through the were initially found in mouse Sertoli cells. 28,29 Given that
activation of TLR2 and TLR4.22 Therefore, apoptotic germ the TAM/Gas6 system is a negative regulator of TLR sig-
cells and residual bodies, if not removed in time, may naling,13 it is reasonable to speculate that TAM recep-
release endogenous TLR ligands and trigger an inflam- tors inhibit TLR-initiated innate immune responses in
matory response through TLR activation (Figure 3.2). The Sertoli cells because TAM are abundantly expressed in
most well-characterized endogenous TLR ligands include these cells. The mechanisms by which TAM receptors
high-mobility group box 1 (HMGB1) and several heat inhibit TLR signaling in Sertoli cells have been investi-
shock proteins (HSPs), which induce endogenous inflam- gated. 30 TAM−/− Sertoli cells display excessive activation
mation through the activation of TLR2 and TLR4.23,24 of TLR3 signaling characterized by the overexpression
Notably, HMGB1 and HSPs are abundantly expressed in of immunoregulatory cytokines. The TAM/Gas6 system
The innate defense system in the testis  55

upregulates the suppression of cytokine signaling, which damage to the seminiferous epithelium. Using the EAO
in turn inhibits TLR-driven expression of cytokines in model, we found that Axl and Mer receptors coopera-
Sertoli cells (Figure 3.2). Several TLRs are also expressed tively regulated systemic immune tolerance to male germ
in mouse Leydig cells and can be activated by their cell antigens.36 Although Axl and Mer double knockout
respective ligands. 31 Axl and Mer are also expressed in (AM−/−) male mice were fertile and had no signs of auto-
Leydig cells and inhibit TLR-initiated innate immune immune orchitis, they were susceptible to EAO induc-
responses. tion and AM−/− mice developed severe EAO after a single
TLR-initiated innate immune responses in Sertoli and immunization with germ cell antigens.36 Mild EAO was
Leydig cells may contribute to testicular defense against observed in Axl−/− and Mer−/− mice after similar immuni-
microbial infection. However, the innate immune response zation. By contrast, a single immunization did not induce
in testicular cells needs to be tightly controlled by the EAO in Tyro3−/− and wild-type (WT) mice. These observa-
negative regulators because an excessive innate immune tions suggest that Axl and Mer, but not Tyro3, cooperatively
response may disrupt testicular immunoprivileged status regulate systemic immune tolerance to male germ cell
and impair spermatogenesis. Accordingly, TAM−/− male antigens. Accordingly, antigen-presenting cells, includ-
mice spontaneously develop autoimmune orchitis charac- ing dendritic cells and macrophages, express Axl and Mer
terized by progressive degeneration of germ cells accom- but not Tyro3,13 and the mechanisms by which Axl and
panying leukocyte infiltration into the testis, upregulation Mer regulate systemic immune tolerance to germ cell anti-
of inflammatory cytokines, disruption of blood-testis gens are worthy of further investigation. TAM−/− devel-
barrier (BTB) integrity, and generation of autoantibodies oped autoimmune orchitis spontaneously,32 suggesting
against male germ cells.32 Notably, autoimmune orchitis that Tyro3 is also involved in the maintenance of testicu-
in TAM−/− mice develops progressively after the onset of lar immunoprivileged status. Considering that Tyro3 is
sexual maturation, and during this period a large number exclusively expressed in Sertoli cells,15 Tyro3 may locally
of residual bodies are formed. We speculate that impair- regulate the testicular immunoprivileged environment via
ment in the phagocytic ability of TAM−/− Sertoli cells modulation of Sertoli cell function, although this remains
slows the timely removal of residual bodies. These residual to be clarified. TAM receptors are also essential for tissue
bodies then release endogenous ligands to activate TLRs homeostasis in other immunoprivileged organs, such as
in Sertoli cells. Excessive activation of TLR signaling in the brain and eye, confirming that TAM receptors play a
TAM−/− Sertoli cells leads to the production of high levels universal role in the regulation of immune privilege.39
of inflammatory cytokines. High levels of tumor necrosis
factor alpha (TNF-α), a major inflammatory cytokine, can THE INNATE DEFENSE SYSTEM IN THE TESTIS
alter BTB permeability,33 resulting in the release of auto- Various immune cells, mainly macrophages in testicular
antigens into the interstitial spaces to induce an autoim- interstitial spaces, are believed to be the frontline against
mune response. A high level of TNF-α also induces germ the arrival of microbial pathogens from circulating blood.
cell apoptosis,34 which may amplify endogenous innate However, testicular macrophages display anti-inflammatory
immune responses. phenotypes in favor of the immunoprivileged environ-
ment.40 Therefore, it is doubtful whether testicular mac-
The role of TAM receptors in immune tolerance to male rophages play a crucial role in counteracting microbial
germ cells pathogens.41 Recent studies have revealed that tissue-
Local immunosuppressive mechanisms are important specific testicular cells, including Leydig, Sertoli, and
for maintaining the immunoprivileged status of the tes- germ cells, are equipped with innate immune machinery
tis. Systemic immune tolerance to autoantigens also con- and are probably involved in testicular defense against
tributes to its immunoprivileged environment. Although microbial infections.
local immunosuppression in the testis has been inten-
sively investigated, the mechanisms underlying the sys- PRRs
temic immune tolerance to germ cell antigens are largely Recognition of microbial pathogens by PRRs is the first
unknown. Considering that TAM receptors inhibit auto- step in the initiation of immune responses.42 PRRs recog-
immune disease,35 they may regulate systemic immune nize the conserved molecular structures that are broadly
tolerance to male germ cell autoantigens. This hypothesis shared by microbial pathogens; namely, pathogen-associated
has been investigated using an experimental autoimmune molecular patterns (PAMPs). Upon PAMP recognition,
orchitis (EAO) model36 in rodent animals, a testis-specific PRRs initiate signaling pathways that execute the first line
autoimmune inflammation model used for the investi- of host defense against invading microbes. Further, PRR-
gation of the pathomechanisms of testicular autoimmu- initiated immune responses direct adaptive immunity, the
nity.37 EAO can be induced in animals by immunization second line of host defense.11
with an injection of allogeneic testicular homogenates, Several PRR families have been identified (Figure 3.3).
male germ cell lysates, or viable male germ cells.38 EAO TLRs, retinoic acid-inducible gene (RIG-I)-like receptors
usually develops after three immunizations and is char- (RLRs), and cytosolic DNA sensors are the most well-
acterized by generation of autoantibodies against germ characterized PRR families43 and these PRRs recognize
cell antigens, infiltration of leukocytes into the testis, and a broad spectrum of PAMPs, thereby initiating signaling
56  Testicular immunoregulation

TLR4

MyD88 TRIF RNA virus

DNA virus

Endosome
TLR7, 8 dsRNA
, 9 TLR3

MyD88 DNA
RIG-I/MDA5
TRIF
TRAF3
IRAKs
IPS-1 DNA sensors
TRAF6 Mitochondrion
Bacterium

IRAKs

TRAF6 STING

TBK1
NEMO MAPKs Endoplasmic reticulum
IKKα IKKβ
MyD88

ISG15
TLR1, 2, 5, 6

AP-1 OAS1 IFNR


IκB IRF3 MX1
NF-κB
......

IFN-α, IFN-β
Nucleus

P P P
NF-κB AP-1 IRF3

IFN-α
IFN-β

TNF-α, IL-1β, IL-6, MCP-1.....

Figure 3.3  PRR-initiated innate immune signaling pathways. TLRs are localized on cellular and endosomal membranes. TLRs
initiate MyD88- and/or TRIF-dependent pathways. MyD88-dependent pathway induces the production of the proinflammatory
cytokines, including TNF-α, IL-1β, IL-6, and MCP-1, through the activation of NF-κB and AP-1. TRIF-dependent pathway induces the
production of the proinflammatory cytokines and IFN-α/IFN-β through the activation of NF-κB and IRF3. RIG-I and MDA5 sense
viral dsRNA, thus initiating an IPS-1-dependent pathway. Cytosolic DNA sensors recognize viral and bacterial DNA, thus initiating a
STING-dependent pathway. Both IPS-1- and STING-dependent pathways induce IFN-α and IFN-β production through IRF3 activa-
tion. The IPS-1-dependent pathway also activates NF-κB and induces proinflammatory cytokine production. IFN-α and IFN-β induce
the expression of the antiviral proteins, including ISG15, OAS1, and MX1, in autocrine and paracrine manners. Abbreviations: AP-1,
activator protein 1; dsRNA, double-strained RNA; IKK, IκB kinase; IPS-1, IFN-β promoter stimulator 1; IRAK, interleukin-1 receptor
associated kinases; IRF3, interferon regulatory factor3; ISG-15, IFN-stimulating gene 15; MAPKs, mitogen-activated protein kinases;
MCP-1, monocyte chemotactic protein 1; MDA5, melanoma differentiation-associated protein 5; MX1, MX GTPase 1; MyD88, myeloid
differentiation protein 88; NEMO, NF-κB-essential modulator; OAS1, 2’5’-digodenylate synthetase 1; RIG-I, retinoic acid-inducible
gene I; STING, stimulator of IFN gene; TBK1, TANK-binding kinase 1; TLR, toll-like receptor; TRAF, tumor necrosis factor receptor-
associated factor; TRIF, toll/IL-1R domain-containing adaptor inducing IFN-β.
The innate defense system in the testis  57

pathways that lead to the secretion of immunoregulatory and TLR5 agonists induces the expression of chemokine
cytokines. These cytokines activate immune cells and MCP-1 and adhesion molecule ICAM-1. These two mol-
directly counteract invading pathogens. To date, 13 TLRs ecules are responsible for the migration of monocytes
have been identified in mammals.44 RLRs include two and lymphocytes from blood vessels to inflamed tissues.50
functional members, RIG-I and melanoma differentiation- Therefore, the activation of TLR2 and TLR5 in Sertoli
associated protein 5 (MDA5), both of which recognize cells may promote leukocyte recruitment in response to
double-stranded (ds) RNA produced during viral replica- microbial infection. We demonstrate that mouse Sertoli
tion, thus coordinating antiviral responses via the induc- cells abundantly express TLR2, TLR3, TLR4, and TLR5,
tion of type 1 IFNs.45 The study of cytosolic DNA sensors and these TLRs are activated by their ligands and induce
has progressed rapidly in recent years. Several types of the expression of proinflammatory cytokines and type
cytosolic DNA sensors, including DNA-dependent activa- 1 IFNs.29 These results suggest that TLR-initiated innate
tor of IFN regulatory factor (DAI), IFN-inducible protein immune responses in Sertoli cells directly counteract
16 (p204 in mouse), and cGMP-AMP synthase (cGAS), microbes. Interestingly, TLR3 activation promotes phago-
have been intensively investigated.46 These DNA sensors cytosis of apoptotic germ cells by Sertoli cells,29 suggesting
recognize viral DNA and initiate antiviral responses. that TLR3 may play a role in regulating tissue homeosta-
sis. Another study confirmed that TLR3 induced antiviral
PRR signaling pathways immune responses in mouse Sertoli cells, showing that
PRRs initiate innate immune responses through several TLR3 ligand induced the expression of proinflammatory
signaling pathways (Figure 3.3). TLRs are type 1 trans- cytokines and IFNs through the activation of IRF3 and
membrane proteins that may be localized on plasma or NF-κB.51 In addition to TLRs, cytosolic RNA and DNA
endosomal membranes. Most TLRs exclusively initiate the sensors have been detected in Sertoli cells at relatively low
myeloid differentiation protein 88 (MyD88)-dependent levels compared with Leydig cells.52,53 The potential roles
pathways, with the exception of TLR3 and TLR4. TLR3 of cytosolic RNA and DNA sensors in testicular defense
exclusively initiates the Toll/IL-1R-domain-containing against viral infection are worthy of investigation.
adaptor-inducing IFN-β (TRIF)-dependent pathway,
whereas TLR4 activation triggers both MyD88- and TRIF- PRR-initiated innate immune responses in Leydig
dependent pathways.47 The MyD88-dependent pathway cells
predominantly induces the secretion of proinflamma- Early studies demonstrated that rat Leydig cells express
tory cytokines and chemokines through the activation IFNs and antiviral proteins.54,55 These observations imply
of nuclear factor kappa B (NF-κB). The TRIF-dependent that rat Leydig cells possess antiviral capacities. However,
pathway activates NF-κB and IFN regulatory factor 3 in response to viral infection, human Leydig cells express
(IRF3), thereby leading to the induction of proinflam- relatively low levels of IFNs and antiviral proteins com-
matory cytokines and type 1 IFNs (IFN-α and IFN-β). pared with rat Leydig cells,56 suggesting that the innate
These cytokines promote the recruitment and activation antiviral response in the human testis may be weaker than
of leukocytes and the induction of IFN-inducible antiviral that in the rat testis. This finding may explain why natu-
proteins, thereby counteracting invading microbial patho- ral viral orchitis is frequently observed in humans but not
gens. Moreover, TLR signaling facilitates the maturation in murine animals. Dissection of the mechanisms under-
of antigen-presenting cells, thereby directing adaptive lying the different antiviral defense systems of murine
immunity. RIG-I and MDA5 are cytosolic RNA sensors and human testes may provide novel strategies for the
and recognize the dsRNA formed by various viruses dur- development of preventive and therapeutic approaches
ing replication. RIG-I and MDA5 activation triggers sig- against viral infection in the testis. Recent studies on PRR-
naling using an adaptor IFN-β promoter stimulator-1 initiated innate immune responses in mouse Leydig cells
(IPS-1) that is localized to mitochondria.48 The cytosolic have expanded our understanding of the innate defense
DNA sensor signaling pathway requires the stimulator system of the mouse testis. Mouse Leydig cells abundantly
of IFN gene (STING) as a common adaptor localized to express TLR2, TLR3, and TLR4.31 TLR3 and TLR4 can be
the endoplasmic reticulum.49 IPS-1-dependent signal- activated by their respective ligands in Leydig cells, initi-
ing results in the activation of IRF3 and NF-κB, which ating innate immune responses inducing the production
induces the expression of type 1 IFNs and proinflamma- of proinflammatory cytokines and type 1 IFNs. Moreover,
tory cytokines, whereas the STING-dependent pathway the activation of TLR3 and TLR4 suppresses testosterone
predominantly induces type 1 IFN production through synthesis in Leydig cells. This suppression is an indirect
the activation of IRF3. effect of TLR-induced proinflammatory cytokines because
several cytokines, including IL-1β, IL-6, and TNF-α,
TLR-initiated innate immune responses in Sertoli inhibit steroidogenesis.57 Therefore, the innate immune
cells responses in testicular cells may have detrimental conse-
The role of TLRs in the testis was initially investigated in quences for spermatogenesis.
mouse Sertoli cells, with Riccioli et al. demonstrating that Viral infection in the male genital system is of consid-
TLR2, TLR4, TLR5, and TLR6 were expressed in mouse erable concern, not only because of its detrimental effect
Sertoli cells.28 Stimulation of Sertoli cells with TLR2 on male fertility, but also the possibility of pathogen
58  Testicular immunoregulation

transmission to sexual partners and the fetus.58 Various TLR3 is expressed in mouse male germ cells.61
viruses, such as mumps virus (MuV), human immuno- Spermatogonia and spermatocytes constitutively express
deficiency virus (HIV), and hepatitis virus, can infect the TLR3, and TLR3 ligands induce the production of pro-
testis and impair testicular function. The innate antiviral inflammatory cytokines and type 1 IFNs. These germ
response in the testis is of particular importance because cells also express antiviral proteins via TLR3 signaling.
viral infection may lead to testicular dysfunction and male Considering that spermatogonia and spermatocytes are
infertility. Major testicular cells, including somatic and located on both sides of the BTB, TLR3-initiated antiviral
germ cells, produce IFNs in response to viral infection. response in male germ cells may contribute to testicular
IFNs induce the expression of various antiviral proteins defense against viral infections from the circulating blood
in testicular cells, suggesting that the testis possesses its and the ascending genital tract. TLR11 is abundantly
own antiviral defense system.59 The PRR-initiated innate expressed in late-stage male germ cells, predominantly in
antiviral response in Leydig cells has been recently inves- round spermatids.62 TLR11 is recognized by Toxoplasma
tigated in mice. In addition to TLR3, which recognizes gondii and uropathogenic Escherichia coli (UPEC), thereby
dsRNA and induces an innate immune response,31 mouse initiating innate immune responses against these patho-
Leydig cells also express RIG-I and MDA5.52 The acti- gens in mice.63,64 However, functional TLR11 is absent in
vation of RIG-I and MDA5 with dsRNA in Leydig cells humans.65 Different TLR11 in mice and humans may be
triggers the IPS-1-dependent pathway to induce proin- a reason why T. gondii and UPEC cause pathogenesis in
flammatory cytokine and type 1 IFN production through humans but not in murine animals. T. gondii and UPEC
NF-κB and IRF3 activation. Moreover, major antiviral induce MCP-1, IL-12, IFN-γ, TNF-α, and IL-6 expression
proteins, including IFN-stimulating gene 15 (ISG15), in mouse germ cells, suggesting that male germ cells are
2’5’-oligodenylate synthetase1 (OAS1), and Mx GTPase1 involved in testicular defense against T. gondii and UPEC
(MX1), are upregulated in Leydig cells. ISG15, OAS1, and infections.62 Round spermatids also express MDA5, sug-
MX1 can amplify antiviral signaling, degrade viral RNA, gesting that late-stage male germ cells are also equipped
and block viral gene transcription, respectively, thereby with innate antiviral machinery.52 The innate immune
limiting viral replication within infected cells. Notably, responses in the late stages of male germ cells appear to
RIG-I and MDA5 signaling in Leydig cells inhibits tes- be particularly important for testicular defense against
tosterone synthesis, indicating that RNA-triggered innate microbial infections from the ascending genital tract
antiviral response perturbs testicular function.52 within the seminiferous tubules, as these compartments
DNA sensor signaling has also been demonstrated are separated from the immune components in the inter-
in Leydig cells. p204 and its signaling adaptor, STING, stitial spaces by the BTB.
are constitutively expressed in mouse Leydig cells53 and Although in vitro studies have demonstrated that cer-
p204 activation induces the expression of type 1 IFNs tain PRRs initiate innate immune responses in male
and antiviral proteins via a STING-dependent pathway. germ cells, the response grades are relatively low com-
p204 signaling does not significantly induce proinflam- pared with those in testicular somatic cells.61,62 However,
matory cytokine production in Leydig cells. Notably, the male germ cells can effectively resist microbial infec-
p204-initiated antiviral response in Leydig cells does not tions. Microorganisms rarely replicate in male germ cells,
affect testosterone synthesis. In agreement with these which suggests that these cells possess other mechanisms
findings, RNA viruses, such as MuV and HIV, usually to counteract microbial infections in addition to PRR-
induce orchitis and perturb male fertility, whereas DNA initiated innate immune responses. The abundant antimi-
viruses rarely cause orchitis.58 Based on these observa- crobial defensins in the genital tract may play a role in the
tions, we speculate that the induction of DNA sensor defense against microbial infections.66 The contribution
signaling is an ideal strategy for enhancing testicular of male germ cells to testicular defense against microbial
defense against viral infection and protecting testicular infections is worthy of further investigation.
function.
TLR signaling in testicular macrophages
PRR-initiated innate immune responses in male Testicular macrophages are major populations of immune
germ cells cells in the testicular interstitial spaces and represent
During puberty, more than 90% of testicular cells are germ approximately 20% of total interstitial cells. These macro-
cells and reside within the seminiferous tubules. Microbial phages are believed to constitute the frontline of testicular
pathogens from the blood circulation and the ascending innate defense against microbial infections.67 However,
genital tract can reach male germ cells localized to both testicular macrophages display immunosuppressive phe-
sides of the BTB. Male germ cells at certain stages produce notypes by predominantly producing anti-inflammatory
IFNs after stimulation with Sendai virus60 and spermato- cytokines.40 Interestingly, TLR signaling is inhibited by
gonia constitutively express IFN-α, IFN-γ, and antiviral UPEC infection in testicular macrophages.68 Treatment
proteins.54,55 These early observations suggest that male of testicular macrophages with UPEC does not induce
germ cells possess antiviral activities. Additionally, PRR- secretion of proinflammatory cytokines because of block-
initiated innate immune responses in male germ cells have age of the MyD88-dependent signaling pathway. In con-
been recently revealed. trast, UPEC predominantly induces the expression of
Concluding remarks  59

type 1 IFNs and chemokines through the activation of the virus sensor signaling neither induces proinflamma-
TRIF-dependent signaling pathway.68 Therefore, testicu- tory cytokine production nor inhibits testosterone syn-
lar macrophages display an antiviral response but prevent thesis in Leydig cells. 53 In contrast, RNA virus sensor
proinflammatory cytokine secretion.41 The mechanisms signaling significantly induces proinflammatory cyto-
underlying the subdued inflammatory response of tes- kine production and inhibits testosterone synthesis. 52
ticular macrophages have been recently investigated.69 We speculate that downregulation of testosterone and
In comparison with peritoneal macrophages, testicular upregulation of proinflammatory cytokines in Leydig
macrophages express relatively low levels of TLR pathway- cells in response to RNA viruses contributes to detri-
related  genes. TLR4 ligand does not activate NF-κB mental consequences in the testis.
because of a lack of IκBα degradation by ubiquitination in In addition to viruses, certain bacterial and parasitic
testicular macrophages. However, TLR3 and TLR4 ligands infections may also lead to testicular dysfunction. Sexually
indeed induce low levels of proinflammatory cytokines transmitted bacteria, most frequently Neisseria gonor-
independent of NF-κB activation. The levels of proinflam- rhoeae and Chlamydia trachomatis, usually cause orchi-
matory cytokines produced by testicular macrophages tis and epididymitis,74 impairing male fertility. Bacterial
are much lower than those produced by peritoneal mac- infections are generally susceptible to antibiotic treatment
rophages. Moreover, testicular macrophages produce high but may lead to infertility if not treated in time. T. gondii
levels of IL-10, an anti-inflammatory cytokine, in response can be transmitted to humans from cats and is associated
to TLR4 activation, confirming that testicular macro- with impairment of testicular function and male fertility,75
phages favor testicular immune privilege.69 Therefore, the although there is no evidence that T. gondii directly infects
role of testicular macrophages in the testicular defense the testis.
against microbial infections needs redefining. Autoimmune orchitis is chronic inflammation in the
testis characterized by the generation of autoantibodies
ORCHITIS against male germ cells and the impairment of spermato-
The mammalian testis possesses a unique immune envi- genesis. Autoimmune orchitis may lead to male steril-
ronment because of its immunoprivileged status and ity.37 Pathomechanisms underlying autoimmune orchitis
locally effective innate defense system.2 The maintenance have been investigated using EAO models.38
of immune homeostasis is essential for normal testicular
function. However, certain pathological conditions, such CONCLUDING REMARKS
as microbial infection, physical trauma, testicular tumors, The mammalian testis is a remarkable immunoprivi-
and even environmental toxicants, may disrupt testicu- leged site essential for protecting immunogenic germ
lar immune homeostasis and this may lead to orchitis, an cells from detrimental defense responses. However,
etiological factor in male infertility.70 Orchitis can be clas- the testis adopts a local innate defense system against
sified as infectious or autoimmune. microbial infection. Testicular immunoregulation is a
Infectious orchitis is caused by microbial infection. broad field worthy of further investigation, in which
In particular, a broad spectrum of viruses cause orchi- several aspects need prioritizing in future research, as
tis and usually impair male fertility. 58 The well-known follows. (1) Mechanisms underlying systemic immune
viral orchitis is caused by MuV. MuV orchitis in prepu- tolerance to male germ cells are largely unknown. In
berty is usually transient and fully recovers in about 2 this context, the role of TAM receptors in the regula-
weeks. However, MuV orchitis in adult men frequently tion of immune tolerance is worth in-depth investiga-
cause infertility.71 Viral orchitis can also be caused tion. (2) TLR2 and TLR4 cooperatively mediated EAO
by HIV infection, and HIV orchitis severely impairs induction. Endogenous TLR ligands in male germ cells
testicular function.72 Moreover, some other viruses, remain to be identified. (3) Although various PRRs ini-
including hepatitis C virus (HCV), coxsackievirus, tiate innate immune responses in testicular cells, the
influenza virus, and dengue virus, may induce orchi- roles of these PRRs in counteracting natural microbes
tis. 58 Although certain viruses, such as MuV, HIV, and in the testis have yet to be elucidated. (4) Male germ
HCV, can be detected in the testis of infected individ- cells display an active defense system against micro-
uals, viral titers are usually very low and it is unclear bial infections. However, PRR-initiated innate immune
whether these viruses directly perturb testicular func- responses are relatively weak in germ cells compared
tion. Vaccination with inactivated MuV may occasion- with somatic cells. Other mechanisms against micro-
ally lead to orchitis, suggesting that MuV orchitis can bial infection in male germ cells remain to be identified.
be indirectly caused by systemic immune-mediated (5) Systemic immune-response-mediated testicular dys-
mechanisms.73 The mechanisms by which viral infec- function is worth clarifying. Inflammatory cytokine
tion induces orchitis remains to be clarified. It should storm in response to microbial infection may result in
also be noted that RNA viral infection more frequently testicular dysfunction. Identification of key cytokines
induces orchitis than DNA viral infection, and the that impair testicular function may aid the develop-
underlying mechanism of this is worthy of further ment of therapeutic strategies for immunological tes-
investigation. Recent studies have shown that DNA ticular dysfunction.
60  Testicular immunoregulation

ACKNOWLEDGMENTS 18. Xiong W, Wang H, Wu H et al. Apoptotic spermato-


This work was supported by the Major State Basic Research genic cells can be energy sources for Sertoli cells.
Project of China (Nos. 2015CB943001, 2016YFA0101001) Reproduction. 2009;137(3):469–479.
and CAMS Initiative for Innovative Medicine (Nos. 19. Xiong W, Chen Y, Wang H et al. Gas6 and the Tyro
2017-IZM-B&R-06, 2017-IZM-3-007). 3 receptor tyrosine kinase subfamily regulate the
phagocytic function of Sertoli cells. Reproduction.
REFERENCES 2008;135(1):77–87.
1. Li N, Wang T, Han D. Structural, cellular and molec- 20. Nagata S, Hanayama R, Kawane K. Autoimmunity and
ular aspects of immune privilege in the testis. Front the clearance of dead cells. Cell. 2010;140(5):619–630.
Immunol. 2012;3:152. 21. Hanayama R, Tanaka M, Miyasaka K et al. Autoimmune
2. Zhao S, Zhu W, Xue S et al. Testicular defense sys- disease and impaired uptake of apoptotic cells in MFG-
tems: Immune privilege and innate immunity. Cell E8-deficient mice. Science. 2004;304(5674):1147–1150.
Mol Immunol. 2014;11(5):428–437. 22. Zhang X, Wang T, Deng T et al. Damaged sper-
3. Perez CV, Theas MS, Jacobo PV et al. Dual role of matogenic cells induce inflammatory gene expres-
immune cells in the testis: Protective or pathogenic sion in mouse Sertoli cells through the activation
for germ cells? Spermatogenesis. 2013;3(1):e23870. of Toll-like receptors 2 and 4. Mol Cell Endocrinol.
4. Chen Q, Deng T, Han D. Testicular immunoregu- 2013;365(2):162–173.
lation and spermatogenesis. Semin Cell Dev Biol. 23. Vabulas RM, Ahmad-Nejad P, da Costa C et al.
2016;59:157–165. Endocytosed HSP60s use toll-like receptor 2 (TLR2)
5. Robinson DR, Wu YM, Lin SF. The protein tyro- and TLR4 to activate the toll/interleukin-1 recep-
sine kinase family of the human genome. Oncogene. tor signaling pathway in innate immune cells. J Biol
2000;19(49):5548–5557. Chem. 2001;276(33):31332–31339.
6. Hafizi S, Dahlback B. Gas6 and protein S. Vitamin 24. Park JS, Svetkauskaite D, He Q et al. Involvement
K-dependent ligands for the Axl receptor tyrosine of toll-like receptors 2 and 4 in cellular activation
kinase subfamily. FEBS J. 2006;273(23):5231–5244. by high mobility group box 1 protein. J Biol Chem.
7. Lemke G. Biology of the TAM receptors. Cold Spring 2004;279(9):7370–7307.
Harb Perspect Biol. 2013;5(11):a009076. 25. Biggiogera M, Tanguay RM, Marin R et al.
8. Lu Q, Lemke G. Homeostatic regulation of the Localization of heat shock proteins in mouse male
immune system by receptor tyrosine kinases of the germ cells: An immunoelectron microscopical study.
Tyro 3 family. Science. 2001;293(5528):306–311. Exp Cell Res. 1996;229(1):77–85.
9. Lemke G, Rothlin CV. Immunobiology of the TAM 26. Zetterstrom CK, Strand ML, Soder O. The high
receptors. Nat Rev Immunol. 2008;8(5):327–336. mobility group box chromosomal protein 1 is
10. Takeda K, Akira S. Toll-like receptors. Current expressed in the human and rat testis where it may
Protocols in Immunology. 2015;109(1):14.12.1–14.12.10. function as an antibacterial factor. Hum Reprod.
11. Iwasaki A, Medzhitov R. Control of adaptive immu- 2006;21(11):2801–2809.
nity by the innate immune system. Nat Immunol. 27. Aslani F, Schuppe HC, Guazzone VA et al. Targeting
2015;16(4):343–353. high mobility group box protein 1 ameliorates tes-
12. Liew FY, Xu D, Brint EK et al. Negative regulation of ticular inflammation in experimental autoimmune
toll-like receptor-mediated immune responses. Nat orchitis. Hum Reprod. 2015;30(2):417–431.
Rev Immunol. 2005;5(6):446–458. 28. Riccioli A, Starace D, Galli R et al. Sertoli cells initi-
13. Rothlin CV, Ghosh S, Zuniga EI et al. TAM recep- ate testicular innate immune responses through TLR
tors are pleiotropic inhibitors of the innate immune activation. J Immunology. 2006;177(10):7122–7130.
response. Cell. 2007;131(6):1124–1136. 29. Wu H, Wang H, Xiong W et al. Expression patterns
14. Lu Q, Gore M, Zhang Q et al. Tyro-3 family recep- and functions of toll-like receptors in mouse sertoli
tors are essential regulators of mammalian sper- cells. Endocrinology. 2008;149(9):4402–4412.
matogenesis. Nature. 1999;398(6729):723–728. 30. Sun B, Qi N, Shang T et al. Sertoli cell-initiated
15. Wang H, Chen Y, Ge Y et al. Immunoexpression of testicular innate immune response through toll-
Tyro 3 family receptors—Tyro 3, Axl, and Mer—And like receptor-3 activation is negatively regulated
their ligand Gas6 in postnatal developing mouse tes- by Tyro3, Axl, and Mer receptors. Endocrinology.
tis. J Histoch Cytochem. 2005;53(11):1355–1364. 2010;151(6):2886–2897.
16. Chen Y, Wang H, Qi N et al. Functions of TAM 31. Shang T, Zhang X, Wang T et al. Toll-like receptor-
RTKs in regulating spermatogenesis and male fertil- initiated testicular innate immune responses in mouse
ity in mice. Reproduction. 2009;138(4):655–666. Leydig cells. Endocrinology. 2011;152(7):2827–2836.
17. Nakanishi Y, Shiratsuchi A. Phagocytic removal 32. Zhang Y, Li N, Chen Q et al. Breakdown of immune
of apoptotic spermatogenic cells by Sertoli cells: homeostasis in the testis of mice lacking Tyro3, Axl
Mechanisms and consequences. Biol Pharm Bull. and Mer receptor tyrosine kinases. Immunol Cell
2004;27(1):13–16. Biol. 2013;91(6):416–426.
References 61

33. Li MW, Xia W, Mruk DD et al. Tumor necrosis fac- 51. Starace D, Galli R, Paone A et al. Toll-like receptor
tor {alpha} reversibly disrupts the blood-testis barrier 3 activation induces antiviral immune responses in
and impairs Sertoli-germ cell adhesion in the semi- mouse Sertoli cells. Biol Reprod 2008;79(4):766–775.
niferous epithelium of adult rat testes. J Endocrinol. 52. Zhu W, Chen Q, Yan K et al. RIG-I-like receptors
2006;190(2):313–329. mediate innate antiviral response in mouse testis.
34. Theas MS, Rival C, Jarazo-Dietrich S et al. Tumour Mol Endocrinol. 2013;27(9):1455–1467.
necrosis factor-alpha released by testicular macro- 53. Zhu W, Liu P, Yu L et al. p204-initiated innate anti-
phages induces apoptosis of germ cells in autoim- viral response in mouse Leydig cells. Biol Reprod.
mune orchitis. Hum Reprod. 2008;23(8):1865–1872. 2014;91(1):8.
35. Artis D, Tschopp J. Innate immunity. Curr Opin 54. Melaine N, Lienard MO, Guillaume E et al.
Immunol. 2010;22(1):1–3. Production of the antiviral proteins 2’5’oligoad-
36. Li N, Liu Z, Zhang Y et al. Mice lacking Axl and Mer enylate synthetase, PKR and Mx in interstitial
tyrosine kinase receptors are susceptible to experi- cells and spermatogonia. J Reprod Immunol.
mental autoimmune orchitis induction. Immunol 2003;59(1):53–60.
Cell Biol. 2015;93(3):311–320. 55. Dejucq N, Lienard MO, Guillaume E et al.
37. Jacobo P, Guazzone VA, Theas MS et al. Testicular Expression of interferons-alpha and -gamma in tes-
autoimmunity. Autoimmun Rev. 2011;10(4):201–204. ticular interstitial tissue and spermatogonia of the
38. Naito M, Terayama H, Hirai S et al. Experimental rat. Endocrinology. 1998;139(7):3081–3087.
autoimmune orchitis as a model of immuno­ 56. Le Tortorec A, Denis H, Satie AP et al. Antiviral
logical male infertility. Med Mol Morphol. 2012;​ responses of human Leydig cells to mumps virus
45(4):185–189. infection or poly I:C stimulation. Hum Reprod.
39. Deng T, Chen Q, Han D. The roles of TAM receptor 2008;23(9):2095–2103.
tyrosine kinases in the mammalian testis and immu- 57. Bornstein SR, Rutkowski H, Vrezas I. Cytokines
noprivileged sites. Front Biosci. 2016;21:316–327. and steroidogenesis. Mol Cell Endocrinol.
40. Winnall WR, Muir JA, Hedger MP. Rat resident tes- 2004;215(1–2):135–141.
ticular macrophages have an alternatively activated 58. Dejucq N, Jegou B. Viruses in the mammalian male
phenotype and constitutively produce interleukin-10 genital tract and their effects on the reproductive
in vitro. J Leukoc Biol. 2011;90(1):133–143. system. Microbiol Mol Biol Rev. 2001;65(2):208–231.
41. Bhushan S, Hossain H, Lu Y et al. Uropathogenic 59. Dejucq N, Chousterman S, Jegou B. The testicular
E.  coli induce different immune response in tes- antiviral defense system: Localization, expression,
ticular and peritoneal macrophages: Implications and regulation of 2’5’ oligoadenylate synthetase,
for testicular immune privilege. PloS One. double-stranded RNA-activated protein kinase, and
2011;6(12):e28452. Mx proteins in the rat seminiferous tubule. J Cell
42. Janeway CA, Jr. Approaching the asymptote? Biology. 1997;139(4):865–873.
Evolution and revolution in immunology. Cold 60. Dejucq N, Dugast I, Ruffault A et al. Interferon-
Spring Harb Symp Quant Biol. 1989;54 Pt 1:1–13. alpha and -gamma expression in the rat testis.
43. Kumar H, Kawai T, Akira S. Pathogen recognition Endocrinology. 1995;136(11):4925–4931.
by the innate immune system. Int Rev Immunology. 61. Wang T, Zhang X, Chen Q et al. Toll-like receptor
2011;30(1):16–34. 3-initiated antiviral responses in mouse male germ
44. Beutler BA. TLRs and innate immunity. Blood. cells in vitro. Biol Reprod. 2012;86(4):106.
2009;113(7):1399–1407. 62. Chen Q, Zhu W, Liu Z et al. Toll-like receptor
45. Ireton RC, Gale M, Jr. RIG-I like receptors in antivi- 11-initiated innate immune response in male mouse
ral immunity and therapeutic applications. Viruses. germ cells. Biol Reprod. 2014;90(2):38.
2011;3(6):906–919. 63. Zhang D, Zhang G, Hayden MS et al. A toll-like
46. Wu J, Chen ZJ. Innate immune sensing and signal- receptor that prevents infection by uropathogenic
ing of cytosolic nucleic acids. Annu Rev Immunology. bacteria. Science. 2004;303(5663):1522–1526.
2014;32:461–488. 64. Yarovinsky F, Zhang D, Andersen JF et al. TLR11
47. Kawai T, Akira S. The role of pattern-recognition activation of dendritic cells by a protozoan profilin-
receptors in innate immunity: Update on Toll-like like protein. Science. 2005;308(5728):1626–1629.
receptors. Nat Immunol. 2010;11(5):373–384. 65. Lauw FN, Caffrey DR, Golenbock DT. Of mice and
48. Loo YM, Gale M, Jr. Immune signaling by RIG-I-like man: TLR11 (finally) finds profilin. Trends Immunol.
receptors. Immunity. 2011;34(5):680–692. 2005;26(10):509–511.
49. Paludan SR, Bowie AG. Immune sensing of DNA. 66. Com E, Bourgeon F, Evrard B et al. Expression of anti-
Immunity. 2013;38(5):870–880. microbial defensins in the male reproductive tract of
50. Springer TA. Traffic signals for lymphocyte recircu- rats, mice, and humans. Biol Reprod. 2003;68(1):95–104.
lation and leukocyte emigration: The multistep para- 67. Hedger MP. Macrophages and the immune responsive-
digm. Cell. 1994;76(2):301–314. ness of the testis. J Reprod Immunol. 2002;57(1–2):19–34.
62  Testicular immunoregulation

68. Bhushan S, Tchatalbachev S, Klug J et al. 72. Poretsky L, Can S, Zumoff B. Testicular dysfunction
Uropathogenic Escherichia coli block MyD88- in human immunodeficiency virus-infected men.
dependent and activate MyD88-independent sig- Metab Clin Exp. 1995;44(7):946–953.
naling pathways in rat testicular cells. J Immunol. 73. Clifford V, Wadsley J, Jenner B et al. Mumps vac-
2008;180(8):5537–5547. cine associated orchitis: Evidence supporting a
69. Bhushan S, Tchatalbachev S, Lu Y et al. Differential potential immune-mediated mechanism. Vaccine.
activation of inflammatory pathways in testicular 2010;28(14):2671–2673.
macrophages provides a rationale for their subdued 74. Trojian TH, Lishnak TS, Heiman D. Epididymitis
inflammatory capacity. J Immunol. 2015;194(11):​ and orchitis: An overview. Am Fam Physician.
5455–5464. 2009;79(7):583–587.
70. Schuppe HC, Meinhardt A, Allam JP et al. Chronic 75. Dalimi A, Abdoli A. Toxoplasma gondii and male
orchitis: A neglected cause of male infertility? reproduction impairment: A new aspect of toxo-
Andrologia. 2008;40(2):84–91. plasmosis research. Jundishapur J Microbiol.
71. Philip J, Selvan D, Desmond AD. Mumps orchitis 2013;6(8).
in the non-immune postpubertal male: A resurgent
threat to male fertility? BJU Int. 2006;97(1):138–141.
Inflammation and spermatogenesis
4
MARIA SUSANA THEAS, PATRICIA VERÓNICA JACOBO, CECILIA VALERIA PÉREZ,
VANESA ANABELLA GUAZZONE, and LIVIA LUSTIG

HUMAN ORCHITIS parenchyma is damaged, such as neoplasia, trauma, toxic


The testis is an immunologically privileged site with a agents, and cryptorchidia.10–13 Continuous release of sper-
unique immunosuppressor microenvironment able to tol- matic antigens from impaired ST induces disruption of
erate haploid germ cell antigens that appear at puberty and tolerance and induction of an autoimmune inflamma-
to easily accept tissue grafts. The existence of the blood- tory response.14 Orchitis may also have a genetic basis as a
testis barrier (BTB), the secretion of immunosuppressor manifestation of polyglandular autoimmune syndrome. In
mediators by somatic cells (mainly Sertoli cells), and the particular, this rare autosomal recessive disease is caused
immunomodulatory function of regulatory T (Treg) cells by mutation of the AIRE gene or is related to a defect in
contribute to immunoprivilege. Treg cell function.15,16
However, this immunosuppressed microenvironment
does not prevent the testis from developing inflammatory ANTIBODIES TO SPERMATIC ANTIGENS
and immune reactions in response to different stressors Although cellular immune response is the main patho-
such as pathogens or tissue damage. genic mechanism in autoimmune orchitis, the presence
Infection and inflammation are widely accepted as of antisperm antibodies (ASAs) and immune complex
important etiological factors of subfertility or infertility. deposits at the seminiferous tubular walls also contribute
Prevalence rates up to 15% have been reported in patients to testicular inflammation.17
examined in infertility clinics.1 The most frequent orchitis ASAs may be detected in serum, sperm, seminal plasma,
or epididymo-orchitis induced by specific pathogens occur and also in follicular fluid or cervicovaginal secretions in
by retrograde ascent of different urethral bacterial patho- women. Patients with a clinical history of testis inflam-
gens (Escherichia coli, Chlamydia trachomatis, and Neisseria mation associated with damage frequently present high
gonorrhea, among others) or systemic viral infections. The ASAs levels in spermatozoa and seminal plasma. As well,
best-known orchitogenic viruses in humans are mumps 40%–70% of vasectomized men and patients with con-
virus and human immunodeficiency virus (HIV). Mumps genital absence of the vas deferens18 develop antibodies to
virus induces innate immune responses in mice through sperm antigens. ASAs IgG type present in seminal plasma
retinoic acid-inducible gene signaling and TLR2 expressed interacts with sperm plasma membrane antigens, affect-
by Sertoli and Leydig cells, which results in the produc- ing sperm motility or inducing sperm agglutination,19
tion of proinflammatory cytokines and chemokines.2 while ASAs IgA type detected in the sperm head interferes
Both viruses are able to replicate in Leydig cells in vitro, mainly with the fertilization process. The detection of
inhibiting testosterone.3,4 Acquired immune deficiency autoantibodies against protein disulfide isomerase ER60,
syndrome patients often suffer from oligozoospermia or expressed in the developing acrosome, has been proposed
azoospermia and hypogonadism.5 Immunosuppression as a tool for diagnosis of infertility associated to testicular
is also a potential mechanism for HIV persistence in the inflammation.20
testis.6
Orchitis is characterized by impairment of seminiferous EXPERIMENTAL MODELS OF AUTOIMMUNE ORCHITIS
tubules (STs) presenting germ cell sloughing and degener- Experimental models of orchitis contributed significantly
ation associated with interstitial and peritubular immune to understanding of testis immunoregulation and inter-
infiltrates of T cells and macrophages that may eventually actions among immune, somatic, and germ cells. Acute
invade STs. Granulomas may also occur in severe orchitis. orchitis can be induced by injection of lipopolysaccha-
Increase of Th1 and Th17 proinflammatory cytokines have ride21 or human chorionic gonadotrophin.22 However,
been demonstrated in human orchitis associated with the experimental model of chronic autoimmune orchi-
oligozoospermia and fibrosis contributing to transient tis (EAO) better mimics human orchitis associated with
or permanent subfertility or infertility.1,7,8 Mast cells and infertility. The classical model of EAO has been induced
peritubular cells are involved in testicular fibrosis mainly in guinea pig, mice, and rats using as immunogen a tes-
through increased production of profibrotic cytokines.9 ticular homogenate obtained from adult animals in adju-
Lymphomonocyte infiltration is also frequent in vants.23–25 Sperm cells without adjuvants have also been
pathologies other than infection in which the testicular used to induce EAO.26 With the utilization of a proteomic

63
64  Inflammation and spermatogenesis

approach with sera of rats and mice with EAO or by inject- PATHOGENESIS OF AUTOIMMUNE ORCHITIS
ing recombinant proteins, several immune-dominant We will analyze the main features of the immune
autoantigens have been identified in orchitis. Zonadhesin, response induced in rats immunized with testicular
expressed on the outer acrosomal membrane of sperma- homogenate in Freund’s complete adjuvant and Bordetella
tids and sperm and ODF2 present in the outer dense fibers pertussis as coadjuvant.25 Focal orchitis develops 50 days
of sperm flagellum are testis-specific, and other nonorgan- after the first immunization and severe orchitis after 80
specific antigens such as heat shock protein 70 or RNA days. Focal orchitis is characterized by multiple foci of
binding protein H1 have also been detected.27–29 STs with different degrees of germ cell sloughing asso-
Other models of chronic EAO can be induced by ciated with discrete peritubular immune cell infiltrates,
manipulation of the immune system, such as thymectomy mainly of dendritic cells (DCs), macrophages, and lym-
in 3-day-old mice, which induces autoimmune diseases phocytes. In severe orchitis, aspermatogenesis occurs
in several organs including the testis30 or after infection in most of the STs in which only spermatogonia, a few
of the testis with viruses such as Sendai virus (closely basal spermatocytes, and Sertoli cells are present (Figure
related to mumps virus) or myxoma virus.31,32 Unilateral 4.1). Abundant immune cell infiltrates are present in the
infection with bacteria (Listeria monocytogenes)21 induces interstitium, but unlike EAO in mice or human orchitis,
autoimmune orchitis in the contralateral testis.33 In this the inflammatory cells are not detected inside damaged
last model, orchitis was also passively transferred to unin- STs. Most are atrophic and the number of apoptotic post-
fected recipients with CD4+ T cells from infected donors.34 meiotic germ cells increases. Granulomas are frequently

(a1) 100 µm (a2) 50 µm (a3) 20 µm

(b1) 100 µm (b2) 50 µm (b3) 20 µm

*
*

(c1) 100 µm (c2) 50 µm (c3) 20 µm

Figure 4.1  Microphotographs of testis sections. (a) Normal rat showing seminiferous tubules (ST) with normal spermatogen-
esis and no signs of interstitial inflammation. (b) Rat immunized with testis homogenate and adjuvants showing a focus of ST with
germ cell loss or aspermatogenesis (*) intermingled with normal ST and mild interstitial immune cell infiltrates (focal orchitis).
(c) Rat immunized with testis homogenate and adjuvants showing severe orchitis. The whole section presents aspermatogenic ST (*)
associated with interstitial and perivascular cell infiltrates of macrophages and lymphocytes. Hematoxylin eosine
Pathogenesis of autoimmune orchitis  65

observed. Proinflammatory mediators, secreted mainly (Figure 4.2). An increase in BTB permeability together
by immune cells, increase in the testis of EAO rats, gener- with a reduction in expression of the tight junction mol-
ating an inflammatory microenvironment responsible for ecule occludin and the gap junction molecule connexin-43
impairment of testicular function. were detected in the testis of EAO rats. Reduced adhe-
sion ability of germ cells and the increased expression of
Blood-testis barrier adherent junction proteins, N-cadherin and α-catenin,
BTB is a key player in the maintenance of testicular in Sertoli cells are also associated with germ cell slough-
immunoprivilege. Its structure, which involves specialized ing.35,36 Several inflammatory mediators and cytokines,
cell junctions between adjacent Sertoli cells close to the base- increased during testicular inflammation, are described as
ment membrane, prevents entry of leukocytes and antibodies modulators of junction dynamics and BTB function. IL-6
into seminiferous tubules and limits interaction among germ disrupts the Sertoli cell tight junction barrier in vitro via
cell autoantigens and the immune system. During testicular the p38 MAPK signaling pathway and by inhibiting deg-
inflammation, BTB structure and function are impaired radation of BTB constitutive proteins.37 In vivo, IL-6 was

N-cadherin
Connexin 43

Claudin 11

Occludin
α-catenin
β-catenin

p120
ZO-1
Actin

IL17R

IL6R

TNFR1
(a) IL-17
IL-6
TNF-α
NO

(b)

Figure 4.2  Schematic drawing illustrating BTB under normal (a) and inflammatory conditions (b). (a) BTB consists of coexisting
integral membrane proteins of the adherens junctions (cadherins), gap junctions (connexins), and tight junctions (claudins, occludin)
between adjacent Sertoli cells. Each integral membrane protein is associated with its corresponding adaptors (ZOs, catenins, and
p120) that link them to the actin cytoskeleton. (b) Proinflammatory cytokines (IL-6, TNF-α, IL-17) and nitric oxide (NO) contribute to
BTB impairment. Decreased occludin and connexin-43 expression, delocalization of claudin-11, and upregulation of N-cadherin and
α-catenin are also shown.
66  Inflammation and spermatogenesis

shown to induce a reduction in occludin expression after with EAO a significant increase in the number of CD4+
local administration to adult rat testes.38 Tumor necrosis and CD8+ cells, including pathogenic Th1, Th17 subsets,
factor alpha (TNF-α) and transforming growth factor beta and Foxp3+ T cells, occurs. 51 Th1- and Th17-type immune
(TGF-β) can alter testicular tight junction function by response was characterized by secretion of TNF-α and
accelerating endocytosis of integral membrane proteins.38 interferon gamma (IFN-γ) (Th1 cytokines) and IL-17
IL-1α induces BTB disruption but exerts its effect on the and IL-23 (Th17 cytokines) were observed in the testis
Sertoli cell actin cytoskeleton network without affecting throughout the two phases of EAO. The number of CD4+
levels of junction proteins.39 IL-17 disrupts the Sertoli cell T cells producing TNF-α, IFN-γ, or IL-17 peaked dur-
barrier both in vitro and in vivo, decreasing the expres- ing the focal phase of EAO; however, these cells declined
sion of occludin.40 As well, nitric oxide (NO) has been during the severe phase of the disease. CD8+ T cells were
shown to regulate testicular adherens and tight junction also found to express Th1 and Th17 cytokines along the
dynamics.41,42 course of the disease, with these cells predominating
in the severe phase of EAO. Testicular IL-17 and IL-23
Immune cells content increased in EAO with maximum levels in the
DCs are specialized sentinel cells that bridge the innate severe phase, suggesting their contribution to mainte-
and adaptive immune system. DCs are present in the tes- nance of chronic testicular inflammation.52 The highest
ticular interstitium and their numbers strongly correlate number of CD4+ T cells producing TNF-α and IFN-γ
with development of tissue damage in the inflamed tes- at EAO onset in rats is consistent with the major role
tes.43 DCs from normal and orchitic testis express simi- attributed to these cells in the development of murine
lar intensity levels of major histocompatibility complex EAO. 53–56
class II and costimulatory molecules (CD80 and CD86). CD4+ Foxp3+ Treg cells also accumulated in the testis
However, a significantly higher mRNA expression of IL-12 with EAO, reaching a peak at the focal phase, then dras-
and chemokine receptor CCR7 was detected in isolated tically declining during the severe phase of the disease.
testicular DCs from rats with EAO compared to normal In contrast, the number of CD8+Foxp3+ Treg cells that
rats.44 While IL-12 supports differentiation of the Th1 sub- increased in the focal phase remained stable during the
set of helper T cells, the upregulation of CCR7 is required severe phase of EAO.51 Along EAO development, testicular-
for entry and homing of DCs into lymph nodes (LNs) in draining LNs, but not non-draining LNs, are strategically
order to present antigens to antigen-specific T cells. In fact, enriched for antigen-specific CD4+CD25+Foxp3+ Treg
during the development of orchitis, testicular-draining cells. We reported that CD4+ Treg cells expressing high
LNs express the CCR7 ligand, CCL19, and present a sig- levels of CD25 derived from testicular-draining LNs of
nificant increase in the percentage of DCs compared to both normal and EAO rats suppress T cell proliferation
testicular-nondraining LNs. Functional analyses show in vitro, exhibit an antigen-experienced status, and also
that DCs isolated from testis and testicular-draining LN express TGF-β, a suppressor of effector T cells. However,
from rats undergoing orchitis significantly enhanced pro- Treg cells derived from testicular-draining LNs of rats
liferation of naïve T cells. This supports the notion that with EAO exhibit a stronger antigen-specific prolifera-
DCs from chronically inflamed testis are immunogenic, tive response and suppressive capacity compared to cells
present antigens to T cells, and stimulate an autoimmune derived from normal rats.57 Our results showed that Treg
response against testicular antigens, thus causing impair- cells with the potential to inhibit autoimmune response
ment of spermatogenesis and immunological infertil- against germ cells are present in the testis and in testis-
ity 44,45 (Figure 4.3). draining LNs. However, these cells fail to effectively con-
Macrophages also play the role of antigen-presenting trol testicular inflammation and tissue destruction since
cells and release inflammatory cytokines and chemokines multiple mechanisms may disturb local immune regula-
that contribute to EAO development. In fact, in vivo deple- tion: (1) inadequate ratio effector/Treg cells (2) intrinsic
tion of these cells with clodronate-containing liposomes functional defects in Treg cell populations, (3) patho­
in rats undergoing EAO decreased incidence and severity genic  Th17 cells resistant to Treg cell regulation, and
of testicular damage. Testicular macrophage population (4)  impairment of Treg cell function by inflammatory
is heterogeneous; it includes resident macrophages (ED2+ milieu. We infer that these last two mechanisms are cen-
cells), inflammatory monocytes recently arrived from cir- tral to understanding why inflammation progresses in the
culation (ED1+ cells), and double-positive (ED1+ ED2+) testis despite the presence of increased numbers of func-
subpopulation, the major contributor to the macrophage tional Treg cells.
number increase in EAO.46 Inflammatory chemokines Testosterone levels were found to be decreased in serum
CCL2, CCL3, and CCL4 regulate macrophage recruit- of rats in the early phase of EAO.58 Fijak et al.59 reported
ment within the interstitium via CCR2, CCR1, and CCR5 that testosterone supplementation effectively reduced dis-
receptors.47–49 ease development in rats with EAO exerting a systemic
The existence of scarce CD4+ and CD8+ T lympho- effect by inhibiting proinflammatory Th1-specific cyto-
cytes and natural killer cells in normal testis of human kine production in testicular-draining LNs and by induc-
and rodents has been established. 34,50,51 In testis of rats ing suppressive Treg cells in the testis.
Pathogenesis of autoimmune orchitis  67

Immunization Testicular
Normal testis
site draining lymph node draining lymph node

Treg
ST
Th17
L

CD8

Th1
DC

Chemokines
IFN-γ
TNF-α
Ag TGF-β
Mφ IL-6
IL-12
IL-17
Orchitic testis

Figure 4.3  Schematic drawing illustrating some features of experimental autoimmune orchitis induction. During the immuni-
zation period, the autoimmune response initiates in the immunization site draining lymph nodes (LN) where dendritic cells (DCs)
and macrophages capture and present the antigen (Ag). The presentation of self-Ag induces activation of specific autoreactive T
cells. Subsequently, DC-sensitized lymphocytes migrate by chemokines to the testis where they contribute to testicular inflamma-
tion thereby causing tissue damage. Spermatic antigens released to the interstitium during the course of the disease are processed
by inflammatory testicular DCs that migrate to testicular-draining LN and activate T lymphocytes. Under inflammatory conditions
with increased levels of cytokines, immature testicular DC overcome immune tolerance (privilege). This view is supported by the
finding that testicular DC in EAO showed increased CCR7 expression, whereas testicular-draining LN express the CCR7 ligand CCL19,
a chemokine involved in DC traffic. Progressive amplification of the autoimmune response finally leading to chronification of EAO
may result from continuous antigen presentation to lymphocyte by DCs in testicular-draining LN and in the testis. L: Leydig cell. ST:
seminiferous tubules ϕ: macrophages.

Germ cell apoptosis mediators compared to normal testis. Specifically, inflammatory


Spermatogonial stem cells reside on the basement mem- macrophages upregulate expression of inducible (iNOS)
brane of STs in intimate contact with Sertoli cells and and constitutive NOS isoforms, whereas resident mac-
with factors generated in the interstitium. In this complex rophages, less sensitive to inflammatory microenviron-
microenvironment (niche) stem cells are able to self-renew ment stimuli, upregulate only iNOS.61 NO released from
and differentiate into committed progenitors that sustain EAO macrophages activates external and mitochondrial
spermatogenesis. apoptotic pathways and also contributes to support high
Under inflammatory conditions spermatocytes and intratesticular testosterone content.62 Administration
spermatids are the main target of immunological attack; of Nω-Nitro-L-arginine methyl ester hydrochloride
these cells die by apoptosis through external and mito- (L-NAME), a competitive inhibitor of NOS, to rats
chondrial pathways, whereas basal germ cells are pro- undergoing EAO decreased incidence and severity of the
tected from death by overexpressing Bcl-260 (Figure 4.4). disease by preventing testicular NO production and pro-
Immune cells play an active role in promoting germ tecting germ cells from apoptosis.61
cell death. In EAO, an increased number of macrophages Testicular macrophages from rats with orchitis
and a rise in nitric oxide synthase (NOS) activity and release a high amount of TNF-α and in vitro neutral-
expression generate a threefold increase in NO content ization of this cytokine with etanercept (Embrel®)
68  Inflammation and spermatogenesis

Sertoli
Apoptosis cell

↓ Bcl-2
Cytochrome c
Postmeiotic
↑ Bax
germ cells
Caspase 9
activation
Sertoli NO
cell Caspase 8 Caspase 3
activation activation

NO
Apoptosis
NO

NO
NO
NO

SURVIVAL Premeiotic
Bcl-2 germ cells
Bcl-2
Bcl-2

IL-6 IL-6R Myoid cells


TNF-α TNFR1
Fas L Fas R
Nitric oxide

Figure 4.4  Schematic drawing illustrating molecules and pathways involved in germ cell apoptosis in orchitis. In EAO, spermato-
cytes and spermatids upregulate death receptors TNFR1 and Fas. TNF-α produced by interstitial macrophages activates caspase 8,
which in turn activates executioner apoptotic caspase 3. FasL shed from interstitial lymphocytes or expressed by neighboring germ
cells trigger FasR activation in a paracrine or autocrine pathway, inducing apoptotic death. NO released by interstitial macrophages
activates both external pathway (caspase 8 activation) and mitochondrial pathway, promoting release of cytochrome c from mito-
chondria and caspase 9 activation followed by caspase 3 execution of apoptosis. Release of cytochrome c from mitochondria is
facilitated by increased Bax/Bcl-2 ratio in the mitochondria. Cytokines such as IL-6 and IL-17 contribute to germ cell death. Whereas
postmeiotic germ cells present in the adluminal compartment die by apoptosis, basal premeiotic germ cells are protected by upreg-
ulation of the antiapoptotic Bcl-2 protein.

reverts the effect of TNF-α on germ cell apoptosis. The associated with germ cell sloughing. 35 Similarly, in vivo
fact that 40% of apoptotic germ cells express TNFR1 administration of IL-17 induces recruitment of inflam-
in EAO testis suggests the contribution of other pro- matory cells into the testicular interstitium, impairment
apoptotic factors in the induction of germ cell death.63 of BTB function, and indirectly, germ cell apoptosis.40
During the severe phase of EAO the number of IL-6R+ Membrane-bound Fas L (mFasL), like other members
germ cells and IL-6 production by inflammatory mac- of the TNF family of death ligands, can be processed by
rophages increases. This cytokine is able to induce germ proteolytic shedding to an active soluble form (sFasL).68
cell death in vitro.64 This apoptotic effect may be the Testicular macrophages do not express membrane or intra-
result of direct activation of apoptotic pathways, or cellular FasL; however, testicular CD4+ and CD8+ T cells
more likely, IL-6 may generate a microenvironment expressing mFasL may be the source of the increased sFasL
permissive to the effect of proapoptotic factors such as present in the extracellular milieu in severe orchitis.69 FasL
TNF-α or IFN-γ.65–67 IL-6 injected intratesticularly into Strep (a Strep-Tag molecule fused to three monomers of
normal adult rats induces focal testicular inflammation the extracellular domain of FasL) injected in rats can enter
References 69

Table 4.1  Factors used to reduce inflammation in experimental autoimmune orchitis.


Agent Action
Deoxyspergualin Inhibits lymphocyte IFN gamma production71
Mn Ab to IFN-γ Antiviral and immune-modulatory capacity72
Testosterone Stimulates differentiation of Treg cells73
Melanocyte stimulating hormone, TGF- β Anti-inflammatory action74
Secretory leukocyte Immunosuppressor75
protease inhibitor (SLPI)
Ethyl piruvate Blocks high mobility group box protein 1’s inflammatory action76
Intermedin Antioxidant capacity and proinflammatory cytokine production77
L-NAME Competitive inhibitor of NOS62
Galectin-1 Anti-inflammatory action78

seminiferous tubules. The fact that this molecule induces REFERENCES


germ cell Fas activation in vitro, triggering apoptosis, 1. Schuppe HC, Meinhardt A, Allam JP et al. Chronic
highlights sFasL as a key molecule involved in germ cell orchitis: A neglected cause of male infertility?
death. In EAO, apoptotic spermatocytes and spermatids Andrologia. 2008;40:84–91.
express Fas or FasL or coexpress both molecules, indicat- 2. Wu H, Shi L, Wang Q et al. Mumps virus-induced
ing involvement of the Fas-FasL system in germ cell death innate immune responses in mouse Sertoli and
in an autocrine and/or paracrine way.70 Leydig cells. Sci Rep. 2016;6:19507.
3. Mouchel T, Le Goffic R, Patard JJ et al. Le virus
TREATMENT OF ORCHITIS ourlien et l´orchite: Vers une approche physio-
Symptomatic treatment, bed rest, and antibiotics for pathologique. Progr Urol. 2002;12:124–128.
specific bacterial infections and eventually IFN-α2B in 4. Le Goffic R, Mouchel T, Aubry F et al. Production
mumps orchitis are used to reduce testicular pain and of the chemokines monocyte chemotactic protein-1,
inflammation in patients with acute orchitis. regulated on activation of normal T cell expressed
No specific treatment exists for chronic orchitis. and secreted protein, growth-related onco-gene,
Experimentally, several drugs have been used to reduce and interferon-γ-inducible protein-10 is induced by
testicular inflammation (Table 4.1). Corticoids have been the Sendai virus in human and rat testicular cells.
used in the past in infertile patients with high levels of Endocrinology. 2002;143:1434–1440.
ASAs. However, at present, intracytoplasmic sperm injec- 5. Dejucq N, Jégou B. Viruses in the mammalian male
tion represents the chosen treatment for infertility of these genital tract and their effects on the reproductive
patients. system. Microbiol Mol Biol Rev. 2001;65:208–231.
6. Winnall WR, Lloyd SB, De Rose R et al. Simian
PERSPECTIVES immunodeficiency virus infection and immune
Infertility affects 16%–20% of couples in reproductive responses in the pig-tailed macaque testis. J Leukoc
age,13 and inflammation of the male reproductive tract is Biol. 2015;97:599–609.
considered an important etiological factor. Diagnosis of 7. Suominen J, Söderström KO. Lymphocyte infil-
human autoimmune orchitis associated with subfertility tration in human testicular biopsies. Int J Androl.
or infertility, a chronic inflammatory pathology, is cur- 1982;5:461–466.
rently possible only by histopathological evaluation of tes- 8. Duan YG, Yu CF, Novak N et al. Immunodeviation
tis biopsy, an invasive procedure. Although detection of towards a Th17 immune response associated with
ASAs as well as immune complexes is helpful, there is a testicular damage in azoospermic men. Int J Androl.
need to find reliable peripheral markers of testis inflam- 2011;e536–545.
mation. Deeper understanding of mechanisms by which 9. Frungieri MB, Weidinger S, Meineke V et al.
pathogenic immune cells and Tregs interact in testis Proliferative action of mast-cell tryptase is
under inflammation as well as identification of mediators mediated by PAR2, COX2, prostaglandins, and
involved in germ cell damage and spermatogenic arrest PPAR gamma. Possible relevance to human
will facilitate exploration of specific therapies to reduce fibrotic disorders. Proc Natl Acad Sci U S A.
inflammation and reinitiate spermatogenesis. In patholo- 2002;99:15072–15077.
gies like orchitis and spermatogenic arrest, regeneration of 10. Frungieri MB, Calandra RS, Lustig L et al. Number,
spermatogenesis seems to be possible since spermatogonia distribution pattern, and identification of macro-
remain within the STs despite the loss of postmeiotic germ phages in the testes of infertile men. Fertil Steril.
cells.79,80 2002;78:298–306.
70  Inflammation and spermatogenesis

11. Schuppe HC, Meinhardt A. Immune privilege and 26. Itoh M, Hiramine C, Hojo K. A new murine model
inflammation of the testis. Chem Immunol Allergy. of autoimmune orchitis induced by immunization
2005;88:1–14. with viable syngeneic testicular germ cells alone.
12. Nistal M, Riestra ML, Paniagua R. Focal orchi- I  Immunological and histological studies. Clin Exp
tis in undescended testes. Arch Pathol Lab Med. Immunol. 1991;83:137–142.
2002;126:64–69. 27. Wheeler KM, Tardif S, Rival C et al. Regulatory
13. Jahnukainen K, Jorgensen N, Pöllänen P et al. T cells control tolerogenic versus autoimmune
Incidence of testicular mononuclear cell infiltrates response to sperm in vasectomy. Proc Natl Acad Sci
in normal human males and in patients with germ U S A. 2011;108:7511–7516.
cell neoplasia. Int J Androl 1995;18:313–320. 28. Fijak M, Iosub R, Schneider E et al. Identification of
14. Fraczek M, Kurpisz M. Mechanisms of the immunodominant autoantigens in rat autoimmune
harmful effects of bacterial semen infection on orchitis. J Pathol. 2005;207:127–138.
ejaculated human spermatozoa: Potential inflam- 29. Terayama H, Hirai S, Naito M et al. Specific autoan-
matory markers in semen. Folia Histochem Cytobiol. tigens identified by sera obtained from mice that are
2015;53:201–217. immunized with testicular germ cells alone. Sci Rep.
15. Kisand K, Peterson P. Autoimmune polyendocri- 2016;6:35599.
nopathy candidiasis ectodermal dystrophy. J Clin 30. Sakaguchi S, Sakaguchi N, Asano M et al.
Immunol. 2015;35:463–478. Immunologic self-tolerance maintained by activated
16. Chan AY, Anderson MS. Central tolerance to self T cells expressing IL-2 receptor alpha-chains (CD25).
revealed by the autoimmune regulator. Ann NY Acad Breakdown of a single mechanism of self-tolerance
Sci. 2015;1356:80–89. causes various autoimmune diseases. J Immunol.
17. Restrepo B, Cardona Maya W. Anticuerpos anti- 1995;155:1151–1164.
espermatozoides y su asociación con la fertilidad. 31. Melaine N, Ruffault A, Dejucq-Rainsford N et
Actas Urol Esp. 2013;37:571–578. al. Experimental inoculation of the adult rat tes-
18. D’Cruz OJ, Haas GG Jr, de La Rocha R et al. tis with Sendai virus: Effect on testicular mor-
Occurrence of serum antisperm antibodies in patients phology and leukocyte population. Hum Reprod.
with cystic fibrosis. Fertil Steril. 1991;56:519–527. 2003;18:1574–1579.
19. Cui D, Gangwei H, Yonggang S et al. Antisperm 32. Fountain S, Holland MK, Hinds LA et al. Interstitial
antibodies in infertile men and their effect on semen orchitis with impaired steroidogenesis and sper-
parameters: A systematic review and meta-analysis. matogenesis in the testes of rabbits infected with an
Clinica Chimica Acta. 2015;444:29–36. attenuated strain of myxoma virus. J Reprod Fertil.
20. Fijak M, Schneider E, Klug J et al. Autoantibodies 1997;110:161–169.
against protein disulfide Isomerase ER-60 are a diag- 33. Mukasa A, Hiromatsu K, Matsuzaki G et al. Bacterial
nostic marker for low-grade testicular inflammation. infection of the testis leading to autoaggressive
Hum Reprod. 2014;29:2382–2392. immunity triggers apparently opposed responses of
21. Reddy MM, Mahipal SV, Subhashini J et al. Bacterial αβ and γδ T cells. J Immunol. 1995;155:2047–2056.
lipopolysaccharide-induced oxidative stress in the 34. Mukasa A, Yoshida H, Kobayashi N et al. γδ T cells in
impairment of steroidogenesis and spermatogenesis infection-induced andautoimmune-induced testicu-
in rats. Reprod Toxicol. 2006;22:493–500. lar inflammation. Immunology. 1998;95:395–401.
22. Assmus M, Svechnikov K, von Euler M et al. Single 35. Pérez CV, Sobarzo CM, Jacobo PV et al. Loss of
subcutaneous administration of chorionic gonado- occludin expression and impairment of blood-testis
tropin to rats induces a rapid and transient increase barrier permeability in rats with autoimmune orchi-
in testicular expression of pro-inflammatory cyto- tis: Effect of interleukin 6 on Sertoli cell tight junc-
kines. Pediatr Res. 2005;57:896–901. tions. Biol Reprod. 2012;87:1–12.
23. Freund J, Lipton MM, Thompson GE. 36. Pérez C, Sobarzo C, Jacobo P et al. Impaired expres-
Aspermatogenesis in guinea pig induced by tes- sion and distribution of adherens and gap junction
ticular tissue and adjuvant. J Exp Med. 1953;97:​ proteins in the seminiferous tubules of rats undergoing
711–726. autoimmune orchitis. Int J Androl. 2011;34:e566–577.
24. Kohno S, Munoz JA, Williams TM et al. 37. Zhang H, Yin Y, Wang G et al. Interleukin-6 dis-
Immunopathology of murine experimental allergic rupts blood-testis barrier through inhibiting pro-
orchitis. J Immunol. 1983;130:2675–2682. tein degradation or activating phosphorylated ERK
25. Doncel GF, Di Paola JA, Lustig L. Sequential study in Sertoli cells. Sci Rep. 2014;4:4260.
of the histopathology and cellular and humoral 38. Li MWM, Mruk DD, Lee WM et al. Cytokines and
immune response during the development of junction restructuring events during spermatogen-
autoimmune orchitis in Wistar rats. Am J Reprod esis in the testis: An emerging concept of regulation.
Immunol. 1989;20:44–51. Cytokine Growth Factor Rev. 2009;20:329–338.
References 71

39. Sarkar O, Mathur PP, Cheng CY et al. Interleukin 1 52. Jacobo P, Pérez CV, Theas MS et al. CD4+ and
alpha (IL1A) is a novel regulator of the blood-testis CD8+ T cells producing Th1 and Th17 cytokines are
barrier in the rat. Biol Reprod. 2008;78:445–454. involved in the pathogenesis of autoimmune orchitis.
40. Pérez CV, Pellizzari EH, Cigorraga SB et al. Reproduction. 2011;141:249–258.
IL17A impairs blood-testis barrier integrity and 53. Mahi-Brown CA, Tung KS. Activation require-
induces testicular inflammation. Cell Tissue Res. ments of donor T cells and host T cell recruitment
2014;358:885–898. in adoptive transfer of murine experimental auto-
41. Lee NP, Cheng CY. Nitric oxide/nitric oxide syn- immune orchitis (EAO). Cell Immunol. 1989;124:​
thase, spermatogenesis, and tight junction dynam- 368–379.
ics. Biol Reprod. 2004;70:267–276. 54. Yule TD, Tung KS. Experimental autoimmune
42. Lee NP, Mruk DD, Wong CH et al. Regulation of orchitis induced by testis and sperm antigen-
Sertoli-germ cell adherens junction dynamics in the specific T cell clones: An important pathogenic
testis via the nitric oxide synthase (NOS)/cGMP/ cytokine is tumor necrosis factor. Endocrinology.
protein kinase G (PRKG)/beta-catenin (CATNB) 1993;133:​1098–1107.
signaling pathway: An in vitro and in vivo study. Biol 55. Yule TD, Mahi-Brown CA, Tung KS. Role of testicu-
Reprod. 2005;73:458–471. lar autoantigens and influence of lymphokines in
43. Rival C, Lustig L, Radu I et al. Identification of a testicular autoimmune disease. J Reprod Immunol.
dendritic cell population in normal testis and in 1990;18:89–103.
chronically inflamed testis of rats with autoimmune 56. Terayama H, Naito M, Qu N et al. Intra-testicular
orchitis. Cell Tissue Res. 2006;324:311–318. expression of mRNAs of both interferon γ and tumor
44. Rival C, Guazzone VA, von Wulffen W et al. necrosis factor α is significantly increased in experi-
Expression of co-stimulatory molecules, chemokine mental autoimmune orchitis in mice. J Reprod Dev.
receptors and proinflammatory cytokines in den- 2011;57:296–302.
dritic cells from normal and chronically inflamed rat 57. Jacobo P, Guazzone VA, Pérez CV et al. CD4+
testis. Mol Hum Reprod. 2007;13:853–861. Foxp3+ regulatory T cells in autoimmune orchitis:
45. Guazzone VA, Hollwegs S, Mardirosian M Phenotypic and functional characterization. Am J
et  al. Characterization of dendritic cells in tes- Reprod Immunol. 2015;73:109–125.
ticular draining lymph nodes in a rat model of 58. Suescun MO, Calandra RS, Lustig L. Alterations of
experimental autoimmune orchitis. Int J Androl. testicular function after induced autoimmune orchi-
2011;34:276–289. tis in rats. J Androl. 1994;15:442–448.
46. Rival C, Theas MS, Suescun MO et al. Functional 59. Fijak M, Schneider E, Klug J et al. Testosterone
and phenotypic characteristics of testicular mac- replacement effectively inhibits the development of
rophages in experimental autoimmune orchitis. experimental autoimmune orchitis in rats: Evidence
J Pathol. 2008;215:108–117. for a direct role of testosterone on regulatory T cell
47. Guazzone VA, Rival C, Denduchis B et al. Monocyte expansion. J Immunol. 2011;186:5162–5172.
chemoattractant protein-1 (MCP-1/CCL2) in experi- 60. Theas MS, Rival C, Jarazo-Dietrich S et al. TNF-α
mental autoimmune orchitis. J Reprod Immunol. released by testicular macrophages induces apoptosis
2003;60:143–157. of germ cells in autoimmune orchitis. Hum Reprod.
48. Guazzone VA, Jacobo P, Theas MS et al. Cytokines 2008;23:1865–1872.
and chemokines in testicular inflammation: A brief 61. Jarazo-Dietrich S, Jacobo P, Pérez CV et al. Up regu-
review. Microsc Res Tech. 2009;72:620–628. lation of nitric oxide synthase-nitric oxide system in
49. Guazzone VA, Jacobo P, Denduchis B et al. the testis of rats undergoing autoimmune orchitis.
Expression of cell adhesion molecules, chemokines Immunobiology. 2012;217:778–787.
and chemokine receptors involved in leukocyte 62. Jarazo Dietrich S, Fass MI, Jacobo PV et al. Inhibition
traffic in rats undergoing autoimmune orchitis. of NOS-NO system prevents autoimmune orchitis
Reproduction. 2012;143:651–662. development in rats: Relevance of NO released by
50. Tompkins AB, Hutchinson P, de Kretser DM et testicular macrophages in germ cell apoptosis and
al. Characterization of lymphocytes in the adult testosterone secretion. PLoS One. 2015;10:e0128709.
rat testis by flow cytometry: Effects of activin and 63. Suescun MO, Rival C, Theas MS et al. Involvement
transforming growth factor beta on lymphocyte of tumor necrosis factor-alpha in the pathogen-
subsets in vitro. Biol Reprod. 1998;58:943–951. esis of autoimmune orchitis in rats. Biol Reprod.
51. Jacobo P, Guazzone VA, Jarazo-Dietrich S et al. 2003;68:2114–2121.
Differential changes in CD4+ and CD8+ effector and 64. Rival C, Theas MS, Guazzone VA et al. Interleukin-6
regulatory T lymphocyte subsets in the testis of rats and IL-6 receptor cell expression in testis of rats
undergoing autoimmune orchitis. J Reprod Immunol. with autoimmune orchitis. J Reprod Immunol.
2009;81:44–54. 2006;70:43–58.
72  Inflammation and spermatogenesis

65. Minami R, Muta K, Ilseung C et al. Interleukin-6 73. Fijak M, Damm LJ, Wenzel JP et al. Influence of
sensitizes multiple myeloma cell lines for apop- testosterone on inflammatory response in tes-
tosis induced by interferon-alpha. Exp Hematol. ticular cells and expression of transcription fac-
2000;28:244–255. tor Foxp3 in T cells. Am J Reprod Immunol.
66. Oritani K, Tomiyama Y, Kincade PW et al. Both 2015;74:12–25.
Stat3-activation and Stat3-independent bcl-2 down- 74. Ito T, Ito N, Betterman A et al. Collapse and resto-
regulation are important for interleukin-6-induced ration of MHC class I dependent immune privilege.
apoptosis of 1A9-M cells. Blood. 1999;93:​1346–1354. Am J Pathol. 2004;164:623–644.
67. Boer U, Fennekohl A, Puschel GP. Sensitization by 75. Guazzone VA, Guerrieri D, Jacobo P et al.
interleukin-6 of rat hepatocytes to tumor necro- Microencapsulated secretory leukocyte protease
sis factor alpha-induced apoptosis. J Hepatol. inhibitor decreases cell-mediated immune response
2003;38:728–735. in autoimmune orchitis. Life Sci. 2011;89:100–106.
68. Kayagaki N, Kawasaki A, Ebata T et al. 76. Aslani F, Schuppe HC, Guazzone VA et al. Targeting
Metalloproteinase-mediated release of human Fas high mobility group box protein 1 ameliorates tes-
Ligand. J Exp Med. 1995;182:1777–1783. ticular inflammation in experimental autoimmune
69. Jacobo PV, Fass M, Pérez C et al. MS. Involvement orchitis. Hum Reprod. 2015;30:417–431.
of soluble Fas ligand in germ cell apoptosis in tes- 77. Li L, Ma P, Liu Y et al. Intermedin attenuates LPS-
tis of rats undergoing autoimmune. Cytokine. induced inflammation in the rat testis. PLoS One.
2012;60:385–392. 2013 8:e65278.
70. Theas S, Rival C, Lustig L. Germ cell apoptosis in 78. Pérez CV, Gómez LG, Gualdoni GS et al. Dual
autoimmune orchitis: Involvement of the Fas-FasL roles  of endogenous and exogenous galectin-1 in
system. Am J Reprod Immunol. 2003;50:166–176. the control of testicular immunopathology. Sci Rep.
71. Ablake M, Itoh M, Kaneko T et al. Short-term 2015;5:12259.
prophylaxis with deoxyspergualin prevents tes- 79. Hentrich A, Wolter M, Szardening-Kirchner C et al.
ticular autoimmunity in mice. Eur J Pharmacol. Reduced numbers of Sertoli, germ, and spermato-
2002;450:209–212. gonial stem cells in impaired spermatogenesis. Mod
72. Itoh M, Yano A, Xie Q et al. Essential pathogenic role Pathol. 2011;24:1380–1389.
for endogenous interferon-gamma (IFN-γ) during 80. Sousa M, Cremades N, Alves C et al. Developmental
disease onset phase of murine experimental autoim- potential of human spermatogenic cell co-cultured
mune orchitis. I. In vivo studies. Clin Exp Immunol. with Sertoli cells. Hum Reprod. 2002;17:161–172.
1998;111:513–520.
Junctional adhesion molecule (JAM)
family
5
Recent findings and their role and regulation
in spermatogenesis
KUN HUANG and WING-YEE LUI

INTRODUCTION and designate them as JAM-A, JAM-B, and JAM-C.11 All


Extensive and timely restructuring of cell junctions takes JAM family proteins are characterized by a V-type and a
place at the interface between adjacent Sertoli cells and C2-type immunoglobulin-like domain in their extracel-
between Sertoli cells and germ cells during spermatogen- lular domain followed by a transmembrane segment and
esis. Restructuring of junctions at the blood-testis barrier a cytoplasmic tail with a PDZ-binding motif (Phe-Leu-
(BTB) allows the transit of germ cells across the BTB at Val) (Figure 5.1). The three classical JAMs share up to 32%
stage VIII so the germ cells enter the apical compartment of amino acid sequence identity12 and also share ~15%
for further development. After entering the apical com- sequence homology with the nonclassical JAM members.
partment, developing germ cells remain attached to the The nonclassical JAMs are diverged from the classical JAM
seminiferous epithelium (Sertoli cells) for physical support by their cytoplasmic tails.13 The cytoplasmic tails of the
and at the same time have to keep migrating toward the classical JAMs are relatively conserved in length and motif
tubular lumen for further differentiation. Therefore, cell composition.14 PDZ-containing proteins such as ZO-1,
junctions formed at the BTB and between Sertoli germ cell AF-6, CASK, PAR-3, and MUPP-1 bind onto the PDZ
interface undergo dynamic restructuring. Such restruc- motif of JAM-A15–18 while ZO-1 and PAR-3 bind to JAM-B
turing can be achieved by the timely and spatial changes and JAM-C.19 ZO-1, MUPP-1, and LNX2 bind to CAR.20–22
of structural proteins such as occludin, claudin, and junc- Interaction of PDZ motif with various PDZ-containing
tional adhesion molecules (JAMs), peripheral proteins proteins is involved in intracellular signal transduction
such as zonula occludens and catenin, as well as signal- and an array of cellular functions including cell polar-
ing molecules including JNK and p38 kinase. JAM is the ity, cell migration, and barrier permeability (Table 5.1). In
family of Ca2+-independent cell adhesion molecules found the extracellular domain, a tripeptide motif (Arg-Val-Glu
at both the BTB and Sertoli germ cell interface. In this for JAM-A, Arg-Leu-Glu for JAM-B, and Arg-Ile-Glu for
review, we discuss the role of classical JAMs and coxsacki- JAM-C) in the V-type domain acts as a dimerization motif
evirus and adenovirus receptor (CAR) in the testis as well and another tripeptide linker sequence (Val-Leu-Val) con-
as their regulatory mechanisms. We highlight some recent nects the two immunoglobulin domains to impose a bent
findings of classical JAMs and CAR in other tissue models conformation (Figure 5.1).23–25
and hope that this information can serve as the blueprint
for planning new studies to delineate the unknown func- Homophilic and heterophilic trans-interaction
tions in the testis and the regulation of JAMs and CAR Crystal structures have shown that two JAM-A molecules
during spermatogenesis. on the same plasma membrane form cis-dimer via the
dimerization motif in the V-type Ig domain.23 Based on
GENERAL PROPERTIES OF JAM FAMILY MEMBERS
the bent conformation and the positioning of the dimer-
Molecular structure of classical JAMs and CAR ization motif, Kostrewa et al. have proposed that cis-dimer
Proteins of the JAM family are type I glycoprotein belong- appears like a U shape on the cell surface and forms a two-
ing to the immunoglobulin superfamily (IgSF). The JAM dimensional network by trans-interaction of their extra-
family consists of seven members including three classical cellular domain with cis-dimer on apposing membrane.23
members (JAM-A, JAM-B, and JAM-C) and four nonclas- Having the conserved dimerization motif between the
sical members (CAR, endothelial cell-selective adhesion classical JAMs, JAM-B and JAM-C might dimerize in a
molecule [ESAM], JAM-L, and JAM-4).1–10 Like other similar manner as JAM-A for the establishment of adhe-
junction protein families, the nomenclature of JAM pro- sive interaction.23 These motifs may also be involved in het-
teins is diverse because JAMs have been identified by sev- erophilic cis-dimer between the classical JAM members.26
eral research groups. For instance, VE-JAM and human Trans-homophilic interactions of JAM-A have been con-
JAM-2 are the aliases of JAM-B. Efforts were made in 2003 firmed using recombinant soluble JAM-A (rsJAM-A) and
to standardize the nomenclature of the classical JAMs anti-JAM-A Fab BV11 (a monoclonal antibody targeting

73
74  Junctional adhesion molecule (JAM) family

cis/trans-Interaction
trans-hetero-dimer e.g. JAM-A/αLβ2
cis-dimer trans-homo-dimer e.g. JAM-B/-C JAM-A/αVβ1
Cytosol
PAR-3
ZO-1

αLb2
JAM-A (LFA-1)
JAM-C JAM-a
β α
Extracellular region
α β
Dimerization JAM-A
motif V-type domain JAM-B
αVb1
Tripeptide linker
C-type domain

PAR-3 PAR-3
Type II ZO-1 ZO-1
PDZ binding motif AF-6
Monomer of CASK
Classical JAMs MUPP-1
(JAM-A, -B, -C)
and CAR

Figure 5.1  Molecular structures of JAMs and their modes of interactions. Classical JAMs and CARs share structural homology
with two extracellular Ig-like domains, a transmembrane domain and a cytoplasmic tail. Two JAMs on the same plasma membrane
form cis-dimers followed by the formation of trans-interaction between cis-dimer on the plasma membrane of an apposing cell,
resulting in the formation of trans-homo-dimer or trans-heterodimer. JAMs also interact with integrin.

Table 5.1  Classical JAMs and CAR.


Extracellular Intracellular Tissue
Member partner partner Function distribution Knockout mouse phenotype
JAM-A αVβ3 ZO-1, AF-6 Reovirus attachment Blood, bone marrow, Colonic injury46
αLβ2 PAR-3 Barrier permeability brain, heart, intestine, Enhanced thrombotic function
CASK Cell polarity kidney, liver, lung, of platelets75
MUPP-1 Leukocyte transmigration pancreas, skin, Hepatocyte apoptosis and
Angiogenesis spleen, testis attenuation of neutrophil
infiltration76
Shape alterations in corneal
epithelial47
Male subfertility36,61
JAM-B JAM-C ZO-1, PAR-3 Barrier permeability Heart, kidney, lymph Normal mouse development62
α4β1 Leukocyte migration nodes, intestine,
testis
JAM-C JAM-B ZO-1, PAR-3 Cell-cell adhesion Brain, bone marrow, Growth retardation and
CAR Cell polarity heart, lung, liver, pulmonary dysfunction77
αMβ2 Leukocyte migration kidney, spleen, testis, Severe hydrocephalus78
αXβ2 Barrier permeability nerve Male-specific infertility37
αVβ3 Angiogenesis
Nerve conduction
Spermatogenesis
CAR JAM-C ZO-1, LNX2, Coxsackievirus and Brain, kidney, liver, Dilated intestine and atrophy of
JAM-L MUPP-1, MAGI-1 adenovirus entry lung, intestine, skin, the exocrine pancreas and
Leukocyte migration testis, pancreas, atrioventricular block79
Barrier permeability cornea
Spermatogenesis
General properties of JAM family members  75

specifically to rsJAM-A dimers but not monmers).27 It was cells. Immunofluorescence staining has shown that JAM-C
found that anti-JAM-A Fab BV11 inhibits the binding of is widely distributed on the germ cell surface and a portion
fluid-phase rsJAM-A to immobilized rsJAM-A.27 Trans- of JAM-C protein is concentrated in the anterior area of
homophilic interactions of JAM-C have also been confirmed round spermatogenic cells.37 As spermiogenesis proceeds,
by a similar approach. A dose-dependent binding of Fc JAM-C is confined to the head region of the elongated
fusion protein of rsJAM-C to immobilized rs-JAM-C was spermatids at the junctional plaques and colocalized with
reported.25 Surface plasmon resonsance analysis has fur- JAM-B expressed in Sertoli cells.37 Heterophilic interaction
ther confirmed the specificity of the homophilic interac- of JAM-B/JAM-C between Sertoli cells and germ cells at
tion of JAM-C.25 the apical ES provides a structural interlock for the attach-
Apart from homophilic interaction, classical JAMs can ment of elongated spermatids on the seminiferous epithe-
form trans-heterophilic interaction with the JAM family. lium prior to spermiation.37 CAR is present in Sertoli cells
Among the classical JAMs, JAM-B/JAM-C heterophilic and all types of germ cells at different developmental stages
interactions have been reported,6 heterophilic interactions from spermatogonia to spermatozoa.32,38 CAR is localized
of JAM-A with classical JAMs have not yet been detected. at the BTB and the apical ES.32,38 Preferential expression of
JAM-B/JAM-C heterophilic interaction is achieved JAM-A, JAM-B, JAM-C, and CAR in different cell types
through the dimerization motifs in the V domain, while and compartments of the seminiferous epithelium con-
the C domain is believed to be important for stabilization tribute to the heterogeneity and specific properties of the
of the interaction.28 Dynamic light scattering (DLS) analy- intercellular junction complexes.
sis and analytical ultracentrifugation (AUC) have revealed
that JAM-B/JAM-C heterodimer is a preferential inter- Functions of classical JAMs and CAR
action compared with JAM-C homodimer formation.28 JAM family proteins not only function as key molecules
This heterophilic interaction enables specific properties of for leukocyte transmigration, but also are involved in vari-
intercellular junction complexes that play important roles ous cellular functions and physiological processes such as
in leukocyte adhesion and migration.28 barrier function, cell adhesion, cell polarity, and pathogen-
Trans-heterophilic interactions are not restricted to the esis (Table 5.1). For leukocyte transmigration, readers are
classical JAMs; the classical JAMs interact heterophilically strongly encouraged to read Weber et al.,26 Wang et al.,39
with nonclassical JAMs and various integrins (Table 5.1). and Bazzoni40 for a more comprehensive view of the topic.
For instance, JAM-A can interact with β2 of LFA-1 (αLβ2) In this chapter, we focus our discussion on the findings
and αVβ3.29,30 JAM-B can interact with integrin α4β131 of classical JAMs and CAR on barrier function, cell adhe-
while JAM-C can interact with many heterophilic inter- sion, and cell polarity in other epithelia, and highlight the
acting partners such as CAR and various integrins includ- recent advances in the studies of classical JAMs and CAR
ing αVβ3, αΜβ1, and αXβ2.28,32–34 A broad spectrum of in the testis.
trans-interaction of JAMs within their family members
and with other transmembrane proteins is a notable and Barrier function
unique feature that supports JAMs to exert a wide range of Studies in various epithelial cells and endothelial cells
biological functions ranging from cell adhesion, polarity, have confirmed that classical JAMs and CAR are the
and leukocyte migration. key molecules that regulate barrier permeability. Several
lines of evidence in epithelial and endothelial cell models
Tissue distribution and localization in the testis have confirmed that JAMs are found in the region of the
Classical JAMs and CAR are present in many tissues tight junction (TJ) when JAMs are transfected. However,
including brain, heart, lung, skin, and testis (Table 5.1). transfected JAMs are localized at distinct subcompart-
Classical JAMs are expressed in epithelial cells, endothe- ments of the TJs.41 Exogenous JAM-A in Madin-Darby
lial cells, and leukocytes, while CAR is expressed mainly canine kidney (MDCK) cells has been found to colo-
in epithelial cells, endothelial cells, and cardiomyocytes. calize partially with ZO-1 at the TJs, while JAM-B is
As well as being involved in leukocyte transmigration, found to be diffuse at the lateral membrane. Transfected
the JAM family is also involved in an array of cellular JAM-C is found to colocalize perfectly at the TJs.41
functions such as cell adhesion and polarity. Both classical Distinct subcompartmental localization of JAMs at the
JAMs and CAR can be found in the testis and are crucial for site of TJs reflects the heterogeneity of JAM members in
spermatogenesis. JAM-A is highly expressed in Sertoli cells nature. Furthermore, the differential expression of JAMs
and can be found at the BTB.35 Immunohistochemistry in different organs are crucial for the establishment of
staining has indicated that JAM-A expression is stage- ­t issue-specific TJs/AJs. For instance, the BTB in the testis
dependent. JAM-A expression reaches its highest at stages constituted by TJs and basal ES is classified as one of the
IX–XIV and its lowest at stages IV–VI.35 Germ cells express tightest BTBs where JAM-A and JAM-B are localized.37
a moderate level of JAM-A and are localized on the cell sur- In contrast to the BTB, high endothelial venules (HEVs),
face of spermatozoa.36 Unlike JAM-A, JAM-B is only pres- which support high levels of lymphocyte extravasation
ent in Sertoli cells.37 It is mainly localized at the BTB and from the blood, express high levels of JAM-B and JAM-C
the apical ectoplasmic specialization (ES) between Sertoli but not JAM-A.41 In the seminiferous epithelium, the
cells and germ cells. JAM-C is solely expressed by germ apical ES does not coexist with TJs, where only JAM-B
76  Junctional adhesion molecule (JAM) family

and JAM-C are present to form heterophilic interaction the site of cell-cell contact but not to tight junctions, which
between Sertoli cells and the elongated spermatids.37 suggests that CAR may function as an adherens junction
Several in vitro studies have demonstrated that JAM-A protein.55
homodimers are critical for epithelial barrier function.42–44 In the testis, JAM-B/JAM-C heterophilic interaction is a
For instance, antibodies against JAM-A have shown to crucial adhesive structure between Sertoli cells and elon-
inhibit the recovery of the barrier function after TJ dis- gated spermatids at the apical ES of the seminiferous epi-
ruption in T84 and SK-CO15 epithelial cells.42,43 siRNA thelium prior to spermiation.37 JAM-B/JAM-C interlocks
knockdown of JAM-A increases the barrier permeability not only function as junction complexes, but also act as a
as shown by reduced transepithelial resistance (TER) and platform for actin bundling and are involved in the assem-
increased FITC-dextran flux and also alters the epithelial bly of cell polarity complexes that are crucial for spermatid
cell morphology via Rap1-mediated regulation of β2 integ- differentiation.37
rin.44 Antibody blockade of JAM-A not only affects epithe-
lial TJ barrier function, but disrupts corneal endothelial Cell polarity
barrier function and results in corneal swelling.45 In addi- Polarity is a fundamental property of all eukaryotic cells
tion, mice having JAM-A knockout display an increase in and is crucial to many aspects such as cell differentia-
intestinal mucosal and corneal epithelial permeability.46–48 tion, cell migration, and morphogenesis.56 The establish-
Collectively, both in vitro and in vivo data strongly sug- ment of apicobasal polarity and proper formation of tight
gest a central role of JAM-A in the regulation of the barrier junctions requires the interaction of cell polarity proteins
function. In contrast, JAM-C seems to exert the opposite and junction proteins.56 Par3/Par6/aPKC complex is a
effect on barrier permeability. Inhibition of JAM-C causes cell polarity complex in which Par3 and Par6 are linked
a significant reduction in endothelial permeability in vitro through aPKC. Studies have shown that Par3 directly
and in vivo.49 Although classical JAMs share similar struc- associates with JAM-A.16,57 JAM-A is colocalized with
ture, they differentially regulate barrier permeability. ZO-1 and E-cadherin to form the spotlike adherens junc-
CAR is found to colocalize with TJ proteins such as tion at the initial cell-cell contact during junction forma-
occludin and ZO-1 in various tissues such as kidney and tion. Par3 and aPKC are found after formation of spotlike
brain as well as cultured epithelial cells.50 In vitro studies adherens junctions.19,58,59 The association between JAM-A
have revealed that CAR mediates homotypic interaction.20 and Par3 provides a way for anchoring the Par3/Par6/
Overexpression of CAR in airway epithelial cells increases aPKC complex at TJs, resulting in the establishment of a
TER, while soluble CAR or anti-CAR antibody cause TJ fully polarized epithelial cell.57 Subsequent studies have
disruption in airway epithelial cells and MDCK cells.20,51 further confirmed that JAM-B and JAM-C, but not CAR,
In addition, yeast two-hybrid analyses have revealed that strongly associate with Par3, thus suggesting that classical
cytoplasmic domain of CAR directly interacts with vari- JAMs are involved in the establishment of cell polarity.19
ous TJ-associated molecules such as MUPP-1, MAGI-1, However, in the testis, JAM-C localization in sperma-
and TAPP1.21,52 These data further support the idea that tids shows a little overlap with Par337 By coimmunopre-
CAR is a crucial player in barrier function. cipitation, JAM-C is found to associate with Par6, Cdc42,
In the testis, JAM-A, JAM-B, and CAR are found at PKCλ, and PATJ,37 suggesting that JAM-C can interact
the site of the BTB.35,37 Knockdown of CAR by siRNA with two polarity protein complexes, CRB3/Pals1/PATJ
perturbs the Sertoli cell TJ barrier function as evidenced and Par3/Par6/aPKC. Subsequent studies have confirmed
by the decrease in TER, while overexpression of CAR in that JAM-C forms a stable complex with Pals1 and Par6 at
Sertoli cells exerts the opposite effect.53 More importantly, the apical ES to confer adhesion and polarity.60 The inter-
CAR not only functions as a TJ structural protein but also action of Src kinase with Pals1 and Par6 can destabilize
acts as a regulatory protein in the seminiferous epithe- JAM-C/Pals1/Par6 complex and results in detachment of
lium. The siRNA knockdown of CAR induces endocytosis spermatids from the seminiferous epithelium.60
of occludin via modulation of the phosphorylation status
of occludin, resulting in TJ disruption.53 Knockout phenotypes in male reproductive system
Through loss-of-function studies, the physiological roles
Cell adhesion of JAMs have been identified (Table 5.1). A wide range
Studies have clearly demonstrated that heterophilic inter- of physiological processes such as spermatogenesis and
actions among JAM members or between JAMs and leukocyte transmigration requires temporal and spatial
integrins contribute to cell adhesion. JAM-B and JAM-C expression as well as the participation of JAMs. JAMs are
undergo heterophilic interaction in cell-cell contacts and also involved in various diseases including viral infection
this interaction is important to facilitate the adhesion of and cancer development. This review focuses on the role
T natural killer and dendritic cells to the endothelium.54 of JAMs in male reproductive functions, and therefore we
JAM-B also modulates JAM-C/αMβ2 integrin-mediated will put more emphasis on knockout mice displaying male
adhesion of cultured leukocytes to the lymph node section reproductive defects.
by controlling the accessibility of JAM-C to αMβ2 inte- Among classical JAMs and CAR models, knockout of
grin.28 Apart from classical JAMs, studies performed in JAM-A and JAM-C exhibit notable abnormality in sper-
nonpolarized cells have revealed that CAR is localized to matogenesis. Detailed investigation of localization of
Future perspectives and concluding remarks  77

JAM-A has found that JAM-A is present in spermatids and motifs, which leads to an additive effect on Sp1- and NRSF-
mature spermatozoa and epididymal cauda sperms.36 Male mediated JAM-B transactivation, while TGF-β2 inhibits
JAM-A knockout mice produce progeny with altered sex JAM-B transcription by the activation of Smad proteins.68
ratio coupled with reduced litter size.61 They also exhibit Interferon-gamma (IFNγ) and tumor necrosis factor alpha
sperm motility defects such as a significant reduction of (TNF-α) are two major cytokines that are elevated dur-
the percentage of motile spermatozoa and progressive and ing testicular inflammation and cause reduced fertility.
hyperactivated motility.36 Sperms from JAM-A knockout Studies in our laboratory have revealed that IFNγ+TNF-α
mice also display abnormal morphology in the mitochon- disrupt testicular cell adhesion by exerting a synergistic
drial sheaths and in the flagella.36 effect on CAR downregulation.69 IFNγ+TNF-α treatment
JAM plays a role in the recruitment of cell polarity com- inhibits the binding of the basal transcription factors and
plex for the establishment of polarity.16,57 Loss of JAM-C promotes the binding of negative regulators including
hinders the recruitment of Par6/Cdc42/PKCλ complex to NFkB subunits and Sp1 to the CAR promoter region.69
the anterior region of spermatid head for the polarization IFNγ+TNF-α also upregulates CAR protein degradation
of round spermatids, resulting in male infertility.37 Testis via ubiquitin-proteasome and NFkB pathways.69 In endo-
from JAM-C knockout mouse is significantly reduced in thelial cells, IFNγ+TNF-α and basic fibroblast growth fac-
size and has no elongated spermatids in the seminiferous tor (bFGF) can induce the redistribution of JAM-A from
tubules.37 It is known that JAM-B is the heterophilic inter- the TJs toward the luminal surface of the endothelium.30,70
acting partner of JAM-C at the apical ES of the seminif- Taken collectively, hormones and cytokines are key regu-
erous epithelium. However, JAM-B-deficient mice show lators to modulate the expression and availability of JAMs
normal reproductive function without any detectable and CAR in both epithelial and endothelial cells.
sperm abnormality.62 Still, we could not rule out the func- MicroRNAs (miRNAs) are small noncoding RNAs
tional role of JAM-B in spermatogenesis since the fertil- that control gene expression at both transcriptional and
ity tests were observed using mice at age 8 weeks old62; translational levels and have been identified to regulate
it remains unknown if abnormalities of spermatogenesis the expression of JAMs. For instance, in breast cancer,
might appear in aging JAM-B-null mice as was reported miR-145 is downregulated and miR-495 is upregulated
in occludin knockout.63 It is also possible that other JAMs and they both target JAM-A.71,72 Overexpression of miR-
may complement one another in some ways due to func- 145 downregulates JAM-A and causes the restructuring
tional redundancy. of the actin cytoskeleton, resulting in decreased motility
and invasiveness,71 while inhibition of JAM-A by miR-495
Regulation of JAMs and CAR in the testis promotes breast cancer cell migration.72 Efforts should be
Deciphering the regulation of JAMs and CAR definitely made to examine if JAMs and CAR in the testis can be
helps gain a better understanding of spermatogenesis and regulated by miRNA.
pathogenesis. The following section reviews some of the Proteolytic cleavage of JAMs has been reported in endo-
recent studies that are pertinent to the regulation of JAMs thelial models. The process itself as well as the cleaved
and CAR. fragments are crucial for the control of the JAM func-
Both in vitro and in vivo studies have shown that JAMs tion. For instance, a significant increase in disintegrin-/
and CAR can be regulated by hormones such as angioten- ADAM17-mediated JAM-A cleavage has been reported in
sin and cytokines such as transforming growth factor-βs inflamed vascular endothelium.73 Soluble JAM-A has been
(TGF-βs). For instance, angiotensin II upregulates the shown to reduce the transendothelial migration of neutro-
expression of JAM-A and contributes to the progression phils in vitro and decreases the infiltration of neutrophil
of hypertension.64 The JAM-A-associated hypertension in vivo.73 Soluble JAM-C is found to be elevated in the
can be attenuated by the use of blockers targeting ANGII serum of patients with rheumatoid arthritis and is capa-
signaling.64 Knockdown of TGF-β receptors and canoni- ble of inducing angiogenesis.74 Like JAM-A, the cleavage
cal Smad signaling causes the upregulation of the JAM-A of JAM-C is mediated partially by disintegrin, ADAM10,
level and inhibits cell invasion in MDA-MB-231 cells.65 and ADAM17.74 Up to now, no studies have been done to
Studies have shown that TGF-β1 significantly inhibits examine if proteolytic cleavage of JAMs occurs in the tes-
JAM-A gene transcription via the activation of Smads tis, let alone its regulatory effects on spermatogeneis.
and promotes JAM-A protein degradation via the activa-
tion of p54 JNK signaling.65 In the testis, TGF-β3 regu- FUTURE PERSPECTIVES AND CONCLUDING REMARKS
lates JAM-B expression to facilitate the disassembly of the There is no doubt that the JAM family is a group of impor-
BTB and the apical ES.66 TGF-β3 regulates cell junction tant cell adhesion proteins involved in the process of germ
restructuring via ERK1/2- and p54 JNK-mediated JAM-B cell development and differentiation. Some studies have
mRNA destabilization and Smad-dependent JAM-B pro- been performed to investigate the regulation of JAMs
tein degradation.66 In addition, TGF-β3 increases in the and CAR during spermatogenesis, such as the regulatory
kinetics of JAM-A and occludin endocytosis and blockade mechanisms involved in the timely expression of JAMs
of TβR1 can abolish TGF-β3-induced JAM-A endocyto- and CAR in normal or pathological conditions. Further
sis.67 IL-1α promotes JAM-B expression by facilitating the studies are warranted to address the many questions that
binding of positive transcription factors to the cis-acting remain unknown and unaddressed. For example, how
78  Junctional adhesion molecule (JAM) family

does the JAM protein complex interact and regulate the 10. Nasdala I. A transmembrane tight junction protein
dynamics of junction restructuring during spermatogen- selectively expressed on endothelial cells and plate-
esis? Are sJAMs or sCAR present in the testis, and if so, lets. J Biol Chem. 2002;277:16294–16303.
what are their functions? What is the mechanism and 11. Muller WA. Leukocyte-endothelial-cell interactions
signaling involved in cleavage of JAMs? What regulatory in leukocyte transmigration and the inflammatory
mechanisms are involved in recycling and degradation of response. Trends Immunol. 2003;24:327–334.
JAMs and CAR in the testis? Are miRNAs present and 12. Aurrand-Lions MA, Duncan L, Du Pasquier L,
involved in the regulation of JAMs and CAR in the testis? Imhof BA. Cloning of JAM-2 and JAM-3: An emerg-
The answers may provide important information on how ing junctional adhesion molecular family? Curr Top
junction restructuring between adjacent Sertoli cells and/ Microbiol Immunol. 2000;251:91–98.
or between Sertoli cells and germ cells via the JAM family 13. Bazzoni G. The JAM family of junctional adhesion
proteins occurs. molecules. Curr Opin Cell Biol. 2003;15:525–530.
14. Bradfield PF, Nourshargh S, Aurrand-Lions M,
ACKNOWLEDGMENT Imhof BA. JAM family and related proteins in leu-
The work was funded by Hong Kong Research Grants kocyte migration (Vestweber series). Arterioscler
Council (HKU774213) and HKU/CRCG Seed Funding for Thromb Vasc Biol. 2007;27:2104–2112.
Basic Research. 15. Ebnet K, Schulz CU, Meyer Zu Brickwedde MK,
Pendl  GG, Vestweber D. Junctional a­ dhesion mol-
REFERENCES ecule  interacts with the PDZ domain-­ containing
1. Martin-Padura I. Junctional adhesion molecule, a proteins  AF-6 and ZO-1. J Biol Chem. 2000;275:​
novel member of the immunoglobulin superfam- 27979–27988.
ily that distributes at intercellular junctions and 16. Itoh M, Sasaki H, Furuse M et al. Junctional adhesion
modulates monocyte transmigration. J Cell Biol. molecule (JAM) binds to PAR-3: A possible mecha-
1998;142:117–127. nism for the recruitment of PAR-3 to tight junctions.
2. Williams LA, Martin-Padura I, Dejana E, Hogg N, J Cell Biol. 2001;154:491–498.
Simmons DL. Identification and characterisation of 17. Martínez-Estrada OM, Villa A, Breviario F et al.
human junctional adhesion molecule (JAM). Mol Association of junctional adhesion molecule with
Immunol. 1999;36:1175–1188. calcium/calmodulin-dependent serine protein kinase
3. Palmeri D, van Zante A, Huang CC, Hemmerich (CASK/LIN-2) in human epithelial caco-2 cells. J
S, Rosen SD. Vascular endothelial junction- Biol Chem. 2001;276:9291–9296.
associated molecule, a novel member of the 18. Hamazaki Y, Itoh M, Sasaki H, Furuse M, Tsukita
immunoglobulin superfamily, is localized to S. Multi-PDZ domain protein 1 (MUPP1) is con-
intercellular boundaries of endothelial cells. J Biol centrated at tight junctions through its possible
Chem. 2000;275:19139–19145. interaction with claudin-1 and junctional adhesion
4. Cunningham SA. A novel protein with homology molecule. J Biol Chem. 2002;277:455–461.
to the junctional adhesion molecule. Characteriza­ 19. Ebnet K, Aurrand-Lions M, Kuhn A et al. The junc-
tion of leukocyte interactions. J Biol Chem. 2000;​ tional adhesion molecule (JAM) family members
275:34750–34756. JAM-2 and JAM-3 associate with the cell polarity
5. Aurrand-Lions M, Duncan L, Ballestrem C, Imhof protein PAR-3: A possible role for JAMs in endothe-
BA. JAM-2, a novel immunoglobulin superfamily lial cell polarity. J Cell Sci. 2003;116:3879–3891.
molecule, expressed by endothelial and lymphatic 20. Cohen CJ, Shieh JT, Pickles RJ et al. The coxsacki-
cells. J Biol Chem. 2001;276:2733–2741. evirus and adenovirus receptor is a transmembrane
6. Arrate MP, Rodriguez JM, Tran TM, Brock TA, component of the tight junction. Proc Natl Acad Sci
Cunningham SA. Cloning of human junctional U S A. 2001;98:15191–15196.
adhesion molecule 3 (JAM3) and its identifica- 21. Coyne CB, Voelker T, Pichla SL, Bergelson JM.
tion as the JAM2 counter-receptor. J Biol Chem. The coxsackievirus and adenovirus receptor  inter­
2001;276:45826–45832. acts with the multi-PDZ domain protein-1
7. Liang TW. Vascular endothelial-junctional adhe- (MUPP1) within the tight junction. J Biol Chem.
sion molecule (VE-JAM)/JAM 2 interacts with T, 2004;279:48079–48084.
NK, and dendritic cells through JAM3. J Immunol. 22. Mirza M, Raschperger E, Philipson L, Pettersson RF,
2002;168:1618–1626. Sollerbrant K. The cell surface protein coxsackie-and
8. Hirabayashi S. JAM4, a junctional cell adhesion adenovirus receptor (CAR) directly associates with
molecule interacting with a tight junction protein, the Ligand-of-Numb Protein-X2 (LNX2). Exp Cell
MAGI-1. Mol Cell Biol. 2003;23:4267–4282. Res. 2005;309:110–120.
9. Moog-Lutz C. JAML, a novel protein with character- 23. Kostrewa D. X-ray structure of junctional adhesion
istics of a junctional adhesion molecule, is induced molecule: Structural basis for homophilic adhesion
during differentiation of myeloid leukemia cells. via a novel dimerization motif. Embo J. 2001;20:​
Blood. 2003;102:3371–3378. 4391–4398.
References 79

24. Prota AE. Crystal structure of human junc- 38. Wang CQ, Mruk DD, Lee WM, Cheng CY. Coxsackie
tional adhesion molecule 1: Implications for and adenovirus receptor (CAR) is a product of Sertoli
reovirus binding­. Proc Natl Acad Sci U S A. and germ cells in rat testes which is localized at the
2003;100:5366–5371. Sertoli–Sertoli and Sertoli–germ cell interface. Exp
25. Santoso S, Orlova VV, Song K et al. The homophilic Cell Res. 2007;313:1373–1392.
binding of junctional adhesion molecule-C mediates 39. Bazzoni G. Pathobiology of junctional adhesion mol-
tumor cell-­endothelial cell interactions. J Biol Chem. ecules. Antioxid Redox Signal. 2011;15:1221–1234.
2005;​280:36326–36333. 40. Garrido-Urbani S, Bradfield P, Imhof B. Tight junc-
26. Weber C, Fraemohs L, Dejana E. The role of junc- tion dynamics: The role of junctional adhesion mol-
tional adhesion molecules in vascular inflammation. ecules (JAMs). Cell Tissue Res. 2014;355:701–715.
Nat Rev Immunol. 2007;7:467–477. 41. Aurrand-Lions M, Johnson-Leger C, Wong C, Du
27. Bazzoni G, Martìnez-Estrada OM, Mueller F et al. Pasquier L, Imhof BA. Heterogeneity of endothelial
Homophilic interaction of junctional adhesion mol- junctions is reflected by differential expression and
ecule. J Biol Chem. 2000;275:30970–30976. specific subcellular localization of the three JAM
28. Lamagna C, Meda P, Mandicourt G et al. Dual inter- family members. Blood. 2001;98:3699–3707.
action of JAM-C with JAM-B and αMβ2 integrin: 42. Liu Y, Nusrat A, Schnell FJ et al. Human junction
Function in junctional complexes and leukocyte adhesion molecule regulates tight junction resealing
adhesion. Mol Biol Cell. 2005;16:4992–5003. in epithelia. J Cell Sci. 2000;113:2363–2374.
29. Ostermann G, Weber KS, Zernecke A, Schroder A, 43. Mandell KJ, McCall IC, Parkos CA. Involvement of
Weber C. JAM-1 is a ligand of the [beta]2 integrin the junctional adhesion molecule-1 (JAM1) homodi-
LFA-1 involved in transendothelial migration of leu- mer interface in regulation of epithelial barrier func-
kocytes. Nat Immunol. 2002;3:151–158. tion. J Biol Chem. 2004;279:16254–16262.
30. Naik MU, Mousa SA, Parkos CA, Naik UP. Signaling 44. Mandell KJ, Babbin BA, Nusrat A, Parkos CA.
through JAM-1 and αVβ3 is required for the angio- Junctional adhesion molecule 1 regulates epithelial
genic action of bFGF: Dissociation of the JAM-1 and cell morphology through effects on [beta]1 integrins
αVβ3 complex. Blood. 2003;102:2108–2114. and Rap1 activity. J Biol Chem. 2005;280:11665–11674.
31. Cunningham SA, Rodriguez JM, Arrate MP, Tran 45. Mandell KJ, Holley GP, Parkos CA, Edelhauser HF.
TM, Brock TA. JAM2 interacts with α4β1. Facilitation Antibody blockade of junctional adhesion molecule-
by JAM3. J Biol Chem. 2002;277:27589–27592. A in rabbit corneal endothelial tight junctions pro-
32. Mirza M, Hreinsson J, Strand M-L et al. duces corneal swelling. Invest Ophthalmol Vis Sci.
Coxsackievirus and adenovirus receptor (CAR) is 2006;47:2408–2416.
expressed in male germ cells and forms a complex 46. Laukoetter MG, Nava P, Lee WY et al. JAM-A regu-
with the differentiation factor JAM-C in mouse tes- lates permeability and inflammation in the intestine
tis. Exp Cell Res. 2006;312:817–830. in vivo. J Exp Med. 2007;204:3067–3076.
33. Santoso S, Sachs UJH, Kroll H et al. The junctional 47. Kang LI, Wang Y, Suckow AT et al. Deletion of
adhesion molecule 3 (JAM-3) on human platelets is JAM-A causes morphological defects in the corneal
a counterreceptor for the leukocyte integrin Mac-1. epithelium. Int J Biochem Cell Biol. 2007;39:576–585.
J Exp Med. 2002;196:679–691. 48. Khounlotham M, Kim W, Peatman E et al.
34. Li X, Stankovic M, Lee BP-L et al. JAM-C induces Compromised intestinal epithelial barrier induces
endothelial cell permeability through its association adaptive immune compensation that protects from
and regulation of β3 integrins. Arterioscler Thromb colitis. Immunity. 2012;37:563–573.
Vasc Biol. 2009;29:1200–1206. 49. Orlova VV, Economopoulou M, Lupu F, Santoso S,
35. Xia W, Wong CH, Lee NP, Lee WM, Cheng CY. Chavakis T. Junctional adhesion molecule-C regu-
Disruption of Sertoli-germ cell adhesion function lates vascular endothelial permeability by modulat-
in the seminiferous epithelium of the rat testis can ing VE-cadherin-mediated cell-cell contacts. J Exp
be limited to adherens junctions without affecting Med. 2006;203:2703–2714.
the blood–testis barrier integrity: An in vivo study 50. Raschperger E, Thyberg J, Pettersson S et al. The
using an androgen suppression model. J Cell Physiol. ­coxsackie-and adenovirus receptor (CAR) is an in
2005;205:141–157. vivo marker for epithelial tight junctions, with a
36. Shao M, Ghosh A, Cooke VG, Naik UP, Martin- potential role in regulating permeability and tissue
DeLeon PA. JAM-A is present in mammalian sper- homeostasis. Exp Cell Res. 2006;312:1566–1580.
matozoa where it is essential for normal motility. 51. Walters RW, Freimuth P, Moninger TO et al.
Dev Biol. 2008;313:246–255. Adenovirus fiber disrupts CAR-mediated inter-
37. Gliki G, Ebnet K, Aurrand-Lions M, Imhof BA, cellular adhesion allowing virus escape. Cell.
Adams RH. Spermatid differentiation requires 2002;110:789–799.
the assembly of a cell polarity complex down- 52. Coyne CB, Bergelson JM. CAR: A virus receptor
stream of junctional adhesion molecule-C. Nature. within the tight junction. Adv Drug Deliv Rev. 2005;​
2004;431:320–324. 57:869–882.
80  Junctional adhesion molecule (JAM) family

53. Su L, Mruk DD, Cheng CY. Regulation of the blood- 67. Xia W, Wong EW, Mruk DD, Cheng CY. TGF-
testis barrier by coxsackievirus and adenovirus recep- β3 and TNFα perturb blood–testis barrier (BTB)
tor. Am J Physiol Cell Physiol. 2012;303:C843–C853. dynamics by accelerating the clathrin-mediated
54. Liang TW, Chiu HH, Gurney A et al. Vascular endocytosis of integral membrane proteins: A new
endothelial­-junctional adhesion molecule (VE-­JAM)/​ concept of BTB regulation during spermatogenesis.
JAM 2 interacts with T, NK, and dendritic cells Dev Biol. 2009;327:48–61.
through JAM 3. J Immunol. 2002;168:1618–1626. 68. Wang Y, Lui W-Y. Opposite effects of interleukin-1α
55. Noutsias M, Fechner H, de Jonge H et al. Human and transforming growth factor-β2 induce stage-
coxsackie-adenovirus receptor is colocalized with specific regulation of junctional adhesion mol-
integrins αVβ3 and αVβ5 on the cardiomyocyte ecule-B gene in Sertoli cells. Endocrinology.
sarcolemma and upregulated in dilated cardiomy- 2009;150:2404–2412.
opathy implications for cardiotropic viral infections. 69. Gao Y, Lui W-Y. Synergistic effect of interferon-
Circulation. 2001;104:275–280. gamma and tumor necrosis factor-alpha on cox-
56. Shin K, Fogg VC, Margolis B. Tight junctions and cell sackievirus and adenovirus receptor expression:
polarity. Annu Rev Cell Dev Biol. 2006;22:207–235. An explanation of cell sloughing during testicular
57. Ebnet K, Suzuki A, Horikoshi Y et al. The cell polar- inflammation in mice. Biol Reprod. 2014;90:59.
ity protein ASIP/PAR-3 directly associates with 70. Pajukanta P. Familial combined hyperlipidemia
junctional adhesion molecule (JAM). EMBO J. is associated with upstream transcription factor 1
2001;20:3738–3748. (USF1). Nat Genet. 2004;36:371–376.
58. Suzuki A, Yamanaka T, Hirose T et al. Atypical pro- 71. Götte M, Mohr C, Koo C et al. miR-145-dependent
tein kinase C is involved in the evolutionarily con- targeting of junctional adhesion molecule A and
served par protein complex and plays a critical role modulation of fascin expression are associated with
in establishing epithelia-specific junctional struc- reduced breast cancer cell motility and invasiveness.
tures. J Cell Biol. 2001;152:1183–1196. Oncogene. 2010;29:6569–6580.
59. Suzuki A, Ishiyama C, Hashiba K et al. aPKC kinase 72. Cao M, Nie W, Li J et al. MicroRNA-495 induces
activity is required for the asymmetric differentiation breast cancer cell migration by targeting JAM-A.
of the premature junctional complex during epithe- Protein Cell. 2014;5:862–872.
lial cell polarization. J Cell Sci. 2002;115:3565–3573. 73. Koenen RR, Pruessmeyer J, Soehnlein O et al.
60. Wong EW, Mruk DD, Lee WM, Cheng CY. Par3/ Regulated release and functional modulation of
Par6 polarity complex coordinates apical ectoplas- junctional adhesion molecule A by disintegrin
mic specialization and blood–testis barrier restruc- metalloproteinases. Blood. 2009;113:4799–4809.
turing during spermatogenesis. Proc Natl Acad Scie 74. Rabquer BJ, Amin MA, Teegala N et al. Junctional
U S A. 2008;105:9657–9662. adhesion molecule-C is a soluble mediator of angio-
61. Cooke VG, Naik MU, Naik UP. Fibroblast growth genesis. J Immunol. 2010;185:1777–1785.
factor-2 failed to induce angiogenesis in junctional 75. Naik MU, Stalker TJ, Brass LF, Naik UP. JAM-A
adhesion molecule-deficient mice. Arterioscler Thromb protects from thrombosis by suppressing integrin
Vasc Biol. 2006;26:2005–2011. αIIbβ3-dependent outside-in signaling in platelets.
62. Sakaguchi T, Nishimoto M, Miyagi S et al. Putative Blood. 2012;119:3352–3360.
“stemness” gene jam-B is not required for mainte- 76. Khandoga A, Kessler JS, Meissner H et al. Junctional
nance of stem cell state in embryonic, neural, or hema- adhesion molecule-A deficiency increases hepatic
topoietic stem cells. Mol Cell Biol. 2006;26:​6557–6570. ischemia-reperfusion injury despite reduction of
63. Takehashi M, Kanatsu-Shinohara M et al. Production neutrophil transendothelial migration. Blood. 2005;​
of knockout mice by gene targeting in multipotent 106:725–733.
germline stem cells. Dev Biol. 2007;312:344–352. 77. Imhof B, Zimmerli C, Gliki G et al. Pulmonary dys-
64. Xu H, Oliveira-Sales EB, McBride F et al. function and impaired granulocyte homeostasis
Upregulation of junctional adhesion molecule-A result in poor survival of Jam-C-deficient mice. J
is a putative prognostic marker of hypertension. Pathol. 2007;212:198–208.
Cardiovasc Res. 2012;96:552–560. 78. Wyss L, Schäfer J, Liebner S et al. Junctional
65. Wang Y, Lui W-Y. Transforming growth factor-β1 adhesion molecule (JAM)-C deficient C57BL/6
attenuates junctional adhesion molecule-A and con- mice develop a severe hydrocephalus. PloS One.
tributes to breast cancer cell invasion. Eur J Cancer. 2012;7:e45619.
2012;48:3475–3487. 79. Pazirandeh A, Sultana T, Mirza M et al. Multiple
66. Zhang X, Lui W-Y. Transforming growth factor-β3 phenotypes in adult mice following inactivation of
regulates cell junction restructuring via MAPK- the Coxsackievirus and Adenovirus receptor (Car)
mediated mRNA destabilization and Smad- gene. PLoS One. 2011;6:e20203.
dependent protein degradation of junctional adhesion
molecule B (JAM-B). Biochim Biophys Acta (BBA)–
Gene Regulatory Mechanisms. 2015;1849:601–611.
Sertoli cell immune regulation
within the testis
6
GURVINDER KAUR, KANDIS WRIGHT, ROBIN HANNAH GREER, KARL MUELLER,
ALLAN HAYNES, and JANNETTE M. DUFOUR

INTRODUCTION supported by models of autoimmune orchitis where loss of


It is estimated that 8%–15% of couples worldwide are spermatogenesis resulting in infertility is due to an auto-
infertile. Of these, 40%–60% are infertile due to a male- immune reaction against the germ cells.8,9 Testis immune
related infertility problem.1,2 Causes include trauma, infec- privilege is clinically relevant as humans can also develop
tion, varicoceles, and environmental toxins.3 Associated autoimmune orchitis.4
inflammation can evoke an immune response to auto- Previously it was believed that the unique physiologi-
antigenic germ cells.4 Consistently, in developed nations cal conditions of the testis, such as the lower temperature
sperm autoantibodies have been detected in 5%–12% of of the scrotum, impaired lymphatic drainage or high zinc
male infertility cases.5 Under normal circumstances the concentrations were responsible for testis immune privi-
unique immune privilege environment of the testis pro- lege (Figure 6.2) (reviewed in Kaur et al.10). However, when
tects the germ cells from these autoimmune reactions. the testis was positioned in the abdominal cavity at nor-
Sertoli cells (SCs) play a central role in creation and main- mal core body temperature, it maintained the ability to
tenance of this unique environment, which is essential for protect transplanted cells. Similarly, studies of testicular
the immune protection of autoantigenic germ cells. This lymphatic drainage revealed a fully functional lymphatic
chapter will focus on the unique immunoregulatory abili- system. The testis has elevated levels of zinc. Given that
ties of the SCs necessary for immune protection of germ high levels of zinc can suppress allograft rejection, it was
cells. thought that this could be responsible for testis immune
privilege. However, transplantation studies demonstrated
TESTIS IMMUNE PRIVILEGE that zinc was not the key component. Pancreatic islets,
The testis is the central male reproductive organ; as such, which contain high levels of zinc, are rejected when trans-
it is the site of androgen production and spermatogenesis. planted as allografts or xenografts, and when parathy-
Morphologically, the testis is comprised of the seminifer- roid allografts were transplanted into the prostate, which
ous tubules separated by the interstitial space. The semi- contains the highest zinc concentration of all organs, the
niferous tubules consist of more than 80% of the mass of parathyroid grafts were similarly rejected (reviewed in
the adult testis and are composed of SCs and developing Kaur et al.10).
germ cells surrounded by single (rodents) or multiple Other studies examining the main cellular compo-
(nonhuman primates and humans) layers of peritubu- nents of the testis point to SCs as important players
lar myoid cells. The interstitium consists of Leydig cells in testis immune privilege (Figure 6.2). Foreign tissue
(cells that produce androgens), macrophages, blood ves- transplanted in testes depleted of germ cells, either
sels, lymphocytes, and fibroblasts (Figure 6.1). During by cold testicular ischemia, placement of testes in the
spermatogenesis, which takes place in the seminifer- abdominal cavity, or irradiation, continue to enjoy
ous tubules, the undifferentiated spermatogonial cells prolonged graft survival (reviewed in Kaur et al.10).
undergo three phases of development: mitosis (prolifera- Furthermore, depletion of Leydig cells still resulted in
tion, where spermatogonia undergo self-renewal or dif- the immune protection of allografts transplanted into
ferentiation), meiosis (spermatocyte DNA recombination, the testes (reviewed in Kaur et al.10), although there
reduction, and division), and spermiogenesis (spermatid is evidence that Leydig cells (by secreting androgens)
differentiation), to generate highly specialized spermato- and germ cells, while not essential, contribute to tes-
zoa. As the germ cells progress through development, they tis immune privilege. Additionally, the immune cells
begin to express novel antigens that first appear after the located within testis and testicular draining lymph
establishment of systemic self-tolerance. Despite the pres- nodes are of an altered phenotype (M2 macrophages,
ence of new surface and intracellular proteins, an immune immature dendritic cells, and regulatory T cells) and
response is generally not elicited against these germ cells6,7 contribute to the unique testicular immune environ-
due to the immune privileged environment within the tes- ment (Figure 6.2). However, immune cells need to be
tis. The importance of immunological tolerance to the guided by external signals to take on and maintain
spermatocytes and spermatids was demonstrated by the this regulatory phenotype, suggesting that other cells
lysis of these cells after injection of sera collected from within the testis, such as SCs, are actively involved in
rodents immunized with whole semen6,7 and is further creating this immune privileged environment.

81
(a)
Lumen (b)
Spermatid

Round
spermatid SC Adluminal
compartment

Spermatocyte
Tight junction
SC
Spermatogonia nucleus Basal
compartment
Peritubular
myoid cell
T cell DC Blood vessel

Leydig cell Interstitium



50 µm
82  Sertoli cell immune regulation within the testis

(c) (d)

50 µm 50 µm

Figure 6.1  (a) Schematic illustration of a cross section of a seminiferous tubule. Testis interstitium contains Leydig cells, macrophages (MΦ), blood vessels, lymphocytes, and dendritic
cells (DC). The seminiferous epithelium is surrounded by peritubular myoid cells and is composed of SCs and maturing germ cells. Tight junctions between adjacent SCs divide the tubules
into basal and adluminal compartments. (b–d) Testes were collected from a 51-year-old male (b) or 6–8-week old BALB/c mice (c and d), paraffin embedded and sectioned. Tissue sec-
tions were immunostained for Wilms’ tumor 1 to detect SCs (brown color, c) or smooth muscle alpha actin that detects peritubular myoid cells and blood vessels (brown color, b and d).
Sections were counterstained with hematoxylin for cell nuclei (blue color, b–d). All animal experiments were performed in accordance with guidelines of the National Institutes of Health
and Institute for Laboratory Animal Research Care and Use of Laboratory Animals, and TTUHSC Institutional Animal Care and Use Committee approved protocols. For the human sample,
the study was approved by the Institutional Review Board. An informed consent was obtained from the patient.
Blood-testis barrier or SC barrier  83

What is responsible for testis immune privilege?

Impaired lymphatic Lower temperature? Zinc? Blood testis barrier (BTB)?


drainage?
Within minutes, dye and cells injected Allogeniec parathyroid tissue was Transplanted parathyroid tissue Allogeniec or xenogeniec skin and
into the testis drained to lymph nodes. rejected following transplantation into survived in the testis with a high [Zn], parathyroid tissue survived post
the subcutaneous ear (hypothermic), but was rejected in the prostate despite transplantation into the testis
but survived post transplantation in the having the highest [Zn]. interstitum (outside the BTB).
1. Dye or cells cyrptorchid testis (in the abdomen at
2. Lymphatic 1. Parathyroid cells 1. Parathyroid cells
drainage body temperature).
Skin cells
1. Parathyroid cells Scrotol testis Interstitum
Prostate BTB
Epididymis

Dsp LCs
Cyrptorchid
Ear
Scrotol testis testis T-cell
( [Zn]) ( [Zn])
Testis

( Temp) M
( Temp) (Scrotol temp)

DC 2. CELL
3. Lymph nodes 2. REJECTION 3. SURVIVAL 2. SURVIVAL 3. REJECTION SURVIVAL

NO NO NOT REQUIRED NOT SOLE FACTOR

Germ cells (GCs) and


Leydig cells (LCs)? Other immune cells? Sertoli cells (SCs)?
spermatogenesis
Transplanted islet or parathyroid tissue The human testis contains SCs survive long term post allo- and
Islet allografts survived transplantation
survived in both cryptorchid and macrophages, immature dendritic cells, xeno- transplantation without immune
into the testis despite LC depletion
irradiated testis lacking and T-cells. Under certain suppression and they can protect other
with dimethansulphonate (EDP) or
spermatogenesis and GCs. circumstances T-regulatory cells (Treg) co-transplanted cells (islets,
leuprolide (GnRH analong inhibiting
testosterone production). may also be present in the testis hepatocytes, etc.) from tissue rejection.
1. Parathyroid cells
interstitium.
or islet cells
1. EDP or leuprolide Cyrptorchid 1. Islet cells 2. Isolated 3. Combined
Irratiated testis BTB T-cell
testis SCs
LCs
SC
2. Islet SC SC
SC LCs Treg +
cells
Transplant
SC under the
SC No spermatogenesis
M kidney
or GCs
capsule

3. CELL 2. SURVIVAL 3. SURVIVAL DC 1. CELL 2. and 3. CELL


SURVIVAL REJECTION SURVIVAL
NOT REQUIRED NO LIKELY INVOLVED YES

Figure 6.2  Schematic illustration of potential testis immune privilege mechanisms. Lymphatic drainage, scrotal temperature,
zinc concentration, Leydig cells, germ cells, spermatogenesis, and the BTB/SCB are not solely responsible for testis immune privilege,
while SCs are critical in immune privilege. Temperature (temp); zinc concentration ([Zn]); developing spermatogonia (Dsp); macro-
phages (MΦ); dendritic cells (DCs); dimethanulphonate (EDP); gonadotropin-releasing hormone (GnRH); T-regulatory cells (Tregs).

BLOOD-TESTIS BARRIER OR SC BARRIER into mice expressing diphtheria toxin receptor specifi-
SCs extend from the outer edge of the seminiferous tubules cally in SCs, there was a rapid loss of germ cells, and by
to the lumen and completely surround the majority of the 10 days postinjection no germ cells were detected in the
developing germ cells. Tight junctions located along the testis,13,14 indicating loss of spermatogenesis and the stem
basal region between adjacent SCs form a physical barrier cell niche.12,13
that separates the seminiferous tubules into basal (con- In addition to controlling the transport of factors neces-
taining spermatogonia and preleptotene spermatocytes) sary for germ cell development, the BTB/SCB prevents the
and adluminal (containing most spermatocytes and sper- passage of antibodies into the adluminal compartment of
matids) compartments (Figure 6.1). This barrier, known the seminiferous epithelium15,16 and in combination with
as the blood-testis barrier (BTB) or SC barrier (SCB), the peritubular myoid cell layer (Figure 6.1), which forms a
allows SCs to control and regulate the milieu required for semipermeable barrier, inhibits the entry of immune cells
the development of the germ cells by separating the cells into the seminiferous epithelium.17–19 SC ablation studies
within the adluminal compartment from the blood supply found that SCs are necessary for peritubular myoid cell
and through expression of transporters to control passage fate and establishment of the tubule architecture prior
of numerous factors.11 SC ablation studies demonstrated to puberty.12,13 Depletion of SCs in the embryo or in the
that SCs are critical for germ cell survival, maintenance of neonate resulted in loss of tubule architecture, the absence
spermatogenesis and the spermatogonial stem cell niche, of smooth muscle actin, and a decrease in other myoid
as well as survival of adult Leydig cells.12–14 When SCs cell markers, suggesting dedifferentiation of peritubular
were depleted after a single injection of diphtheria toxin myoid cells. In contrast, if SCs were depleted at puberty or
84  Sertoli cell immune regulation within the testis

in the adult, the basic tubule architecture was maintained the majority of the tubules from meiotic arrest (55%) and
and smooth muscle actin expressing peritubular myoid SC-only syndrome (61%) subjects had a punctate claudin
cells were still present, although expression of myoid cell 11 staining pattern that was spread throughout the basal
functional markers was decreased.13,20 Analysis of the and adluminal aspects of the tubules.29
integrity of the BTB/SCB after depletion of SCs in adults The importance of the other components of the BTB/
demonstrated that SCs are necessary to prevent entry of SCB is not clear. ZO-1 and -2 knockout mice are embry-
molecules into the tubules while the remaining peritu- onic lethal and claudin-5 knockout mice die within
bular myoid cells were sufficient to inhibit immune cell 10  days of birth.32,33 In humans ZO-1 has been found to
infiltration. colocalize with claudin 11 and in tubules with carcinoma
Given that the BTB/SCB prevents entry of antibodies in situ ZO-1 and -2 expression is decreased.28,34 Deletion
and immune cells from entering the adluminal compart- of JAM-A results in subfertility in mice, although this is
ment of the seminiferous tubules and that the majority due to a defect in sperm motility as they have normal tes-
of the autoantigenic advanced germ cells are sequestered tis morphology.35 Mice with JAM-B gene disruption are
behind the BTB/SCB, historically testis immune privilege fertile with normal testicular morphology,36 while ZO-3
was attributed solely to the presence of the BTB/SCB. The knockout mice have no apparent phenotype.33 For claudin
BTB/SCB consists of tight junction proteins that include 3, it was initially concluded that the increased permeabil-
members of the claudin family, occludin, zonula occludens ity of the BTB/SCB and germ cell autoantibodies in SC
(ZOs), junctional adhesion molecules (JAMs), and tricel- androgen receptor knockout (SCARKO) mice was due to
lulin.21 In addition to the tight junction proteins, special- decreased claudin 3 expression.37,38 However, it has since
ized adherens junctions (basal ectoplasmic specializations been shown that claudin 3 knockout mice are fertile and
and basal tubulobulbar complex), desmosomes, and gap have an intact barrier function.39 The importance of ZOs,
junctions also contribute to the functionality of the BTB/ JAM-A and –B, and claudin-3 and -5 in human spermato-
SC barrier.21 genesis is not well studied.
Of all the tight junction proteins, occludin and claudin Experimental autoimmune orchitis (EAO) studies sup-
11 are critical for barrier integrity as male mice lacking port the immunological importance of the BTB/SCB. The
occludin or claudin 11 are infertile.22 Histological analy- BTB at the rete testis is incomplete40–43 and leaky to anti-
sis of testes from the occludin knockout mice revealed bodies,44 making this region susceptible to the development
normal testicular morphology, containing germ cells, in of autoimmune orchitis. Under normal circumstances
young animals (6 weeks old), but as the animals aged (40– spermatozoa traverse through this region in immense
60 weeks old) atrophied tubules devoid of germ cells were numbers and yet an immune response is not generated
observed.22 Similarly, a single injection of synthetic occlu- against these germ cells. However, under certain condi-
din peptide resulted in disruption of the BTB/SCB, loss tions an immune response can be triggered. For instance,
of elongated spermatids (day 8 postinjection), and sper- immunization of mice with syngeneic testicular germ cells
matocytes and spermatids (day 27 postinjection) in rats.23 (without adjuvant) or adoptive transfer of autoreactive
While the immune response was not measured in this CD4 T cells leads to EAO. Analysis of the testis revealed
study it is interesting to note that the treatment was revers- that lymphocyte infiltration started in the rete testis with
ible and despite disruption of the BTB/SCB for several lymphocyte migration into the epithelium and epithelial
weeks, normal spermatogenesis was observed by 68 days degeneration followed by infiltration into the rest of the
postinjection. Consistently, in mice treated with a mutant testis and loss of the germ cells.45,46 Overall, it can be con-
occludin peptide, the integrity of the BTB/SCB was dis- cluded that the BTB/SCB is critical for spermatogenesis by
rupted, demonstrated by permeability to inulin, resulting creating a controlled environment for germ cell develop-
in germ cell loss. Nevertheless, antisperm antibodies were ment. While the importance of the tight junction proteins
not detected in serum.24 Claudin 11 knockout mice are in protection of germ cells from an autoimmune response
sterile and their seminiferous tubules contain degenerat- is less clear, sequestering the majority of the autoantigenic
ing germ cells. Despite the lack of tight junctions in these germ cells behind the BTB/SCB is important to decrease
mice, an immune response is not generated as testicular antigen exposure and prevent an immune response.
autoantibodies were not detected in the serum or within
the adluminal compartment of the seminiferous tubules MOVEMENT OF GERM CELLS ACROSS THE BTB/SCB
and CD4 T cell infiltrate was not detected within the tes- If sequestering of germ cells behind the barrier is nec-
tis.25 Interestingly, in humans and guinea pigs, SC tight essary, how do germ cells cross the BTB/SCB to enter
junctions lack occludin,26 suggesting occludin may not be adluminal compartment without exposing the advanced
critical for the integrity of human or guinea pig BTB/SCB. germ cells? The BTB/SCB undergoes extensive restructur-
In testicular biopsies of men with impaired spermato- ing at late stage VIII–early stage IX to allow the transit
genesis the spatial organization of claudin 11 was altered of preleptotene/leptotene spermatocytes from the basal to
and increased claudin 11 expression was detected.27–31 For adluminal compartment. The movement of preleptotene/
instance, 82% of control subject’s seminiferous tubules leptotene spermatocytes across the BTB/SCB never elicits
featured a filamentous claudin 11 staining pattern that an autoimmune reaction, suggesting that restructuring of
was restricted to the basal aspect of the tubule. In contrast, the BTB/SCB is a controlled process. The exact mechanism
Immune regulation by Sertoli cells  85

by which this transit takes place is largely unknown. Several antagonist acyline to suppress spermatogenesis followed
theories for migration of preleptotene/leptotene spermato- by testosterone treatment to reinitiate spermatogenesis.
cytes across the BTB/SCB without compromising its integ- Using permeability tracers of different sizes (0.6, 70, and
rity have been suggested. (1) The “zipper theory” proposes 150 kDa) to emulate factors that could cross the BTB/SC,53
that occludin fibrils positioned above the preleptotene/ it was demonstrated that meiosis occurs when the BTB/
leptotene spermatocytes are dissolved and new occludin SCB is permeable to factors up to 70 kDa (such as growth
fibrils are reformed under these spermatocytes, resulting factors, cytokines, and hormones) but not 150 kDa (such
in their migration across the BTB/SCB.47 (2) The “repeti- as antibodies).
tive removal of membrane segments theory” suggests that Clinically fine needle biopsies are routinely performed
the stress created by upward migration of germ cells alters to obtain human sperm for in vitro fertilization or to
the SC junctional complexes, resulting in proliferation of analyze testis samples. Even though this procedure can
junctional complexes to form intercellular pockets around cause local injury to the seminiferous epithelium, it does
the developing germ cells (except elongated spermatids). not normally lead to autoimmune orchitis.54 Consistently,
These intercellular pockets, containing developing germ traumatic injury by puncturing the testis or epididymis
cells, are sealed at both ends by tight junctions, adherens, multiple times with a needle, resulting in migration of
and gap junctions and traverse through the seminiferous germ cells into the interstitial space, causes immune cell
epithelium (see Mruk and Cheng,21 Pelletier48). (3) The infiltration and granuloma formation in the epididymis,
“junctional restructuring theory” suggest that germ cell which does not occur in the testis.55 Moreover, injections
movement consists of intermittent phases of tight junc- of testicular germ cells, spermatozoa, or Bordetella pertus-
tion disassembly and assembly, tightly controlled by cyto- singens into the testis interstitial space does not result in
kines that are released by SCs and/or germ cells, which granuloma formation or immune cell infiltrate while sim-
allows passage of developing germ cells from the basal to ilar experiments performed in the epididymis leads to in
adluminal compartment (see Mruk and Cheng21). (4) The an increase in inflammatory cells and granuloma forma-
“intermediate cellular compartment theory” supports the tion within the epididymal interstitium.55,56
existence of a third transient compartment, containing Transplantation studies further demonstrate that the
leptotene spermatocytes, in between basal and adlumi- whole testis rather than just the adluminal compartment
nal compartments.49 Once new tight junctions are formed of the testis is immune privileged. For instance, trans-
near the basal end of the transient compartment the old plantation of allogeneic or xenogeneic pancreatic islets
tight junctions are dissolved. The junctional restricting or parathyroid grafts, and allogeneic insulinomas, skin
and intermediate cellular compartment theories gained grafts, or pituitary grafts, into the testis prolonged their
popularity due to recent evidence derived from biochemi- survival compared to other nonimmune privileged sites,
cal studies.21,50 For instance, whole seminiferous tubules even though the transplanted tissue or cells were located
dissected from adult mice were immunostained for clau- in the testis interstitium (outside the BTB/SC barrier)
din 11 and imaged by confocal microscopy to visualize the (Figure 6.2) (extensively reviewed in Kaur et al.10). In
three-dimensional organization of the SC tight junctions. addition, successful syngeneic or allogeneic spermatogo-
Analysis revealed the presence of claudin 11 on both the nial stem cell transplantation resulted in sperm produc-
basal and apical sides of the migrating spermatocytes, tion in chickens, fish, and large animals without the use
resulting in the enclosure of leptotene spermatocytes in an of immune suppression (reviewed in Honaramooz and
intermediate compartment.50 Yang57). Interestingly, transplantation of germ cells did not
evoke an inflammatory immune response even though the
TESTIS IMMUNE PRIVILEGE: NOT MERELY DEPENDENT germ cells colonized the basal compartment of the semi-
ON THE BTB/SCB niferous tubules (reviewed in Kaur et al.,10 Honaramooz
Reducing antigen exposure by sequestering the advanced and Yang57). Collectively these studies demonstrate that
germ cells behind the BTB/SCB is important to prevent germ cells and other cells transplanted into the testis can
an immune response to the advanced germ cells. However, be exposed to the immune system without evoking an
the whole testis, not just the adluminal compartment of immune response, suggesting that other mechanism(s)/
the seminiferous tubules, is immune privileged as dem- factor(s) work in collaboration with the BTB/SCB to create
onstrated by the studies described below. Under normal a tolerogenic environment in the testis.
circumstances preleptotene spermatocytes fail to gener-
ate an immune response even though they are located IMMUNE REGULATION BY SERTOLI CELLS
within the basal compartment of the seminiferous epi- Transplantation studies provide evidence that SCs mod-
thelium, outside of the BTB/SCB, and it has been shown ify the immune response to create an immune privileged
previously that they express antigens that can evoke an environment for their survival and also the survival of
immune response.51 In seasonal breeders the BTB/SCB cotransplanted cells. SCs survive as allografts or xenografts
breaks down during the nonbreeding cycle repeatedly without the use of chronic immune suppression when
exposing the meiotic spermatocytes, and yet, an immune transplanted outside the testis (Figure 6.2). Additionally,
response does not develop.52 In a recent study BTB/SCB SCs provide immune protection to cografted cells such
restructuring was analyzed in rats treated with the GnRH as pancreatic islets to treat diabetes, adrenal chromaffin
86  Sertoli cell immune regulation within the testis

T-cell

Cell mediated Immunoregulatory Sertoli


M
cytotoxicity factors cell Immune tolerant
Apoptosis Apoptosis and environment
DC antibodies complement inhibitors
complement

Figure 6.3  Generalized Sertoli cell modulation of the immune response. Immune-mediated cytotoxicity occurs when antigen
presenting cells (APCs) (macrophages or dendritic cells), B cells, and T cells are activated against the autoimmunogenic germ cells.
SCs express immunoregulatory factors, apoptosis inhibitors, and complement inhibitors to protect cells from detrimental immune
responses and convert the cytotoxic immune cells to be more regulatory. This creates an immune-tolerant environment within the
testis. Pink circles represent SC surface factors. Purple circles represent SC secreted factors. Green circles represent SC intracellular
factors. Macrophages (MΦ); dendritic cells (DCs).

cells, or dopaminergic neurons for neurodegenerative dis- (2) releasing proinflammatory cytokines, or (3) TNF fam-
eases and hepatocytes for liver disease when transplanted ily death receptor signaling. Both the uptake of perforin
across immunological barriers as allografts or xenografts and granzymes and TNF family death receptor signaling
(reviewed in Kaur et al.58). These studies suggest modula- causes apoptosis of target cells.
tion of the immune response by SCs plays a key role in SCs are resistant to complement mediated cell lysis
testis immune privilege. since they survive both when transplanted as discordant
A fully functional immune response is required for pro- xenografts,59,60 which by definition indicates the presence
tecting human beings from deadly pathogens and foreign of antibodies directed against transplanted cells, and when
antigens while at the same time it also has the potential to subjected to an in vitro human antibody/­ complement
negatively impact male reproduction. Thus, regulation of mediated killing assay.61 For example, neonatal pig SCs
the immune system is required for successful spermato- (NPSCs) survive throughout the study (at least 42, 90,
genesis in the testis. Fundamentally, an immune response and 30 days) when transplanted into mice, rats, and dogs,
can be divided into humoral and cell-mediated responses. respectively.59,60 No immune suppression was used in
The cellular components of the humoral and cell-mediated rodent recipients while a short course (10 days) of immu-
immune response mainly include B cells, NK cells, T cells, nosuppression was used in dogs. Recently, it was shown
and antigen-presenting cells (APCs, such as dendritic that NPSCs survive throughout the study (40 days) despite
cells [DCs] and macrophages). Studies investigating the the presence and deposition of preformed antibodies,
immune privilege mechanism of SCs in vitro and in vivo while neonatal pig islets (NPIs) were rejected when trans-
(SC transplantation studies) clearly indicate that SCs have planted as xenografts underneath the kidney capsule of
the capability to modify both the humoral and cell medi- rats.62 SCs express several complement inhibitors includ-
ated immune responses (Figure 6.3). Here, we will spe- ing membrane cofactor protein (MCP; CD46), decay
cifically discuss the factor(s)/mechanism(s) by which SCs accelerating factor (DAF; CD55), SERPING1, clusterin,
regulate immune cells. and CD59.63–66 Comparison of the levels of these comple-
ment inhibitors revealed that NPSCs express MCP and
Humoral immune response DAF at significantly higher levels than the rejected NPIs,62
In response to nonself antigens, B cells produce antibod- suggesting these inhibitors could explain how SCs escape
ies (IgM and IgG) that bind to these antigens and coat the complement mediated cell lysis (Figure 6.4a).
target cells. This results in killing of the target cell through In rodent and human testis, substantial numbers of
the following pathways (Figure 6.4a). Antibodies acti- cells expressing NK cell markers are detected and a sig-
vate the complement cascade leading to formation of the nificant proportion of these cells are NKT cells.67 Further
membrane attach complex (MAC) on the cell membrane analysis of NKT cells from rat testis revealed that under
of the target cells, which leads to complement mediated both unstimulated and phorbol 12-myristate 13-acetate
cell lysis. Additionally, complement cascade activation (PMA)/ionomycin-stimulated conditions, the NKT cells
causes the release of anaphylatoxins, specifically C4a, C3a, produce interleukin (IL)-10,67 suggesting these cells have
and C5a, which mediate acute inflammation by attracting a tolerogenic phenotype in the testis. Studies investigating
immune cells. Lastly, natural killer (NK) cells can initi- the direct effect of SCs on NK cells are limited. In vitro
ate antibody-dependent cellular cytotoxicity by binding to analysis revealed that rat SC secreted factors inhibit rodent
the IgG1 or IgG3 coated cells through their low affinity and human NK cell activity but fail to suppress the lysis of
Fcy receptor. After binding, NK cells kill the target cells YAC-1 cells (mouse lymphoma cell line sensitive to lysis
through several different mechanisms: (1) exocytosis of by NK cells) by rat NK cells.68 In vivo, despite the deposi-
cytoplasmic granules containing perforin and granzymes, tion of IgG antibody, NK cell infiltration was not detected
(a) Humoral immune response
Secreted factors
SERPING1 Generation of
TGF-β (Immunoregulatory factor)
MCP anaphylatoxins
C1 (C4a, C3a, C5a) Thrombospondin 1&2 (Enzymes that active TGF-β)
Target complex DAF
NK-cell Activin A (Activates alternative macrophages)
cell death Galectin-1 (Immunoregulatory factor)
? C4b
C3 C2a Clusterin CCL7 (Immunoregulatory cytokine and chemokine)
Convertase C4b CD59
C2a
CXCL1
SC C5 C3b
Cell SERPING1 (Complement inhibitor)
Convertase (Apoptosis inhibitor)
MAC lysis SERPINA3n
(C5b-9) SERPINB9
?
PGE2 (Anti-inflammatory and T-cell inhibitor)
B-cell PGI2 (Anti-inflammatory)
Antibodies Target cell lysis Intracellular factors
by complement
activation IDO (T-cell inactivation and immune suppression)
PLA2G4A (Anti-inflammatory prostanoid production)
(b) Cell-mediated immune response PLA2G6
PTGES (Anti-inflammatory prostaglandin production)
ivin
Regulatory MΦ
P 1&2, Act
PTGIS
TGF-β, TS in-1, IDO and DC
A, G al e ct
Surface factors
T-cell MCP (CD46) (C3 and C5 convertase inhibitor)
SC
TGF-β DAF (CD55)
,
Galec TSP 1&2
tin-1, Indirect Clusterin (MAC inhibitor)
IDO
n CD59
Target cell A3 Direc
t
PIN 9 B7-H1 (T-cell negative regulatory receptor)
death SER PINB
Regulatory T-cells
SER
(apoptosis) (tregs)

Figure 6.4  Factors expressed by Sertoli cells that modulate the humoral and cell-mediated immune responses. SCs express various intracellular, cell membrane, and secreted factors
that modulate and maintain a tolerogenic environment. (a) Humoral immune response: SCs inhibit complement mediated cell lysis both directly and indirectly. In the smaller box, antibod-
ies (IgG and IgM) bind to the target cell. Binding of the C1q complex to these antibodies results in the formation of the C1 complex, which activates the complement cascade by generating
C3 convertase. C3 is recruited to the C3 convertase and together forms the C5 convertase, which cumulates factors (C5-C9) to produce the membrane attack complex (MAC) on the target
cell surface. Formation of both the C3 and C5 convertases generates anaphylatoxins that increase inflammation, chemotaxis, and immune cell recruitment. SCs inhibit the complement
cascade by expressing the complement inhibitors (pink dots, green text). SCs can inhibit B cell proliferation through unknown factors (black dots), thereby preventing the production of
antibodies and activation of complement cascade indirectly. SCs can also inhibit NK cells. Red arrows represent immune response pathways and green lines represent SC inhibition of
these pathways. (b) Cell-mediated immune response: SCs inhibit T cell mediated apoptosis directly by secreting apoptosis inhibitors (SERPINA3n and SERPINB9; yellow dots) or indirectly by
inhibiting T cell proliferation. Factors expressed by SCs such as transforming growth factor (TGF)-β, thrombospondin (TSP)-1 and -2, activin A, galectin-1, and indoleamine 2,3-dioxygenase
(IDO) are implicated in inducing regulatory antigen presenting cells (APCs) and Tregs. SCs can induce Tregs directly or indirectly through regulatory APCs. Collectively this tolerance results
in survival of the target tissue. Together these factors are crucial in maintaining testicular immune privilege and protecting autoimmunogenic germ cells. SC factors are listed in the upper
right box. Chemokine ligand 7 (CCL7); chemokine ligand 1 (CXCL1); serpin family G member 1 (SERPING1); prostaglandin (PGE2); prostacyclin (PGI2); phospholipase A2 Group IVA (PLA2g4a);
phospholipase A2 Group VI (PLA2g6); prostaglandin E synthase (PTGES); prostaglandin 12 synthase (PTGIST); membrane cofactor protein (MCP); and decay accelerating factor (DAF).
Immune regulation by Sertoli cells  87
88  Sertoli cell immune regulation within the testis

in surviving SC allografts or xenografts or rejecting NPI Tregs. Furthermore, IDO expression by SCs was critical as
xenografts, suggesting that NK cells play a minimal role in SCs silenced for IDO expression failed to protect the NOD
SC graft survival or NPI graft rejection at the kidney site mice from diabetes development.77 Collectively, these data
(unpublished data, Dufour lab, 2012). demonstrate that SC have the capability to generate reg-
ulatory APCs and T cells, which could explain how SCs
Cell mediated immune response protect autoantigenic germ cells (Figure 6.4b).
T cells and APCs are the main constituents of the cell-
mediated immune response. T cells stimulated by APCs POTENTIAL DOWNSIDE OF IMMUNE PRIVILEGE
kill tissue via apoptosis mediated by the Fas-FasL or perforin- Immune privilege has the possibility to be detrimental,
granzyme B pathways. Additionally, antigen-primed, CD4 as the immune system functions to protect the body from
T cells activate macrophages creating a proinflammatory infections and cancerous transformations, and immune
environment that promotes cell death. Several in vitro modulation may pose an increased risk of these events.
and in vivo studies suggest that SCs modulate the overall For example, immune privilege may play an important
immune response (Figure 6.4b). Transplanted SCs have the role in the disease progression of human immunodefi-
capability to escape cell death via apoptosis as they express ciency virus (HIV). It has been proposed that the testis
SERPINA3N and SERPINB9,69–71 both inhibitors of the may act as an HIV sanctuary site and protect the virus
granzyme B apoptosis pathway. While controversial, it has during antiretroviral therapy.86 The potential for testis
also been reported that SCs can inhibit the Fas-FasL path- immune privilege to be used by viral pathogens may not
way (reviewed in Mital et al.72). In vitro experiments using be unique to HIV, and there is some evidence that Ebola
rodent SC conditioned media (SCCM) demonstrated that and Zika viruses may also use the testis as a reservoir. One
SCs are capable of inhibiting the proliferation of acti- study found that all nine males tested had Ebola-virus–
vated lymphocytes (B and T cells).73–75 Treatment with positive semen 3 months after onset of illness, half were
SCCM significantly reduced the IL-2 production and IL-2 positive 4–6 months after the onset of illness, and one
responsiveness by lymphocytes.73,74 Moreover, SCs express quarter were positive 7–9 months after onset of illness.87
several immunoregualtory factors, such as ­ galectin-1, In another study it was reported that Ebola was present in
indoleamine-pyrrole 2,3-dioxygenase (IDO), prosta- the semen of survivors in Liberia for 18 months after infec-
glandin, transforming growth factor (TGF)-β),76–80 and tion.88 Moreover, transmission of the Zika virus through
have the capability to modulate the immune response in the semen from sexual contact has been reported.89
the testis or when transplanted ectopically. For instance, Despite testis being an immune privileged tissue, infec-
testicular macrophages and DCs express MHC-II and tions within the testis are rare67 and genitourinary tract
costimulatory molecules, implying these cells can mount infections (with the exception of viral pathogens) seldom
an effective immune response. Yet upon lipopolysaccha- involve the testis,90,91 suggesting that testis immune privi-
ride stimulation, rodent testicular macrophages express lege is not a permanent phenomenon and can be broken
low levels of proinflammatory cytokines and high levels down if it threatens the existence of the host. On the same
of the anti-inflammatory cytokine, IL-10 (reviewed in note, the rate of cancer in the testis is not different than in
Winnall and Hedger81). Similarly, DCs isolated from the other sites.67 Primary testicular cancer is infrequent, with
testis or testicular lymph nodes are unable to induce T cell germ cell tumors (GCTs) accounting for 1% of malignan-
proliferation.82 cies in males in the United States.92 The low incidence of
There is evidence that SCs induce tolerogenic APCs. infections and cancer has been attributed to activation of
Coculture of immature DCs with SCs or SCCM resulted the innate and adaptive immune responses.4 Innate immu-
in downregulation of MHC-II and costimulatory mol- nity is initiated by recognition of pathogen-associated
ecule (CD80, CD83, and CD86) expression on DCs.83 molecular patterns (found on bacteria, virus fungus, and
Additionally, these DCs displayed reduced phagocytic protozoa) by pattern recognition receptors such as Toll-
activity, expressed low levels of proinflammatory cyto- like receptors (TLRs). So far, 10 and 13 TLRs have been
kines (IL-12p70 and tumor necrosis factor-α) and high identified in human and rodents, respectively.93 Several
levels of anti-inflammatory cytokines (IL-10 and TGF-β). studies have demonstrated that SCs and testicular mac-
These SC conditioned DCs were tolerogenic as they inhib- rophages express a majority of these TLRs and respond
ited T cell proliferation and induced CD4+CD25+Foxp3+ to TLR activation by initiating inflammation, thereby
regulatory T cells (CD4 Tregs). Galectin-1, expressed by clearing pathogens and protecting the host (reviewed in
SCs, was critical as knockdown of galectin-1 in SCs using Hedger,94 Meinhardt and Hedger95). Interestingly, GCTs,
siRNA abrogated the generation of tolerogenic DCs. It particularly seminomas (Figure 6.5), have characteris-
has also been demonstrated in vitro that SCs can directly tic immune cell infiltrates that consist mainly of mac-
induce CD4 Tregs.84,85 Similarly, transplantation of por- rophages and T lymphocytes that correlate with a better
cine SCs resulted in diabetes prevention and reversion prognosis. At the same time there is a lack of suppressor T
in 88% and 81% of the non-obese diabetic (NOD) mice, cells in GCTs, perhaps reflecting a gradual breakdown of
respectively.77 This effect was associated with the induc- testicular immune privilege. Thus active immune surveil-
tion of a regulatory environment by SCs as evidenced lance leads to immune infiltrate, breakdown of the BTB/
by generation of anti-inflammatory cytokines and CD4 SCB, and loss of immune privilege, resulting in a good
References 89

(a) (b)

200 µm 200 µm
(c) (d)

200 µm 50 µm

Figure 6.5  Testicular germ cell tumor. (a) and (b) Hematoxylin and eosin staining of testicular tissue sections collected from
a 36-year-old male. The patient was diagnosed with classic seminoma, Leydig cell hyperplasia, and intratubular germ cell tumor.
(c) and (d) Testicular tissue section collected from the patient was immunostained for monocyte/macrophages using CD14 as a
marker (brown color; Cell Marque, Rocklin, CA). Cell nuclei were counterstained with hematoxylin (blue color, [c] and [d]). This study
was approved by the Institutional Review Board. An informed consent was obtained from the patient.

prognosis.34,96 However, similar to viral pathogens, if a privilege and inflammation is necessary to prevent infec-
lymphoma metastasizes, the testis can act as a reservoir tions and cancer while maintaining male fertility.
for this tissue and increase risk of relapse. For instance,
the testis is a major site for relapse of acute lymphoblastic ACKNOWLEDGMENTS
leukemia4; however, due to advances in modern medicine, This work was supported in part by NIAID grant AI109398
testicular relapse is now a rare event.97 and funding from The Jasper L. and Jack Denton Wilson
Overall, SCs continuously guide the immune cells Foundation. The authors wish to acknowledge the con-
rather than just constantly suppressing the immune tribution of the Texas Tech University Health Sciences
response. Thus, if a foreign pathogen threatens the exis- Center Clinical Research Institute for their assistance with
tence of the host, SCs abandon their motherly duties and this research.
initiate inflammation, resulting in disruption of immune
privilege and spermatogenesis. For instance, Chlamydia REFERENCES
has been detected in urethral or urine samples from 1. Ramzan MH, Ramzan M, Khan MM et al. Human
11%–35% of men with epididymo-orchitis, and acute semen quality and sperm DNA damage assessed
epididymo-orchitis is associated with decreased sperm by comet assay in clinical groups. Turk J Med Sci.
counts and decreased sperm motility.98 2015;45(3):729–737.
2. Kumar N, Singh AK. Trends of male factor infertil-
SUMMARY ity, an important cause of infertility: A review of lit-
Testis immune privilege is necessary to prevent an auto- erature. J Hum Reprod Sci. 2015;8(4):191–196.
immune response to the developing germ cells. SCs play 3. CDC. 2016. Available from: http://www.cdc.gov​
an important role in maintaining this immune privileged /reproductivehealth/infertility/.
environment by sequestering the majority of the germ cells 4. Hedger MP. The Immunophysiology of Male
behind the BTB/SCB (adluminal compartment), thereby Reproduction. 4 ed. New York: Academic Press;2015:
preventing the immune cells from gaining access to these 805–889.
cells and modifying the immune response by expressing 5. Silva CA, Cocuzza M, Borba EF, Bonfa E. Cutting-
several immunomodulatory factors and inducing regula- edge issues in autoimmune orchitis. Clin Rev Allergy
tory cells. However, a delicate balance between immune Immunol. 2012;42(2):256–263.
90  Sertoli cell immune regulation within the testis

6. O’Rand MG, Romrell LJ. Appearance of cell surface 23. Chung NP, Mruk D, Mo MY, Lee WM, Cheng CY.
auto- and isoantigens during spermatogenesis in the A 22-amino acid synthetic peptide corresponding to
rabbit. Dev Biol. 1977;55(2):347–358. the second extracellular loop of rat occludin perturbs
7. Tung PS, Fritz IB. Specific surface antigens on rat the blood-testis barrier and disrupts spermatogenesis
pachytene spermatocytes and successive classes of reversibly in vivo. Biol Reprod. 2001;65(5):1340–1351.
germinal cells. Dev Biol. 1978;64(2):297–315. 24. Wong CH, Mruk DD, Lee WM, Cheng CY. Targeted
8. Jacobo P, Guazzone VA, Theas MS, Lustig L. Testicular and reversible disruption of the blood-testis bar-
autoimmunity. Autoimmun Rev. 2011;10(4):201–204. rier by an FSH mutant-occludin peptide conjugate.
9. Silva CA, Cocuzza M, Carvalho JF, Bonfa E. FASEB J. 2007;21(2):438–448.
Diagnosis and classification of autoimmune orchitis. 25. Gow A, Southwood CM, Li JS et al. CNS myelin and
Autoimmun Rev. 2014;13(4–5):431–434. Sertoli cell tight junction strands are absent in Osp/
10. Kaur G, Mital P, Dufour JM. Testisimmune claudin-11 null mice. Cell. 1999;99(6):649–659.
privilege—Assumptions versus facts. Anim Reprod. 26. Moroi S, Saitou M, Fujimoto K et al. Occludin is
2013;10(1):3–15. concentrated at tight junctions of mouse/rat but not
11. Mital P, Hinton BT, Dufour JM. The blood-testis and human/guinea pig Sertoli cells in testes. Am J Physiol.
blood-epididymis barriers are more than just their 1998;274(6 Pt 1):C1708–1717.
tight junctions. Biol Reprod. 2011;84(5):851–858. 27. Chiba K, Yamaguchi K, Ando M, Miyake H,
12. Smith LB, O’Shaughnessy PJ, Rebourcet D. Cell- Fujisawa M. Expression pattern of testicular claudin-11
specific ablation in the testis: What have we learned? in infertile men. Urology. 2012;80(5):1161 e13–17.
Andrology. 2015;3(6):1035–1049. 28. Fink C, Weigel R, Fink L et al. Claudin-11 is over-
13. Rebourcet D, O’Shaughnessy PJ, Monteiro A et al. expressed and dislocated from the blood-testis
Sertoli cells maintain Leydig cell number and peri- barrier in Sertoli cells associated with testicular
tubular myoid cell activity in the adult mouse testis. intraepithelial neoplasia in men. Histochem Cell Biol.
PLoS One. 2014;9(8):e105687. 2009;131(6):755–764.
14. Shinomura M, Kishi K, Tomita A et al. A novel Amh- 29. Haverfield JT, Meachem SJ, O’Bryan MK, McLachlan
Treck transgenic mouse line allows toxin-dependent RI, Stanton PG. Claudin-11 and connexin-43 display
loss of supporting cells in gonads. Reproduction. altered spatial patterns of organization in men with
2014;148(6):H1–9. primary seminiferous tubule failure compared with
15. Johnson MH, Setchell BP. Protein and immuno- controls. Fertil Steril. 2013;100(3):658–666.
globulin content of rete testis fluid of rams. J Reprod 30. Nah WH, Lee JE, Park HJ, Park NC, Gye MC.
Fertil. 1968;17(2):403–406. Claudin-11 expression increased in spermato-
16. Johnson MH. The distribution of immunoglobulin genic defect in human testes. Fertil Steril.
and spermatozoal autoantigen in the genital tract of 2011;95(1):385–388.
the male guinea pig: Its relationship to autoallergic 31. Stammler A, Luftner BU, Kliesch S et al. Highly
orchitis. Fertil Steril. 1972;23(6):383–392. conserved testicular localization of claudin-11 in
17. Dym M, Fawcett DW. The blood-testis barrier in the rat normal and impaired spermatogenesis. PLoS One.
and the physiological compartmentation of the semi- 2016;11(8):e0160349.
niferous epithelium. Biol Reprod. 1970;3(3):308–326. 32. Nitta T, Hata M, Gotoh S et al. Size-selective loosen-
18. Dym M, Romrell LJ. Intraepithelial lymphocytes in ing of the blood-brain barrier in claudin-5-deficient
the male reproductive tract of rats and rhesus mon- mice. J Cell Biol. 2003;161(3):653–660.
keys. J Reprod Fertil. 1975;42(1):1–7. 33. Xu J, Anuar F, Ali SM, Ng MY, Phua DC, Hunziker
19. Fawcett DW, Leak LV, Heidger PM, Jr. Electron W. Zona occludens-2 is critical for blood-testis
microscopic observations on the structural com- barrier integrity and male fertility. Mol Biol Cell.
ponents of the blood-testis barrier. J Reprod Fertil 2009;20(20):4268–4277.
Suppl. 1970;10:105–122. 34. Fink C, Weigel R, Hembes T et al. Altered expression
20. Rebourcet D, O’Shaughnessy PJ, Pitetti JL et al. of ZO-1 and ZO-2 in Sertoli cells and loss of blood-
Sertoli cells control peritubular myoid cell fate and testis barrier integrity in testicular carcinoma in situ.
support adult Leydig cell development in the prepu- Neoplasia. 2006;8(12):1019–1027.
bertal testis. Development. 2014;141(10):2139–2149. 35. Shao M, Ghosh A, Cooke VG, Naik UP, Martin-
21. Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli- DeLeon PA. JAM-A is present in mammalian sper-
germ cell interactions and their significance in germ matozoa where it is essential for normal motility.
cell movement in the seminiferous epithelium during Dev Biol. 2008;313(1):246–255.
spermatogenesis. Endocr Rev. 2004;25(5):747–806. 36. Sakaguchi T, Nishimoto M, Miyagi S et al. Putative
22. Saitou M, Furuse M, Sasaki H et al. Complex pheno- “stemness” gene jam-B is not required for maintenance
type of mice lacking occludin, a component of tight of stem cell state in embryonic, neural, or hematopoi-
junction strands. Mol Biol Cell. 2000;11(12):4131–4142. etic stem cells. Mol Cell Biol. 2006;26(17):​6557–6570.
References 91

37. Meng J, Greenlee AR, Taub CJ, Braun RE. Sertoli 53. Haverfield JT, Meachem SJ, Nicholls PK, Rainczuk
cell-specific deletion of the androgen receptor com- KE, Simpson ER, Stanton PG. Differential perme-
promises testicular immune privilege in mice. Biol ability of the blood-testis barrier during reinitiation
Reprod. 2011;85(2):254–260. of spermatogenesis in adult male rats. Endocrinology.
38. Meng J, Holdcraft RW, Shima JE, Griswold MD, 2014;155(3):1131–1144.
Braun RE. Androgens regulate the permeability of 54. Mallidis C, Baker HW. Fine needle tissue aspiration
the blood-testis barrier. Proc Natl Acad Sci U S A. biopsy of the testis. Fertil Steril. 1994;61(2):367–375.
2005;102(46):16696–16700. 55. Itoh M, Xie Q, Miyamoto K, Takeuchi Y. Major dif-
39. Chakraborty P, William Buaas F, Sharma M et al. ferences between the testis and epididymis in the
Androgen-dependent Sertoli cell tight junction induction of granulomas in response to extravasated
remodeling is mediated by multiple tight junction germ cells. I. A light microscopical study in mice. Int
components. Mol Endocrinol. 2014;28(7):1055–1072. J Androl. 1999;22(5):316–323.
40. Johnson MH. An immunological barrier in the 56. Itoh M, Chen XH, Takeuchi Y, Miki T. Morphological
guinea-pig testis. J Pathol. 1970;101(2):129–139. demonstration of the immune privilege in the testis
41. Tung KS, Unanue ER, Dixon FJ. Pathogenesis of using adjuvants: Tissue responses of male repro-
experimental allergic orchitis. II. The role of anti- ductive organs in mice injected with Bordetella
body. J Immunol. 1971;106(6):1463–1472. pertussigens. Arch Histol Cytol. 1995;58(5):575–579.
42. Taguchi O, Nishizuka Y. Experimental autoimmune 57. Honaramooz A, Yang Y. Recent advances in applica-
orchitis after neonatal thymectomy in the mouse. tion of male germ cell transplantation in farm ani-
Clin Exp Immunol. 1981;46(2):425–434. mals. Vet Med Int. 2010;2011.
43. Tung KS, Smith S, Teuscher C, Cook C, Anderson RE. 58. Kaur G, Thompson LA, Dufour JM. Therapeutic
Murine autoimmune oophoritis, epididymoorchitis, potential of immune privileged Sertoli cells. Anim
and gastritis induced by day 3 thymectomy. Immuno­ Reprod. 2015;12:105–117.
pathology. Am J Pathol. 1987;126(2):293–302. 59. Dufour JM, Rajotte RV, Kin T, Korbutt GS.
44. Knee RA, Hickey DK, Beagley KW, Jones RC. Immunoprotection of rat islet xenografts by cotrans-
Transport of IgG across the blood-luminal barrier plantation with Sertoli cells and a single injec-
of the male reproductive tract of the rat and the tion of antilymphocyte serum. Transplantation.
effect of estradiol administration on reabsorption of 2003;75(9):1594–1596.
fluid and IgG by the epididymal ducts. Biol Reprod. 60. Gores PF, Hayes DH, Copeland MJ et al. Long-
2005;73(4):688–694. term survival of intratesticular porcine islets in
45. Itoh M, Terayama H, Naito M, Ogawa Y, Tainosho S. nonimmunosuppressed beagles. Transplantation.
Tissue microcircumstances for leukocytic infiltra- 2003;75(5):613–618.
tion into the testis and epididymis in mice. J Reprod 61. Dufour JM, Hamilton M, Rajotte RV, Korbutt
Immunol. 2005;67(1–2):57–67. GS. Neonatal porcine Sertoli cells inhibit human
46. Naito M, Itoh M. Patterns of infiltration of lym- natural antibody-mediated lysis. Biol Reprod.
phocytes into the testis under normal and patho- 2005;72(5):1224–1231.
logical conditions in mice. Am J Reprod Immunol. 62. Wright K, Dziuk R, Mital P, Kaur G, Dufour JM.
2008;59(1):55–61. Xenotransplanted pig Sertoli cells inhibit both the
47. Dym M, Cavicchia JC. Further observations on alternative and classical pathways of complement
the blood-testis barrier in monkeys. Biol Reprod. mediated cell lysis while pig islets are killed. Cell
1977;17(3):390–403. Transplant. 2016;25(11):2027–2040.
48. Pelletier RM. The tight junctions in the testis, epididy- 63. Bailey R, Griswold MD. Clusterin in the male repro-
mis, and vas deferens. In Cereijido M, Anderson J, eds. ductive system: Localization and possible function.
Tight Junctions. New York: CRC Press; 2001:599–628. Mol Cell Endocrinol. 1999;151(1–2):17–23.
49. Russell L. Movement of spermatocytes from the 64. Doyle TJ, Kaur G, Putrevu SM et al. Immunoprotective
basal to the adluminal compartment of the rat testis. properties of primary Sertoli cells in mice: Potential
Am J Anat. 1977;148(3):313–328. functional pathways that confer immune privilege.
50. Smith BE, Braun RE. Germ cell migra- Biol Reprod. 2012;86(1):1–14.
tion across Sertoli cell tight junctions. Science. 65. Lee HM, Oh BC, Lim DP et al. Role of comple-
2012;338(6108):798–802. ment regulatory proteins in the survival of murine
51. Yule TD, Montoya GD, Russell LD, Williams TM, allo-transplanted Sertoli cells. J Korean Med Sci.
Tung KS. Autoantigenic germ cells exist outside the 2007;22(2):277–282.
blood testis barrier. J Immunol. 1988;141(4):1161–1167. 66. Mead R, Hinchliffe SJ, Morgan BP. Molecular clon-
52. Pelletier RM. Cyclic formation and decay of the ing, expression and characterization of the rat ana-
blood-testis barrier in the mink (Mustela vison), logue of human membrane cofactor protein (MCP/
a seasonal breeder. Am J Anat. 1986;175(1):91–117. CD46). Immunology. 1999;98(1):137–143.
92  Sertoli cell immune regulation within the testis

67. Hedger MP. The Immunophysiology of Male 80. Chui K, Trivedi A, Cheng CY et al. Characterization
Reproduction. New York: Academic Press; and functionality of proliferative human Sertoli
2015:805–889. cells. Cell Transplant. 2011;20(5):619–635.
68. Nikolova DB, Kancheva LS, Surneva MD, Martinova 81. Winnall WR, Hedger MP. Phenotypic and functional
YS. Species-specific effect of proteins secreted by heterogeneity of the testicular macrophage popula-
cultured pre-pubertal rat Sertoli cells on natu- tion: A new regulatory model. J Reprod Immunol.
ral killer cell activity. Immunopharmacology. 2013;97(2):147–158.
1992;23(1):15–20. 82. Guazzone VA, Jacobo P, Theas MS, Lustig L. Cytokines
69. Sipione S, Simmen KC, Lord SJ et al. Identification of a and chemokines in testicular inflammation: A brief
novel human granzyme B inhibitor secreted by cultured review. Microsc Res Tech. 2009;72(8):620–628.
Sertoli cells. J Immunol. 2006;177(8):5051–5058. 83. Gao J, Wang X, Wang Y et al. Murine Sertoli cells
70. Bladergroen BA, Strik MC, Bovenschen N et al. promote the development of tolerogenic dendritic
The granzyme B inhibitor, protease inhibitor 9, is cells: A pivotal role of galectin-1. Immunology.
mainly expressed by dendritic cells and at immune- 2016;148(3):253–265.
privileged sites. J Immunol. 2001;166(5):3218–3225. 84. Dal Secco V, Riccioli A, Padula F, Ziparo E,
71. Hirst CE, Buzza MS, Sutton VR, Trapani JA, Filippini A. Mouse Sertoli cells display pheno-
Loveland KL, Bird PI. Perforin-independent expres- typical and functional traits of antigen-presenting
sion of granzyme B and proteinase inhibitor 9 in cells in response to interferon gamma. Biol Reprod.
human testis and placenta suggests a role for gran- 2008;78(2):234–242.
zyme B-mediated proteolysis in reproduction. Mol 85. Campese AF, Grazioli P, de Cesaris P et al.
Hum Reprod. 2001;7(12):1133–1142. Mouse Sertoli cells sustain de novo generation of
72. Mital P, Kaur G, Dufour JM. Immunoprotective Sertoli regulatory T cells by triggering the notch path-
cells: Making allogeneic and xenogeneic transplanta- way through soluble JAGGED1. Biol Reprod.
tion feasible. Reproduction. 2010;139(3):​495–504. 2014;90(3):53.
73. De Cesaris P, Filippini A, Cervelli C et al. 86. Jenabian MA, Costiniuk CT, Mehraj V et al. Immune
Immunosuppressive molecules produced by Sertoli tolerance properties of the testicular tissue as a viral
cells cultured in vitro: Biological effects on lympho- sanctuary site in ART-treated HIV-infected adults.
cytes. Biochem Biophys Res Commun. 1992;186(3):​ AIDS. 2016;30(18):2777–2786.
1639–1646. 87. Deen GF, Knust B, Broutet N et al. Ebola RNA per-
74. Selawry HP, Kotb M, Herrod HG, Lu ZN. sistence in semen of Ebola virus disease survivors—
Production of a factor, or factors, suppressing IL-2 Preliminary report. N Engl J Med. 2015.
production and T cell proliferation by Sertoli cell- 88. Soka MJ, Choi MJ, Baller A et al. Prevention of
enriched preparations. A potential role for islet sexual transmission of Ebola in Liberia through a
transplantation in an immunologically privileged national semen testing and counselling programme
site. Transplantation. 1991;52(5):846–850. for survivors: An analysis of Ebola virus RNA
75. Wyatt CR, Law L, Magnuson JA, Griswold MD, results and behavioural data. Lancet Glob Health.
Magnuson NS. Suppression of lymphocyte prolif- 2016;4(10):e736–743.
eration by proteins secreted by cultured Sertoli cells. 89. Hills SL, Russell K, Hennessey M et al. Transmission
J Reprod Immunol. 1988;14(1):27–40. of Zika virus through sexual contact with travel-
76. Cupp AS, Kim G, Skinner MK. Expression ers to areas of ongoing transmission—Continental
and action of transforming growth factor beta United States, 2016. MMWR Morb Mortal Wkly Rep.
(TGFbeta1, TGFbeta2, and TGFbeta3) during 2016;65(8):215–216.
embryonic rat testis development. Biol Reprod. 90. Krieger JN. Epididymitis, orchitis, and related condi-
1999;60(6):1304–1313. tions. Sex Transm Dis. 1984;11(3):173–181.
77. Fallarino F, Luca G, Calvitti M et al. Therapy of experi- 91. Trojian TH, Lishnak TS, Heiman D. Epididymitis
mental type 1 diabetes by isolated Sertoli cell xenografts and orchitis: An overview. Am Fam Physician.
alone. J Exp Med. 2009;206(11):2511–2526. 2009;79(7):583–587.
78. Dettin L, Rubinstein N, Aoki A, Rabinovich GA, 92. Ehrlich Y, Margel D, Lubin MA, Baniel J. Advances
Maldonado CA. Regulated expression and ultra- in the treatment of testicular cancer. Transl Androl
structural localization of galectin-1, a proapoptotic Urol. 2015;4(3):381–390.
beta-galactoside-binding lectin, during spermato- 93. Roach JC, Glusman G, Rowen L et al. The evolution
genesis in rat testis. Biol Reprod. 2003;68(1):51–59. of vertebrate Toll-like receptors. Proc Natl Acad Sci
79. Wollina U, Schreiber G, Gornig M, Feldrappe S, U S A. 2005;102(27):9577–9582.
Burchert M, Gabius HJ. Sertoli cell expression of 94. Hedger MP. Toll-like receptors and signalling
galectin-1 and -3 and accessible binding sites in nor- in spermatogenesis and testicular responses to
mal human testis and Sertoli cell only-syndrome. inflammation—A perspective. J Reprod Immunol.
Histol Histopathol. 1999;14(3):779–784. 2011;88(2):130–141.
References 93

95. Meinhardt A, Hedger MP. Immunological, paracrine 97. Kulkarni KP, Marwaha RK, Trehan A, Bansal D.
and endocrine aspects of testicular immune privi- Testicular relapse in childhood acute lymphoblas-
lege. Mol Cell Endocrinol. 2011;335(1):60–68. tic leukemia: The challenges and lessons. Indian J
96. Hvarness T, Nielsen JE, Almstrup K, Skakkebaek Cancer. 2010;47(2):134–138.
NE, Rajpert-De Meyts E, Claesson MH. Phenotypic 98. Cunningham KA, Beagley KW. Male genital tract
characterisation of immune cell infiltrates in tes- chlamydial infection: Implications for pathology
ticular germ cell neoplasia. J Reprod Immunol. and infertility. Biol Reprod. 2008;79(2):180–189.
2013;100(2):135–145.
The mitotic phase of spermatogenesis
Recent advances and perspectives
7
KIN LAM FOK and HSIAO CHANG CHAN

INTRODUCTION puzzle from the whole picture. In this chapter, we will


In healthy men, millions of sperm are produced daily in reveal the key concepts and recent findings with a par-
the testis through a process known as spermatogenesis, ticular focus on the emerging regulators of the mitotic
owing to the tremendous replenishing power of sper- phase of spermatogenesis.
matogonia, a heterogeneous population consisting of both
spermatogonial stem cells (SSCs) and committed progeni- ORIGIN AND DIFFERENT SUBPOPULATIONS
tors.1 The spermatogonia need to undergo three phases— OF SPERMATOGONIA
the mitotic phase, the meiotic phase, and the postmeiotic Spermatogonia are originated from gonocytes, also
differentiation phase—to give rise to spermatozoa. In the known as pre- or prospermatogonia. Gonocytes are
mitotic phase, spermatogonia undergo a series of mitosis to positioned within the lumen of the seminiferous tubules
expand the number of daughter cells. These daughter cells before birth. Shortly after birth, they proliferate, dif-
have two fates: (1) self-renewal to maintain the stem cell ferentiate, and migrate from the lumen to the basement
pool, and (2) committed to differentiation and give rise to membrane of the seminiferous tubules where a specialized
primary spermatocytes that enter the meiotic phase. Since microenvironment or niche is formed.11,12 Gonocytes give
the primary spermatocytes will only undergo two rounds rise to two spermatogonial populations, the undifferenti-
of meiotic division whereas the spermatids undergo dif- ated spermatogonia and differentiating spermatogonia.
ferentiation without division, the mitotic phase is consid- The differentiating spermatogonia proceed to the meiotic
ered the foundation that provides a steady supply of a large process and migrate from the basement membrane to the
number of committed progenies while maintaining the adluminal compartment to initiate the first wave of sper-
stem cell pool to sustain spermatogenesis. Perturbation of matogenesis in the juvenile while the undifferentiated
the homeostasis between spermatogonial self-renewal and spermatogonia self-renew and support the steady state of
differentiation has detrimental effects on spermatogenesis. spermatogenesis in the adult.13 In the mouse, the undif-
In the past two decades, a number of valuable tools for ferentiated spermatogonia consist of type A spermatogo-
the study of spermatogonia have been developed. These nia (Aundiff ) that can be further classified into Asingle (As),
include the germ cell transplantation technique that Apaired (Apr), and Aaligned (Aal) spermatogonia while the
allows the assessment of stem cell activity by observ- differentiating spermatogonia are made up of type A1–4,
ing the regeneration of spermatogenesis, 2 the in vitro intermediate, and type B spermatogonia. The mitotic divi-
propagation of spermatogonia that facilitates functional sion of spermatogonia undergoes distinctive incomplete
studies by pharmacological and genetic manipulations, 3,4 cytokinesis where the daughter cells were interconnected
and the fluorescent protein-mediated lineage tracing in with the intercellular cytoplasmic bridge, also known as
transgenic mice models that monitors the migration and the syncytial state.14 The first mitotic division of As sper-
dynamics of live cells in real time.5 In particular, the in matogonia will give rise to Apr spermatogonia. Subsequent
vitro propagation of spermatogonia has recapitulated rounds of mitosis will give rise to Aal spermatogonia of
the mitosis phase of spermatogenesis. In the presence 4–32 cells in length. The spermatogonial stem cells (SSCs)
of a cocktail of growth factors and supporting feeder mainly reside in the As spermatogonia. Interestingly, the
cells or extracellular matrix proteins, primary culture syncytial state spermatogonia undergo stochastic frag-
of spermatogonia can be maintained and expanded in mentation and give rise to As spermatogonia, indicating
vitro. The cultured spermatogonia remain heterogeneous that the hierarchy of Aundiff is reversible and stochastic14
and the stem cell activity is maintained in a subpopu- (Figure 7.1).
lation of cells. Using these valuable tools, a plethora of A number of markers have been identified for different
critical regulators on the self-renewal and differentia- spermatogonial populations. GFRα1 is mainly expressed
tion of spermatogonia has been identified.6–10 However, in As spermatogonia and its expression is decreased grad-
the mitotic phase of spermatogenesis is peculiarly com- ually in Apr and Aal spermatogonia. In contrast, Ngn3 is
plicated, involving the regulation by extrinsic factors weakly expressed in or absent from As spermatogonia
such as paracrine signaling and cell-niche interaction, while its expression increases in Apr and Aal spermatogo-
intrinsic factors such as specific genes and proteins, and nia.15 Expression of c-Kit is only observed in Aal and differ-
the interplay between the two. Nevertheless, the current entiating spermatogonia.16 Thus far, a specific marker for
knowledge appears to represent only a few pieces of the SSC has not been identified. Nonetheless, the expression

94
The niche of spermatogonia  95

Differentiated
c-KIT positive

As Apr Aal–4 Aal (8–32 cells)


Undifferencitated

PAX7 Aal (3, 5 or 6 cells)


ID4
GFRα1
NGN3
PLZF

Complete mitosis Incomplete mitosis Syncytial fragmentation

Figure 7.1  Spermatogonial subpopulations in the mitotic phase of mouse spermatogenesis. Schematic diagram showing the
interconversion between As,Apr, and Aal spermatogonia through cell division and syncytial fragmentation among the Aundiff popula-
tion (bottom panel). These subpopulations differentiate to form the differentiated spermatogonia (upper panel). The corresponding
marker genes expression among the Aundiff is also shown in the bottom panel.

of Id4 or Pax7 marks the majority of SSCs that reside in As macrophages,24 and probably the neighboring spermatogo-
spermatogonia17,18 (Figure 7.1), and the stem cell capacity nia also contribute to the niche through paracrine signaling25
gradually decreases as spermatogonia advance from As to (see section “Autocrine/paracrine regulation of spermatogo-
Aal stages.19,20 While these markers are useful for labeling nia”) (Figure 7.2). Moreover, by chasing the location of Aundiff
different spermatogonial subpopulations, the functional using time-lapse imaging in vivo, it was found that Aundiff
activity of SSCs is defined by the ability to colonize the is biasedly localized to the vascular network. Alteration in
seminiferous tubules and regenerate spermatogenesis vasculature leads to the rearrangement of Aundiff,5 suggest-
after transplantation.21,22 ing that the vasculature pattern may act as a backbone of the
spermatogonial niche in the seminiferous tubules.
THE NICHE OF SPERMATOGONIA Apart from the vasculature niche in the convoluted
Spermatogonia reside in the basal compartment of the seminiferous tubules, a recent study has identified a sec-
seminiferous tubules (i.e., the compartment between ond niche of GFRα1+ Aundiff at the tubuli rectus26 (Figure
the tight junctions of neighboring Sertoli cells, also 7.2), a straight seminiferous tubule that leads the convo-
known as the blood-testis barrier (BTB), and the base- luted seminiferous tubule to the rete testis. This region
ment membrane of the seminiferous tubules.1 Within the comprises a valvelike structure set up by a group of modi-
basal compartment, the Aundiff preferentially localized to fied Sertoli cells (also known as Sertoli valve) that retained
a specialized niche that plays essential roles in modulat- proliferative capacity in adult life as well as a stable number
ing the self-renewal, differentiation, and migration. Aundiff of GFRα1+ Aundiff but not more differentiated germ cells.
migrates to their niche during the establishment from Since proliferating cells often show differential expression
gonocyte and leaves it when they differentiate.5 In a physi- of tight junction molecules compared to nonproliferating
ological condition, the BTB is formed after Aundiff arrives at cells,27 the question as to whether the BTB is functional
the basement membrane due to the immaturity of Sertoli in the Sertoli valve niche remains elusive. This region also
cells in neonatal testis.23 expresses a higher level of the glial-derived neurotrophic
The spermatogonial niche comprises of various cell factor (GDNF), a Sertoli cell-secreted factor essential for
types and extracellular matrix (ECM) proteins. The Sertoli the self-renewal and maintenance of SSCs, compared to
cells and the ECM of the basement membrane are in clos- that in the convoluted seminiferous tubules.26 Therefore,
est proximity with Aundiff in the niche. Aundiff directly inter- the Sertoli valve niche is postulated to serve as a reserve
act with the ECM through cell adhesion molecules. Apart pool of SSCs in the testis. However, the stem cell activity in
from the Sertoli cells, other somatic cells, including the the GFRα1+ Aundiff population of this niche awaits further
Leydig cells, peritubular myoid cells, immune cells such as investigation.
96  The mitotic phase of spermatogenesis

Sertoli valve niche


Nondividing Proliferating
Sertoli cell Sertoli cell Testis

TJs ? GDNF Seminiferous


tubules Rete
As Apr testis
Basement membrane
Blood
Myoid cells stream
GDNF-
Leydig cells positive

Vasculature niche
Nondividing Spermatocytes
Sertoli cell
Epididymis
Nondividing
As TJs Sertoli cell
Apr Aal

Basement membrane
Myoid cells

Leydig cells Blood


vessel
Macrophage

Figure 7.2  Spermatogonial niches in mouse testis. Schematic diagram showing the localization of the Sertoli valve niche near the
rete testis and the vasculature niche at the seminiferous tubules (right panel). The somatic cells within the Sertoli-valve niche include
Sertoli cells (both proliferating and nonproliferating), Leydig cells, and peritubular myoid cells whereas the As and Apr spermatogo-
nia are the only germ cells in this niche (upper left panel). Compared to the vasculature niche, the Sertoli-valve niche expresses a
higher level of GDNF and the tight junctions (TJs) may be absent (as indicated by ? in the upper left panel) due to the proliferation of
Sertoli cells. The Sertoli cells in the vasculature niche are nonproliferative and form the blood-testis barrier with neighbor Sertoli cells
through TJs. Blood vessel and macrophage also contribute to the vasculature niche (bottom left panel). Differentiated spermatogo-
nia (denoted in purple) are also found in the vasculature niche but not in the Sertoli valve niche.

INTERACTION BETWEEN SPERMATOGONIA interact with laminin, β1-integrin is expressed in Aundiff


AND THE NICHE and antibody for β1-integrin has been used for the enrich-
Differentiating spermatogonia are not found at the sper- ment of SSCs.28 β1-integrin plays an indispensable role in
matogonial niche,5 suggesting that spermatogonial differ- the homing of SSCs. Gene knockout or neutralizing anti-
entiation may be associated with cell migration to allow body treatment that deplete functional β1-integrin in pri-
the dissociation of committed progenies from the niche. mary spermatogonial culture decreases the attachment to
On the other hand, a minority of As spermatogonia may laminin and reduces the homing of SSCs after transplanta-
be formed by syncytial fragmentation of Aal spermatogo- tion.29 A subsequent study has demonstrated that Rac1, a
nia.14 These As spermatogonia need to be associated with downstream target of integrin receptor, is required for the
the niche in order to receive support for self-renewal. migration of SSCs across the BTB through regulating the
These studies highlighted the importance of the dynamic level of cell adhesion molecule claudins.23 Intriguingly,
interaction between spermatogonia and the niche. the Rac1-mediated migration is not required for homing of
Similarly, after transplantation of SSCs into the seminifer- transplanted SSCs in neonatal testis where BTB is absent,23
ous tubules, the transplanted cells need to migrate across suggesting the migration during gonocyte to spermatogo-
the BTB and reach the niche in order to regenerate sper- nia transition may be regulated by an independent pathway.
matogenesis. The migration of SSCs to the spermatogonial Apart from the cell adhesion molecules, the differentiat-
niche is described as homing. Although homing of trans- ing spermatogonia also express a range of matrix metallo-
planted SSCs does not occur in physiological condition, proteinases (MMP) that are capable of degrading different
this model provides a unique opportunity for the study of ECM proteins.30,31 The expression of MMP-2, 7, 9, 23, and
spermatogonial migration. CD147, a recognized regulator of MMPs activity and cell
Similar to the migration process in other cell types, the survival, increase along with spermatogonial develop-
migration of spermatogonia involves the modulation of ment in neonatal mice. Depleting CD147 in a differentiat-
cell adhesion and the interaction with ECM (Figure 7.3). ing spermatogonia cell line GC1-spg inhibits the activity
An important ECM protein in the spermatogonial niche of MMP2 and suppresses cell migration.30 Intriguingly,
is laminin. It is abundantly expressed at the basement in contrast to its recognized function in the survival of
membrane of the seminiferous tubules and supports the GC2-spd spermatocyte cell line through interaction with
in vitro propagation of spermatogonia in the absence of its partner TRAF2 that modulates the NFkB signaling,32,33
a feeder layer.4 Among the cell adhesion molecules that CD147 is dispensable for the maintenance of GC1-spg due
Autocrine/paracrine regulation of spermatogonia  97

Self-renewal and maintenance Differentiation


Sertoli cell Spermatocyte Sertoli cell Spermatocyte

TJs TJs
GDNF
FGF2
GFR 1, KITL RA
c-Ret FGFR1 As BMP4 RARs
Wnt5a c-KIT
As Nodal FGF8
Apr Aal Vitamin
CSF-1
Laminin-containing basement membrane Laminin-containing basement membrane

Blood Blood
Leydig cells
Leydig vessel vessel
cells Macrophage Macrophage

Homing and interaction with the niche

Sertoli cell Spermatocyte

CXCL12
CXCR4 TJs
Apr CD147 Aal
As
MMP2
1-integrin
Laminin-containing basement membrane

Leydig cells Blood


vessel
Macrophage

Figure 7.3  Factors involved in different biological processes in the mitotic phase of spermatogenesis. Schematic diagram
showing the factors (highlighted in red) involved in the homing of SSC and the interaction between the niche and different sper-
matogonial population (bottom panel), the self-renewal of SSC and the maintenance of spermatogonia (upper left panel), and the
spermatogonial differentiation (upper right panel). Aundiff (c-Kit–) is denoted in green whereas differentiating spermatognia (c-Kit+)
and primary spermatocyte are denoted in purple.

to the absence of TRAF2 expression.32,33 This suggests a differentiation, and migration of spermatogonia in a para-
primary role of CD147 in spermatogonial migration but crine manner35 (Figure 7.3). Two best-characterized growth
not cell survival. factors required for self-renewal are GDNF and fibroblast
A recent study comparing the transcriptome of Aundiff growth factor 2 (FGF2, also known as bFGF) secreted by
and differentiating spermatogonia during spermatogonial the Sertoli cells. FGF2 may bind to the four known recep-
development has shed light on the molecular mechanism tors FGFR1-4 expressed in spermatogonia,36 whereas
underlying the migration of spermatogonia. The results GDNF binds to the GFRα1-c-Ret coreceptor on sper-
showed that c-Kit+ differentiating spermatogonia express matogonia. Both factors promote the self-renewal of SSC
a higher level of genes involved in epithelial-mesenchymal independently by the activation of AKT and MAP2K1/2
transition (EMT), a process well-established to be involved pathway.37 FGF2 is also expressed by the Leydig cells and
in cell migration compared to that of Id4+ As spermatogo- differentiating germ cells that may contribute to the para-
nia.34 Moreover, genes involved in cell adhesion and ECM crine regulation in vivo.37 Activation of signaling pathways
demonstrated dynamic methylation pattern in As versus by GDNF and FGF2 induces the expression of transcrip-
that in differentiating spermatogonia.34 These results sug- tion factors, Plzf, Bcl6b, and Etv5, that play critical roles
gest that the differentiating spermatogonia possess higher in SSC self-renewal.38,39 The Leydig cells also contribute to
migration ability that may be attributed to the differential the paracrine regulation of spermatogonial self-renewal by
interaction with ECM of the niche. secreting colony-stimulating factor 1 (CSF-1) that stimu-
lates the self-renewal of SSC with modest effect on overall
AUTOCRINE/PARACRINE REGULATION proliferation of spermatogonia.40
OF SPERMATOGONIA Recent studies have demonstrated that a pan-Wnt
The niche where spermatogonia reside contains cyto- antagonist significantly reduces the SSC activity in cul-
kines and growth factors that regulate the self-renewal, ture, suggesting the involvement of Wnt signaling cascade
98  The mitotic phase of spermatogenesis

in modulating SSC self-renewal.41 Subsequent studies have with RA and exacerbates the spermatogonial differen-
shown that Wnt5a directly promotes SSC self-renewal tiation.60 Moreover, it impairs the maintenance of SSC in
through β-catenin-independent pathway whereas Wnt3a primary spermatogonial culture.61 Interestingly, BMP4 is
indirectly modulates SSC self-renewal by promoting the also expressed in spermatogonia in adult testis,59 suggest-
expansion of committed progenitors through β-catenin- ing the potential involvement of spermatogonial cells in
dependent pathway.42 Wnt6 is another Wnt ligand uniquely paracrine regulation.
expressed in Sertoli cells that may contribute to the prolif- Paracrine factors may have dual roles. For example,
eration of Aundiff.43 Apart from the positive regulation on apart from its well-characterized effect on the self-renewal
Aundiff, Wnt4 secreted by the Sertoli cells induces cell death of SSCs, GDNF also regulates SSC homing by acting as a
and decreases the stem cell activity of cultured spermato- chemotactic factor.62 Treatment of GDNF in cultured sper-
gonia.44 These results suggest that the Sertoli-cell-secreted matogonia increases their migration beneath the Sertoli
Wnt ligands are pivotal paracrine signals that differen- cells through the BTB. Expression of a dominant nega-
tially orchestrate the maintenance of Aundiff. tive GDNF receptor reduces the colonization of SSCs after
Paracrine signaling in the niche also regulates sper- transplantation.62 GDNF also increases the expression of
matogonial differentiation. Retinoic acid (RA), an active chemokine receptor type 4 (CXCR4), which binds to the
metabolite of vitamin A, is a well-established spermatogo- Sertoli-cell-secreted chemokine (C-X-C motif) ligand 12
nial differentiation inducer.45,46 In the testis, the biogenesis (CXCL12). CXCL12 treatment increases the migration of
and catabolism of RA are regulated by the expression of cultured spermatogonia and enhances the colonization
CYP26B1 in Sertoli cells and RALDH2 in germ cells.47 RA of transplanted SSCs.62–64 Overexpression of CXCL12 in
generated by the Sertoli cells binds to the nuclear RA recep- Sertoli cells in vivo also enhances the homing of SSCs.
tors (RARs) in spermatogonia and transcriptionally acti- These results suggest the involvement of paracrine signal-
vates or represses gene expression through direct binding ing in regulating the migration of spermatogonia.
to the RA response element (RARE). Thus far, three RARs In contrast with the paracrine growth factors, little is
(RARα, RARβ, and RARγ) have been identified. Knockout known about the autocrine regulation in spermatogo-
of either one of these RARs disrupts spermatogenesis at nia. In 2009, He et al. identified Nodal, a member of the
different stages, indicating their importance in spermato- transforming growth factor-beta superfamily, as the first
genesis.48–50 RA treatment in cultured spermatogonia or in autocrine factor that promotes the proliferation of sper-
mice in vivo both upregulates the expression of transcrip- matogonial stem/progenitor cells.65 A recent study has
tion factors Stra8 and Sall4 that are implicated in sper- shown that FGF8 is another autocrine factor that acts
matogonial differentiation.51–53 RA also downregulates through FGFR1 to elicit signaling required for the main-
Oct4 and Plzf, the transcription factors that are involved tenance of Aundiff.66 FGF8 is expressed in spermatogenic
in spermatogonial self-renewal.45 Recent studies have cells in a seminiferous tubules cycle-dependent manner
shown that ectopic overexpression of RARγ in spermato- while its receptor, FGFR1, is expressed in Aundiff and Sertoli
gonia primes spermatogonial differentiation,20 whereas cells. Overexpression of FGF8 leads to the accumulation
inactivation of RA signaling by expression of a dominant of Aundiff. Knockout of both FGF8 or FGFR1 reduces the
negative RAR abolishes spermatogonial differentiation.54 number of Aundiff. More importantly, conditional knock-
Our recent study has also shown that in a spermatogonial out of FGFR1 in Aundiff blunted the accumulation of Aundiff
cell line C18-4,55 RARα, but not RARβ and RARγ, tran- induced by FGF8 overexpression, suggesting that the
scriptionally activates a membrane channel protein CFTR action of FGF8-FGFR1 signaling is cell-autonomous.66
(our unpublished data), which has been recently shown These studies suggest that spermatogonia are also sub-
to be involved in regulating the differentiation of embry- jected to autocrine regulation.
onic stem cells.56 These results suggest differential roles of Recently, emerging players in the autocrine/­paracrine
RARs in mediating the effect of RA through both direct regulation of spermatogonia have been identified. Sahin
and indirect activation of spermatogonial differentiation et  al. have established a spermatogenesis ­ regeneration
regulators. model by first depleting spermatogenic cells except Aundiff
Besides the biogenesis of RA, the Sertoli cells also secrete using irradiation followed by the induction of spermatogo-
the stem cell factor (also known as c-kit ligand KITL) at nial differentiation by administration of ­gonadotrophin-​
the basal membrane at a particular stage of the cycle of the releasing hormone agonist. Using this model, they observed
seminiferous epithelium when it is known to interact with that the Desert Hedgehog ligand Dhh, the Hedgehog recep-
differentiating type A spermatogonia.57 The stem cell fac- tor Ptc 2, and the receptor downstream signaling mole-
tor is required for the differentiation of Aundiff and mainte- cules Gli1 and Gli2 are expressed in Aundiff, suggesting the
nance of differentiating spermatogonia by binding to the involvement of Hedgehog signaling in the autocrine regu-
c-Kit receptor expressed on the differentiating spermato- lation of spermatogonia.67 Apart from the involvement of
gonia.58 The expression of c-Kit per se is also under para- cytokines and growth factors, a recent study has suggested
crine regulation from Sertoli cells. Bone morphogenetic the involvement of purinergic signaling in the a­ utocrine/
protein 4 (BMP4) secreted by the Sertoli cells in neonatal paracrine regulation of spermatogonia.68 Purinergic sig-
testis exerts a mitogenic effect and induces c-Kit expres- naling is mediated by the binding of extracellular nucle-
sion in cultured spermatogonia.59 It also acts cooperatively otide or nucleoside such as ATP to the P2 receptor. Two
Ubiquitin proteasome system in the regulation of spermatogonia  99

subclasses of P2 receptors are metabotropic P2Y receptors of proteasome activity during gonocyte differentiation.76
(P2YRs) and ionotropic P2X receptors (P2XRs) that acti- Gene array analysis comparing the expression profile of
vate G-protein–coupled signaling and nucleotide-gated ion UPS genes in gonocytes and spermatogonia isolated from
channels, respectively.69 By employing electrophysiological neonatal testis has identified 205 UPS genes. Among them,
techniques, Flect et al. have shown that spermatogonia are 28 genes showed differential expression in gonocyte ver-
sensitive to a board concentration range of extracellular sus spermatogonia. Intriguingly, genes encoding ubiqui-
ATP. They have further demonstrated two modes of ATP tinating enzymes have been demonstrated to have higher
response: high-affinity (10-µM extracellular ATP) and low expression in the gonocytes whereas those encoding for
affinity (>300-µM extracellular ATP), which are mediated deubiquitinating enzymes have higher expression in sper-
by P2X4 and P2X7 receptors, respectively.69 Further study matogonia,76 suggesting the involvement of UPS in the
is required to demonstrate the physiological role of puri- establishment of spermatogonia.
nergic signaling in the autocrine/paracrine regulation in In the mitotic division of spermatogonia, it has been
spermatogonia. proposed that both symmetric and asymmetric division
occurs. Symmetric division allows the stem cell to become
UBIQUITIN PROTEASOME SYSTEM IN THE REGULATION two stem cells or two committed progenies while asym-
OF SPERMATOGONIA metric division allows the stem cell to give rise to a stem cell
Stem cell homoeostasis often requires a precise orches- and a committed progeny.9 The asymmetric segregation of
tration of protein networks. Therefore, it is expected that cell fate determinant could be one plausible mechanism
the ubiquitin-proteasome system (UPS), which is capable for determining the identity of progenies.9 The deubiq-
of regulating protein turnover and activity, could play uitinating enzyme UCH-L1 is expressed in spermatogo-
important roles in this process. The UPS begins with nia and asymmetrically distributed to the two progenies
ubiquitination, a process where ubiquitin, an evolution- in the asymmetric division of spermatogonia. A higher
ary conserved small peptide, is covalently conjugated to level of UCH-L1 is observed in Plzf+ Aundiff spermatogonia
the target proteins. This process includes three different whereas c-Kit+ differentiating spermatogonia inherit low/
enzymes: E1 activating enzyme, E2 conjugating enzyme, undetectable level of UCH-L1.77 This result suggests that
and E3 ubiquitin ligase. The ubiquitin is first activated by UCH-L1 may be involved in regulating the self-renewal
the E1 enzyme at the cost of ATP. The activated ubiquitin and differentiation of spermatogonia.
is then transferred to the E2 enzyme, and in the presence The functional role of UPS in the mitotic phase of sper-
of a E3 ligase the ubiquitin will be conjugated to target matogenesis can be highlighted by the E3 ligases involved
protein in a substrate-specific manner.70,71 Ubiquitination in Skp1-Cullin-F-box (SCF) complex. The SCF complex
can be a single-round or multiple-round event that leads to is one of the major ubiquitin ligases complex required for
monoubiquitination or polyubiquitination, respectively. cell cycle progression.78 The F-box protein component of
In a polyubiquitination event, the incoming or distal ubiq- the SCF complex is an E3 ligase that binds to the complex
uitin is piled up to the previous or proximal ubiquitin at and controls the substrate recognition. Various F-box pro-
one of the seven lysine residues (K) on the ubiquitin pep- teins can bind to the SCF complex, and this variability
tide. Polyubiquitination of K48 chain is often recognized allows the SCF complex to target a range of substrates.79
by the proteasome that leads to the degradation of the Skp2 is an F-box protein required for S phase progression
target protein. Other types of ubiquitination can be rec- by target degradation of cyclin-dependent kinase inhibitor
ognized as posttranslation modifications that regulate the p27.80 A study has shown that knockout of Skp2 reduces
localization and activity of target protein.72 Ubiquitination fertility, which is associated with the progressive loss of
can be reversed by deubiquitinating enzymes (DUBs) spermatogonia and an increase in apoptosis. More impor-
that provide a path for recycling of ubiquitin after deg- tantly, double knockout of Skp2 and p27 partially restored
radation or for modification of ubiquitin chain on target the number of germ cells and the fertility outcome, sug-
proteins.73 Emerging evidence has suggested the involve- gesting that Skp2 is required for the maintenance of sper-
ment of functionally versatile UPS in the mitotic phase of matogonia by targeting p27.81
spermatogenesis.74 β-TrCP is another well characterized E3 ligase of the SCF
The requirement of protein degradation or UPS in the complex that targets a number of the cell cycle and apopto-
regulation of spermatogonia came from two studies that sis regulators. There are two paralogs: β-TrCP1 (also known
employed Rattus and Drosophila as model systems. The as BTRC) and β-TrCP2 (also known as Fbxw11). The two
study in Drosophila has used an unbiased large-scale paralogs possess indistinguishable biochemical properties
RNAi screening approach to identify genes involved in but are varied in their expression. β-TrCP1 demonstrated a
male germline stem cells maintenance and differentia- high expression in the spermatocytes and a median expres-
tion. Complex analysis of the screening result has revealed sion in the spermatogonia. In contrast, β-TrCP2 showed a
the genes involved in protein synthesis and degradation,75 median expression in spermatogonia and low expression in
suggesting the involvement of UPS in the process. The the spermatocytes.82 Of note, the two paralogs showed non-
study in Rattus has shown that the RA-induced differen- redundant roles in the testis. Knockout of β-TrCP1 leads
tiation in gonocytes can be blocked by proteasome inhibi- to impairment in meiotic progression and male subfertil-
tors lactacystin or bortezomib, indicating the requirement ity.83 Intriguingly, loss of total β-TrCP activity by inducible
100  The mitotic phase of spermatogenesis

knockdown of β-TrCP2 in β-TrCP1 knockout mice leads expression, lncRNAs play versatile roles in a range of cellu-
to an ectopic localization of spermatogonia associated with lar processes. lncRNAs can act as baits to attract the bind-
impaired cell junctions. This phenotype is attributed to ing of transcription factors and impair their interaction
the stabilization of Snail1, a bona fide substrate of β-TrCP with DNA and hence their functions. lncRNAs can also
involved in regulating the transcription of cell adhesion act as decoys for RNA species. The binding of lncRNA to
molecules, since knockdown of Snail1 restored the local- mRNA can regulate their degradation and nuclear export.
ization of spermatogonia and rescued the entire spermato- The binding of lncRNA to miRNA can perturb their bind-
genesis. These results suggest that β-TrCP regulates the cell ing to bona fide mRNA targets and thus inhibit their activ-
adhesion of spermatogonia by targeting Snail1. ity. As well, the binding of lncRNA to genomic DNA has
Recently, another F-box protein FBXW7 has been shown also been shown to recruit epigenetic modifiers that mod-
to be expressed specifically in Aundiff and regulate the self- ulate epigenetic events such as methylation and histone
renewal of SSC. Knockout of FBXW7 leads to accumula- modification. Finally, lncRNA acts as a precursor that after
tion of Aundiff and increases the colonization of transplanted further processing by RNase will give rise to miRNA. This
SSC while its overexpression compromises SSC activity. property provides a reservoir of miRNA without transcrip-
Moreover, loss of FBXW7 is associated with an increase tion.91 Intriguingly, emerging evidence has suggested that
of MYC protein. Double knockdown of MYC and MYCN both the miRNA and lncRNA are involved in regulating
mimics the effect of FBXW7 overexpression and obliterate the mitotic phase of spermatogenesis.
SSC activity.84 These results suggest that FBXW7 negatively Using high throughput sequencing techniques, Niu et al.
regulates SSC self-renewal by targeting MYC. have compared the miRNA expression in Thy1+ and Thy- tes-
Apart from the SCF complex, recent studies showed ticular population in 6-day-old mice as well as in cultured
that E3 ligase HUWE1, which is an established tumor sup- spermatogonia. The Thy1+ population is enriched in Aundiff
pressor known to target p53 and MYCN,85–87 is expressed while the majority of the Thy1– population is comprised of
in both spermatogonia and spermatocytes.88 HUWE1 somatic cells. Analysis of the sequencing results showed
protein isolated from testicular lysate can ubiquitinate that ~50% of the annotated miRNAs in available databases
core histone proteins.89 Knockout of HUWE1 specifi- were expressed in these samples, suggesting that miRNA
cally in neonatal germ cells perturbed the transition from is the predominant short ncRNA species in neonatal mice
gonocytes to spermatogonia (our unpublished data). These testis.92 Further characterization by comparing the miRNA
results suggest that HUWE1 positively regulates the estab- profile in Thy1+ and Thy1– population has identified 139 dif-
lishment and maintenance of spermatogonia. Together, ferentially expressed miRNAs, of which 84 were upregu-
these studies suggest the pivotal roles of UPS in regulating lated and 55 were downregulated. They further identified
the self-renewal and maintenance of spermatogonia. miR-21, an antiapoptotic factor, as one of the differentially
expressed miRNA under direct transcriptional control of
NONCODING RNA IN THE REGULATION the GDNF signaling pathway. Moreover, inhibition of miR-
OF SPERMATOGONIA 21 by antagomir oligonucleotide increases apoptosis and
The previous sections have revealed the importance decreases the SSC number and activity.92 These results sug-
of protein as intrinsic factors in regulating the mitotic gest an important role of miR-21 in SSC self-renewal.
phase of spermatogenesis. However, the involvement of He et al. have compared miRNA expression in GFRα1+
genes/nucleic acid should not be overlooked. Mammalian and GFRα1– population by microarray. They have identi-
genome analyses show that the protein-coding genes fied miR-20 and miR-106a as two miRNAs preferentially
only contribute to 5%–10% of the genome. However, the expressed in GFRα1+ spermatogonia and in spermatogo-
remaining >90% of the genome are not junk DNA. Rather, nial cell line C18-4. Transfection of miRNA mimics for
these regions are actively transcribed into RNA, which is these two miRNAs can each promote the proliferation/
classified as noncoding RNA (ncRNA). Depending on the self-renewal of SSCs as reflected by the increased BrdU
size/nucleotide (nt) length, ncRNA can be further divided uptake, the increased expression of proliferation marker
into long (lncRNA; >200 nt) or short (<200 nt) ncRNA. PCNA and self-renewal marker Plzf, and the increased SSC
Short ncRNA, such as PIWI-interacting RNA (piRNA), activity as measured by transplantation assay. Consistent
plays important roles in silencing retrotransposon and with the gain-of-function model, antagomir against these
safeguarding the germline genome during meiotic pro- two miRNAs can each inhibit SSC numbers and activity.
gression.90 MicroRNA (miRNA) is another class of short Further studies have identified Stat3 and Ccnd1 as the
ncRNA that is well reported to regulate gene expres- downstream targets of miR-20 and 106a that contribute to
sion posttranscriptionally by binding to the untranslated SSC self-renewal.93
region of the mRNA. lncRNAs are expressed from a wide Yang et al. have demonstrated the important roles of
variety of genomic locations. They can be expressed from miR-221 and 222 in maintaining SSC stemness by sup-
both autosomes and sex chromosomes in either sense or pressing the translation of c-Kit. In hematopoietic stem
antisense direction. They can be either intergenic or in cells, protein expression of c-Kit is regulated by miR-221
association with a gene at the promoter, exon, intron, or and 222. Similarly, the expression of miR-221 and 222
the untranslated region. Given their broad spectrum of is observed in c-Kit– spermatogonia and upregulated
Metabolism of spermatogonia  101

by GDNF, FGF2, and CSF-1. On the other hand, RA METABOLISM OF SPERMATOGONIA


decreases the expression of miR-221 and 222 in spermato- Spermatogonia is one of the most active stem cell popu-
gonial culture. More importantly, inhibition of miR-221 lations in adults. In order to sustain their cellular activi-
or 222 decreases SSC activity while overexpression of ties, they require a metabolic machinery that converts
either one perturbs spermatogonial differentiation.94 RA the available nutrients in the microenvironment into
also represses the expression of other miRNAs. Tong et al. a sufficient amount of energy. Therefore, it is expected
showed that the miRNAs from two clusters miR17-92 and that the metabolism affects the maintenance of sper-
its paralog cluster miR-106b-25 are both downregulated matogonia. Intriguingly, recent studies suggest that the
by RA in spermatogonial culture. Intriguingly, knock- metabolism of spermatogonia also affects the fate deci-
out of miR17-92 cluster upregulates the expression of sion. The mammalian target of rapamycin (mTOR), an
miRNAs from the miR-106b-25 cluster and leads to mild atypical serine/threonine kinase, is a master regula-
defects in spermatogenesis.95 Huszar and Payne showed tor of cell growth and metabolism.108 mTOR interacts
that expression of miR-146 is markedly decreased by with different protein components to form two protein
RA. Overexpression of miR-146 inhibits the RA-induced complexes, mTORC1 and mTORC2, that have differ-
expression of spermatogonial differentiation mark- ent upstream input and downstream output.108 Among
ers c-Kit, Stra8, and Sohlh1 while inhibition of miR-146 these two complexes, the activity of mTORC1 is pre-
upregulates them.96 Chen et al. showed that the expression cisely regulated in order to maintain the homoeostasis of
of miR202-3p was elevated by GDNF but decreased by spermatogonial self-renewal and differentiation. Plzf, a
RA. Knockout of miR202-3p decreases SSC activity and transcription factor essential for the self-renewal of sper-
induces differentiation into meiotic cells by inhibiting the matogonia, 39 has been shown to induce the expression of
translation of RNA binding protein Rbfox2.97 Together, Redd1, which suppresses mTORC1 activity. Knockout of
these results suggest that miRNAs regulate both sper- Plzf elevates mTORC1 activity that increases cell growth
matogonial self-renewal and differentiation. but decreases self-renewal.109 A recent study has further
A bulk of nonannotated lncRNAs is differentially shown that elevated mTORC1 activity is observed in
expressed in the hallmark stages of testis develop- spermatogonia primed for differentiation.110 Knockout
ment,98–100  suggesting a hidden layer of molecular of mTORC1 inhibitor Tsc2 in the whole spermatogo-
regulation in spermatogenesis.101 Compared with the nial population promotes differentiation and leads to
spermatocyte and spermatid, the spermatogonia express germline degeneration whereas knockout of Tsc2 in dif-
the largest number of lncRNAs. Microarray study showed ferentiating spermatogonia does not compromise the
that among the 24,316 lncRNAs, 17,354 of them are spermatogonial pool.110 These results suggest that the
expressed in spermatogonial populations.102 Despite the activity of mTORC1 is tightly regulated to maintain
abundant expression in spermatogonia, the functions of the fate decision of spermatogonia.
these lncRNAs in the mitotic phase of spermatogenesis The self-renewal of many types of stem cells relies on
remain largely unknown. Nonetheless, emerging evi- anaerobic glycolysis.111 Similarly, glycolysis also enhances
dence suggests that these lncRNAs play important roles the self-renewal of spermatogonia. The glycolytic activity
in spermatogonia. The meiotic recombination hot spot in spermatogonia is coupled with the level of MYC/MYCN
locus (mrhl) is a lncRNA identified during the analysis protein. Loss of MYC/MYCN diminishes glycolysis and
of meiotic recombination.103 Intriguingly, subsequent limits spermatogonial self-renewal. Chemical induction of
silencing studies in a differentiating spermatogonial cell glycolysis promotes spermatogonial self-renewal induced
line GC1-spg showed that mrhl regulates the expression by growth factors in vitro.112 These results suggest that
of genes involved in cell adhesion and differentiation and spermatogonia use glucose as the major energy source. In
negatively regulates Wnt signaling through interaction fact, Aundiff is preferentially located near the blood stream
with p68/Ddx5 RNA helicase.104,105 Reciprocally, treat- where glucose is readily available.
ment of GC1-spg cells with Wnt3a downregulates the Cellular metabolism produces reactive oxygen species
expression of mrhl,106 suggesting a negative feedback loop (ROS) as a byproduct of energy production. Emerging evi-
between mrhl and Wnt signaling. lncRNA033862 is a dence suggests the involvement of ROS in regulating stem
lncRNA expressed in the antisense direction that encom- cell homoeostasis.113 Excessive ROS can induce DNA dam-
passes exon 9 and the intronic sequence of the Gfra1 age that leads to apoptosis or senescence. Therefore, stem
gene. Its expression is tightly dependent on GDNF.107 cells possess antioxidant activity to safeguard genome
lncRNA033862 acts as an antisense transcript that binds integrity. Intriguingly, while excessive ROS also leads to
to Gfra1 mRNA according to the sequence complementar- apoptosis, completely removing the ROS species by scav-
ity and positively regulates the level of Gfra1. Knockdown enger treatment abolishes spermatogonial self-renewal.114
of lncRNA033862 triggers apoptosis and decreases the Moreover, knockout of ROS-producing enzymes, NADPH
stemness of cultured spermatogonia as revealed by trans- oxidase 1 (Nox1) or 3 (Nox3), reduces spermatogonial
plantation.107 These studies suggest the functional impor- self-renewal.114,115 These results suggest that a fine bal-
tance of lncRNAs in the regulation of spermatogonial ance in the level of ROS is required for the maintenance
self-renewal and differentiation. of spermatogonia.
102  The mitotic phase of spermatogenesis

PERSPECTIVES 6. Phillips BT, Gassei K, Orwig KE. Spermatogonial


In this chapter, we have presented the key concepts and the stem cell regulation and spermatogenesis. Philos
recent findings on the mitotic phase of spermatogenesis, Trans R Soc Lond B Biol Sci. 2010;365:1663–1678.
describing the sophisticated networks of nucleic acids and 7. de Rooij DG. The spermatogonial stem cell niche.
proteins that act extrinsically or intrinsically in orches- Microsc Res Tech. 2009;72:580–585.
trating the homeostasis between the self-renewal and 8. Kanatsu-Shinohara M, Takehashi M, Shinohara T.
differentiation of a heterogeneous population of spermato- Brief history, pitfalls, and prospects of mammalian
gonia. However, key questions regarding the biological spermatogonial stem cell research. Cold Spring Harb
properties of spermatogonia remain to be answered. For Symp Quant Biol. 2008;73:17–23.
example, regarding the fate decision in spermatogonia, 9. Oatley JM, Brinster RL. Regulation of spermatogo-
while the asymmetric distribution of the fate determinant nial stem cell self-renewal in mammals. Annu Rev
is observed during division, the syncytial state spermato- Cell Dev Biol. 2008;24:263–286.
gonia undergo stochastic fragmentation that includes a 10. Kanatsu-Shinohara M, Shinohara T. Spermatogonial
chain of cells rather than one daughter cell. Whether the stem cell self-renewal and development. Annu Rev
stochastic fragmentation also involves asymmetric segre- Cell Dev Biol. 2013;29:163–187.
gation of determinants or these are independent mecha- 11. Manku G, Culty M. Mammalian gonocyte and sper-
nisms remain unresolved. Moreover, the determinants, matogonia differentiation: Recent advances and remain-
regardless of nucleic acid or protein, remain to be char- ing challenges. Reproduction. 2015;149:​R139–157.
acterized. Regarding the spermatogonial niche, both the 12. Culty M. Gonocytes, from the fifties to the present:
vasculature niche in convoluted seminiferous tubules and Is there a reason to change the name? Biol Reprod.
the Sertoli valve niche adjacent to the rete testis have not 2013;89:46.
been completely characterized. The question as to whether 13. Yoshida S, Sukeno M, Nakagawa T et al. The first
As spermatogonia residing in these niches are similar round of mouse spermatogenesis is a distinctive
remain to be tested. With the advances in characterizing program that lacks the self-renewing spermatogonia
markers for spermatogonial subpopulations, different sub- stage. Development. 2006;133:1495–1505.
populations can be isolated for single-cell omics analyses. 14. Hara K, Nakagawa T, Enomoto H et al. Mouse
Therefore, we anticipate the discovery of novel regulators spermatogenic stem cells continually interconvert
at a higher resolution. Functional studies of these emerg- between equipotent singly isolated and syncytial
ing regulators will be the keys toward deciphering the states. Cell Stem Cell. 2014;14:658–672.
whole picture of the mitotic phase of spermatogenesis. 15. Nakagawa T, Sharma M, Nabeshima Y, Braun RE,
Yoshida S. Functional hierarchy and reversibility
ACKNOWLEDGMENTS within the murine spermatogenic stem cell compart-
This study was supported in part by National 973 proj- ment. Science. 2010;328:62–67.
ects (2013CB967401 and 2013CB967404), and GRF/ 16. van Pelt AM, de Rooij DG. Synchronization of the
RGC grant  (CUHK 14127316 and 14129316). We thank seminiferous epithelium after vitamin A replace-
Dr. Ruan Yechun (Hong Kong Polytechnic University) for ment in vitamin A-deficient mice. Biol Reprod.
the artwork. 1990;43:363–367.
17. Chan F, Oatley MJ, Kaucher AV et al. Functional
REFERENCES and molecular features of the Id4+ germline stem
1. Oatley JM, Brinster RL. The germline stem cell cell population in mouse testes. Genes Dev. 28,
niche unit in mammalian testes. Physiol Rev. 2014;1351–1362.
2012;92:577–595. 18. Aloisio GM, Nakada Y, Saatcioglu HD et al. PAX7
2. Ogawa T, Arechaga JM, Avarbock MR, Brinster expression defines germline stem cells in the adult
RL. Transplantation of testis germinal cells into testis. J Clin Invest. 2014;124:3929–3944.
mouse seminiferous tubules. Int J Dev Biol. 1997;41:​ 19. Yoshida S. Stem cells in mammalian spermatogen-
111–122. esis. Dev Growth Differ. 2010;52:311–317.
3. Kubota H, Avarbock MR, Brinster RL. Growth fac- 20. Ikami K, Tokue M, Sugimoto R et al. Hierarchical
tors essential for self-renewal and expansion of differentiation competence in response to retinoic
mouse spermatogonial stem cells. Proc Natl Acad Sci acid ensures stem cell maintenance during mouse
U S A. 2004;101:16489–16494. spermatogenesis. Development. 2015;142:1582–1592.
4. Kanatsu-Shinohara M, Miki H, Inoue K et al. Long- 21. Kanatsu-Shinohara M, Inoue K, Miki H et al. Clonal
term culture of mouse male germline stem cells origin of germ cell colonies after spermatogonial
under serum-or feeder-free conditions. Biol Reprod. transplantation in mice. Biol Reprod. 2006;75:68–74.
2005;72:985–991. 22. Zhang X, Ebata KT, Nagano MC. Genetic analy-
5. Yoshida S, Sukeno M, Nabeshima Y. A vasculature- sis of the clonal origin of regenerating mouse sper-
associated niche for undifferentiated spermatogonia matogenesis following transplantation. Biol Reprod.
in the mouse testis. Science. 2007;317:1722–1726. 2003;69:1872–1878.
References 103

23. Takashima S, Kanatsu-Shinohara M, Tanaka T et al. 38. Ishii K, Kanatsu-Shinohara M, Toyokuni S,


Rac mediates mouse spermatogonial stem cell hom- Shinohara T. FGF2 mediates mouse spermatogonial
ing to germline niches by regulating transmigra- stem cell self-renewal via upregulation of Etv5 and
tion through the blood-testis barrier. Cell Stem Cell. Bcl6b through MAP2K1 activation. Development.
2011;9:463–475. 2012;139:1734–1743.
24. DeFalco T, Potter SJ, Williams AV et al. Macrophages 39. Costoya JA, Hobbs RM, Barna M et al. Essential role
contribute to the spermatogonial niche in the adult of Plzf in maintenance of spermatogonial stem cells.
testis. Cell Rep. 2015;12:1107–1119. Nat Genet. 2004;36:653–659.
25. Nagano, MC, Yeh JR. The identity and fate deci- 40. Oatley JM, Oatley MJ, Avarbock MR, Tobias JW,
sion control of spermatogonial stem cells: Where Brinster RL. Colony stimulating factor 1 is an extrin-
is the  point of no return? Curr Top Dev Biol. sic stimulator of mouse spermatogonial stem cell
2013;102:61–95. self-renewal. Development. 2009;136:1191–1199.
26. Aiyama Y, Tsunekawa N, Kishi K et al. A niche for 41. Yeh JR, Zhang X, Nagano MC. Wnt5a is a cell-extrinsic
GFRalpha1-positive spermatogonia in the terminal factor that supports self-renewal of mouse spermato-
segments of the seminiferous tubules in hamster tes- gonial stem cells. J Cell Sci. 2011;124:2357–2366.
tes. Stem Cells. 2015;33:2811–2824. 42. Yeh JR, Zhang X, Nagano MC. Indirect effects of
27. Farkas AE, Capaldo CT, Nusrat A. Regulation of epi- Wnt3a/beta-catenin signalling support mouse sper-
thelial proliferation by tight junction proteins. Ann matogonial stem cells in vitro. PLoS One. 2012;7:​
N Y Acad Sci. 2012;1258:115–124. e40002.
28. Shinohara T, Avarbock MR, Brinster RL. beta1- and 43. Takase HM, Nusse R. Paracrine Wnt/beta-catenin
alpha6-integrin are surface markers on mouse sper- signaling mediates proliferation of undifferentiated
matogonial stem cells. Proc Natl Acad Sci U S A. spermatogonia in the adult mouse testis. Proc Natl
1999;96:5504–5509. Acad Sci U S A. 2016;113:E1489–1497.
29. Kanatsu-Shinohara M, Takehashi M, Takashima S 44. Boyer A, Yeh JR, Zhang X et al. CTNNB1 signaling
et al. Homing of mouse spermatogonial stem cells to in sertoli cells downregulates spermatogonial stem
germline niche depends on beta1-integrin. Cell Stem cell activity via WNT4. 2012;PLoS One 7:e29764.
Cell. 2008;3:533–542. 45. Dann CT, Alvarado AL, Molyneux LA et al.
30. Chen H, Fok KL, Yu S et al. CD147 is required for Spermatogonial stem cell self-renewal requires OCT4,
matrix metalloproteinases-2 production and germ a factor downregulated during retinoic acid-induced
cell migration during spermatogenesis. Mol Hum differentiation. Stem cells 2008;26:2928–2937.
Reprod. 2011;17:405–414. 46. Tong MH, Mitchell D, Evanoff R, Griswold MD.
31. Chen H, Lam Fok K, Jiang X, Chan C. New insights Expression of Mirlet7 family microRNAs in response
into germ cell migration and survival/apoptosis to retinoic acid-induced spermatogonial differentia-
in spermatogenesis: Lessons from CD147. Spermato­ tion in mice. Biol Reprod. 2011;85:189–197.
genesis. 2012;2:264–272. 47. Cunningham TJ, Duester G. Mechanisms of retinoic
32. Chen H, Fok KL, Jiang X et al. CD147 regulates apop- acid signalling and its roles in organ and limb devel-
tosis in mouse spermatocytes but not spermatogo- opment. Nat Rev Mol Cell Biol. 2015;16:110–123.
nia. Hum Reprod. 2012;27:1568–1576. 48. Gely-Pernot A, Raverdeau M, Célébi C et al.
33. Wang C, Fok KL, Cai Z, Chen H, Chan HC. CD147 Spermatogonia differentiation requires retinoic acid
regulates extrinsic apoptosis in spermatocytes receptor gamma. Endocrinology. 2012;153:438–449.
by modulating NFkappaB signaling pathways. 49. Kastner P, Mark M, Leid M et al. Abnormal sper-
Oncotarget. 2017;8(2):3132. matogenesis in RXR beta mutant mice. Genes Dev.
34. Hammoud SS, Low DH, Yi C et al. Transcription 1996;10:80–92.
and imprinting dynamics in developing postna- 50. Chung SS, Sung W, Wang X, Wolgemuth DJ. Retinoic
tal male  germline stem cells. Genes Dev. 2015;29:​ acid receptor alpha is required for synchronization
2312–2324. of spermatogenic cycles and its absence results in
35. Rossi P, Dolci S. Paracrine mechanisms involved in progressive breakdown of the spermatogenic pro-
the control of early stages of Mammalian spermato- cess. Dev Dyn. 2004;230:754–766.
genesis. Front Endocrinol (Lausanne). 2013;4:181. 51. Zhou Q, Li Y, Nie R et al. Expression of stimulated
36. Zhang Y, Wang S, Wang X et al. Endogenously pro- by retinoic acid gene 8 (Stra8) and maturation of
duced FGF2 is essential for the survival and prolifera- murine gonocytes and spermatogonia induced by
tion of cultured mouse spermatogonial stem cells. Cell retinoic acid in vitro. Biol Reprod. 2008;78:537–545.
Res. 2012;22:773–776. 52. Zhou Q, Nie R, Li Y et al. Expression of stimulated
37. Takashima S, Kanatsu-Shinohara M, Tanaka T et al. by retinoic acid gene 8 (Stra8) in spermatogenic cells
Functional differences between GDNF-dependent induced by retinoic acid: An in vivo study in vitamin
and FGF2-dependent mouse spermatogonial stem A-sufficient postnatal murine testes. Biol Reprod.
cell self-renewal. Stem Cell Rep. 2015;4:489–502. 2008;79:​35–42.
104  The mitotic phase of spermatogenesis

53. Hobbs RM, Fagoonee S, Papa A et al. Functional 68. Fleck D, Mundt N, Bruentgens F et al. Distinct puri-
antagonism between Sall4 and Plzf defines germline nergic signaling pathways in prepubescent mouse
progenitors. Cell Stem Cell. 2012;10:284–298. spermatogonia. J Gen Physiol. 2016;148:253–271.
54. Chen Y, Ma L, Hogarth C et al. Retinoid signaling 69. Idzko M, Ferrari D, Eltzschig HK. Nucleotide signal-
controls spermatogonial differentiation by regulat- ling during inflammation. Nature. 2014;509:310–317.
ing expression of replication-dependent core histone 70. Weissman AM. Themes and variations on ubiquity-
genes. Development. 2016;143:1502–1511. lation. Nat Rev Mol Cell Biol. 2001;2:169–178.
55. Hofmann MC, Braydich-Stolle L, Dettin L, Johnson 71. Glickman MH, Ciechanover A. The ubiquitin-
E, Dym M. Immortalization of mouse germ line proteasome proteolytic pathway: Destruction for the
stem cells. Stem Cells. 2005;23:200–210. sake of construction. Physiol Rev. 2002;82:373–428.
56. Liu Z, Guo J, Wang Y et al. CFTR-beta-catenin inter- 72. Komander D. The emerging complexity of protein
action regulates mouse embryonic stem cell differ- ubiquitination. Biochem Soc Trans. 2009;37:937–953.
entiation and embryonic development. Cell Death 73. Reyes-Turcu FE, Ventii KH, Wilkinson KD.
Differ. 2016;24(1):98. Regulation and cellular roles of ubiquitin-specific
57. Manova K, Huang EJ, Angeles M et al. The expres- deubiquitinating enzymes. Annu Rev Biochem.
sion pattern of the c-kit ligand in gonads of mice sup- 2009;78:363–397.
ports a role for the c-kit receptor in oocyte growth 74. Bose R, Manku G, Culty M, Wing SS. Ubiquitin-
and in proliferation of spermatogonia. Dev Biol. proteasome system in spermatogenesis. Adv Exp
1993;157:85–99. Med Biol. 2014;759:181–213.
58. Ohta H, Yomogida K, Dohmae K, Nishimune Y. 75. Yu J, Lan X, Chen X et al. Protein synthesis and
Regulation of proliferation and differentiation in degradation are essential to regulate germline stem
spermatogonial stem cells: The role of c-kit and its cell homeostasis in Drosophila testes. Development.
ligand SCF. Development. 2000;127:2125–2131. 2016;143:​2930–2945.
59. Pellegrini M, Grimaldi P, Rossi P, Geremia R, Dolci S. 76. Manku G, Wing SS, Culty M. Expression of the ubiq-
Developmental expression of BMP4/ALK3/SMAD5 uitin proteasome system in neonatal rat gonocytes
signaling pathway in the mouse testis: A potential and spermatogonia: Role in gonocyte differentiation.
role of BMP4 in spermatogonia differentiation. J Cell Biol Reprod. 2012;87:44.
Sci. 2003;116:3363–3372. 77. Luo J, Megee S, Dobrinski I. Asymmetric distribu-
60. Yang Y, Feng Y, Feng X et al. BMP4 Cooperates with tion of UCH-L1 in spermatogonia is associated with
retinoic acid to induce the expression of differen- maintenance and differentiation of spermatogonial
tiation markers in cultured mouse spermatogonia. stem cells. J Cell Physiol. 2009;220:460–468.
Stem Cells Int. 2016;2016:9536192. 78. Nakayama K, Nagahama H, Minamishima YA et al.
61. Nagano M, Ryu BY, Brinster CJ, Avarbock MR, Targeted disruption of Skp2 results in accumulation
Brinster RL. Maintenance of mouse male germ line of cyclin E and p27(Kip1), polyploidy and centro-
stem cells in vitro. Biol Reprod. 2003;68:2207–2214. some overduplication. EMBO J. 2000;19:2069–2081.
62. Kanatsu-Shinohara M, Inoue K, Takashima S et al. 79. Nakayama KI, Nakayama K. Regulation of the cell
Reconstitution of mouse spermatogonial stem cell cycle by SCF-type ubiquitin ligases. Semin Cell Devl
niches in culture. Cell Stem Cell. 2012;11:567–578. Biol. 2005;16:323–333.
63. Yang QE, Kim D, Kaucher A, Oatley MJ, Oatley JM. 80. Carrano AC, Eytan E, Hershko A, Pagano M. SKP2
CXCL12-CXCR4 signaling is required for the main- is required for ubiquitin-mediated degradation of
tenance of mouse spermatogonial stem cells. J Cell the CDK inhibitor p27. Nat Cell Biol. 1999;1:193–199.
Sci. 2013;126:1009–1020. 81. Fotovati A, Nakayama K, Nakayama KI. Impaired
64. Niu Z, Goodyear SM, Avarbock MR, Brinster RL. germ cell development due to compromised cell
Chemokine (C-X-C) ligand 12 facilitates trafficking cycle progression in Skp2-deficient mice. Cell Div.
of donor spermatogonial stem cells. Stem Cells Int. 2006;1:4.
2016;2016:5796305. 82. Kanarek N, Horwitz E, Mayan I et al. Spermatogenesis
65. He Z, Jiang J, Kokkinaki M, Dym M. Nodal signal- rescue in a mouse deficient for the ubiquitin ligase
ing via an autocrine pathway promotes prolifera- SCF{beta}-TrCP by single substrate depletion. Genes
tion of mouse spermatogonial stem/progenitor cells Dev. 2010;24:470–477.
through Smad2/3 and Oct-4 activation. Stem Cells. 83. Guardavaccaro D, Kudo Y, Boulaire J et al. Control
2009;27:2580–2590. of meiotic and mitotic progression by the F box pro-
66. Hasegawa K, Saga, Y. FGF8-FGFR1 signaling acts as tein beta-Trcp1 in vivo. Dev Cell. 2003;4:799–812.
a niche factor for maintaining undifferentiated sper- 84. Kanatsu-Shinohara M, Onoyama I, Nakayama KI,
matogonia in the mouse. Biol Reprod. 2014;91:145. Shinohara T. Skp1-Cullin-F-box (SCF)-type ubiqui-
67. Sahin Z, Szczepny A, McLaughlin EA et al. Dynamic tin ligase FBXW7 negatively regulates spermatogo-
Hedgehog signalling pathway activity in germline nial stem cell self-renewal. Proc Natl Acad Sci U S A.
stem cells. Andrology. 2014;2:267–274. 2014;111:8826–8831.
References 105

85. Zhao X, Heng JI, Guardavaccaro D et al. The HECT- 100. Sun J, Lin Y, Wu J. Long non-coding RNA expression
domain ubiquitin ligase Huwe1 controls neural dif- profiling of mouse testis during postnatal develop-
ferentiation and proliferation by destabilizing the ment. PLoS One. 2013;8:e75750.
N-Myc oncoprotein. Nat Cell Biol. 2008;10:643–653. 101. Luk AC, Chan WY, Rennert OM, Lee TL. Long non-
86. Inoue S, Hao Z, Elia AJ et al. Mule/Huwe1/Arf-BP1 coding RNAs in spermatogenesis: Insights from
suppresses Ras-driven tumorigenesis by preventing recent high-throughput transcriptome studies.
c-Myc/Miz1-mediated down-regulation of p21 and Reproduction. 2014;147:R131–141.
p15. Genes Dev. 2013;27:1101–1114. 102. Liang M, Li W, Tian H et al. Sequential expression
87. Chen D, Kon N, Li M et al. ARF-BP1/Mule is a criti- of long noncoding RNA as mRNA gene expression
cal mediator of the ARF tumor suppressor. Cell. in specific stages of mouse spermatogenesis. Sci Rep.
2005;121:1071–1083. 2014;4:5966.
88. Liu Z, Miao D, Xia Q, Hermo L, Wing SS. 103. Nishant KT, Ravishankar H, Rao MR. Characteri­
Regulated expression of the ubiquitin protein ligase, zation of a mouse recombination hot spot locus
E3(Histone)/LASU1/Mule/ARF-BP1/HUWE1, dur- encoding a novel non-protein-coding RNA. Mol Cell
ing spermatogenesis. Dev Dyn. 2007;236:2889–2898. Biol. 2004;24:5620–5634.
89. Liu Z, Oughtred R, Wing SS. Characterization 104. Arun G, Akhade VS, Donakonda S, Rao MR. mrhl
of E3Histone, a novel testis ubiquitin protein RNA, a long noncoding RNA, negatively regulates
ligase which ubiquitinates histones. Mol Cell Biol. Wnt signaling through its protein partner Ddx5/
2005;25:2819–2831. p68 in mouse spermatogonial cells. Mol Cell Biol.
90. Chuma S, Nakano T. piRNA and spermatogen- 2012;32:3140–3152.
esis in mice. Philos Trans R Soc Lond B Biol Sci. 105. Akhade VS, Arun G, Donakonda S, Rao MR.
2013;368:20110338. Genome wide chromatin occupancy of mrhl RNA
91. Keniry A, Oxley D, Monnier P et al. The H19 lin- and its role in gene regulation in mouse spermatogo-
cRNA is a developmental reservoir of miR-675 nial cells. RNA Biol. 2014;11:1262–1279.
that suppresses growth and Igf1r. Nat Cell Biol. 106. Akhade VS, Dighe SN, Kataruka S, Rao MR.
2012;14:659–665. Mechanism of Wnt signaling induced down regula-
92. Niu Z, Goodyear SM, Rao S et al. MicroRNA-21 regu- tion of mrhl long non-coding RNA in mouse sper-
lates the self-renewal of mouse spermatogonial stem matogonial cells. Nucleic Acids Res. 2016;44:387–401.
cells. Proc Natl Acad Sci U S A. 2011;108:12740–12745. 107. Li L, Wang M, Wang M et al. A long non-coding
93. He Z, Jiang J, Kokkinaki M et al. MiRNA-20 and RNA interacts with Gfra1 and maintains survival
mirna-106a regulate spermatogonial stem cell renewal of mouse spermatogonial stem cells. Cell Death Dis.
at the post-transcriptional level via targeting STAT3 2016;7:e2140.
and Ccnd1. Stem Cells. 2013;31:2205–2217. 108. Laplante M, Sabatini DM. mTOR signaling in growth
94. Yang QE, Racicot KE, Kaucher AV, Oatley MJ, Oatley control and disease. Cell. 2012;149:274–293.
JM. MicroRNAs 221 and 222 regulate the undif- 109. Hobbs RM, Seandel M, Falciatori I, Rafii S, Pandolfi
ferentiated state in mammalian male germ cells. PP. Plzf regulates germline progenitor self-renewal
Development. 2013;140:280–290. by opposing mTORC1. Cell. 2010;142:468–479.
95. Tong MH, Mitchell DA, McGowan SD, Evanoff 110. Hobbs RM, La HM, Mäkelä JA et al. Distinct
R, Griswold MD. Two miRNA clusters, Mir-17-92 germline progenitor subsets defined through Tsc2-
(Mirc1) and Mir-106b-25 (Mirc3), are involved in the mTORC1 signaling. EMBO Rep. 2015;16:467–480.
regulation of spermatogonial differentiation in mice. 111. Ito K, Suda T. Metabolic requirements for the main-
Biol Reprod. 2012;86:72. tenance of self-renewing stem cells. Nat Rev Mol Cell
96. Huszar JM, Payne CJ. MicroRNA 146 (Mir146) mod- Biol. 2014;15:243–256.
ulates spermatogonial differentiation by retinoic acid 112. Kanatsu-Shinohara, M, Tanaka T, Ogonuki N et al.
in mice. Biol Reprod. 2013;88:15. Myc/Mycn-mediated glycolysis enhances mouse
97. Chen J, Cai T, Zheng C et al. MicroRNA-202 main- spermatogonial stem cell self-renewal. Genes Dev.
tains spermatogonial stem cells by inhibiting cell 2016;30:2637–2648.
cycle regulators and RNA binding proteins. Nucleic 113. Bigarella CL, Liang R, Ghaffari S. Stem cells
Acids Res. 2016;45(7):4142–4157. and the impact of ROS signaling. Development.
98. Bao J, Wu J, Schuster AS, Hennig GW, Yan W. 2014;141:4206–4218.
Expression profiling reveals developmentally regu- 114. Morimoto H, Iwata K, Ogonuki N et al. ROS are
lated lncRNA repertoire in the mouse male germ- required for mouse spermatogonial stem cell self-
line. Biol Reprod. 2013;89:107. renewal. Cell Stem Cell. 2013;12:774–786.
99. Laiho A, Kotaja N, Gyenesei A, Sironen A. 115. Morimoto H, Kanatsu-Shinohara M, Shinohara T.
Transcriptome profiling of the murine testis dur- ROS-Generating Oxidase Nox3 regulates the self-
ing the first wave of spermatogenesis. PLoS One. renewal of mouse spermatogonial stem cells. Biol
2013;8:e61558. Reprod. 2015;92:147.
Genetics of mammalian meiosis
FANG YANG and P. JEREMY WANG
8
INTRODUCTION fetal male germ cells.7 Therefore, retinoic acid and FGF9
In sexually reproducing organisms, diploid germ cells antagonistically regulate the sex-specific timing of meiotic
undergo meiotic cell divisions to produce haploid gametes. entry.
Meiosis occurs in both sexes. After fertilization, the diploid MAX, a partner of MYC family proteins, suppresses the
state is restored in the resulting embryos. Meiosis comprises expression of meiosis genes in embryonic stem cells and
one round of DNA replication followed by two successive germline stem cells.8 Reduction of Max causes precocious
rounds of cell divisions. After DNA replication, each chro- meiotic entry in these stem cells in a Stra8-dependent
mosome consists of two sister chromatids. The chromatids manner, as evidenced by meiotic-like cytological changes,
from the homologous chromosomes are called nonsister but not in fibroblasts. MAX is a component of an atypical
chromatids. During meiosis I, homologous chromosomes polycomb repressive complex 1 (PRC1.6). Therefore, MAX
pair, synapse, recombine, and segregate. Recombination represses meiosis genes in stem cells partly through the
occurs between nonsister chromatids of the homologous PRC1.6 complex.
chromosomes. Meiotic recombination, a hallmark of meio- MEIOC is expressed at the onset of meiosis and pres-
sis, is critical for faithful segregation of homologous chromo- ent throughout the meiotic prophase I.9,10 In mice, germ
somes during the first meiotic cell division. Unlike meiosis cells lacking MEIOC initiate meiosis but proceed to meta-
I, meiosis II involves segregation of sister chromatids and phase precociously without completing chromosomal
thus is similar to mitosis. Because of meiotic recombina- synapsis and meiotic recombination. MEIOC interacts
tion and random chromosome segregation, meiosis shuffles with YTHDC2, an RNA helicase with a YTH domain.
genetic information and thus increases genetic diversity in YTHDC2 is also required for meiosis.11 Both MEIOC and
the offspring, which provides a constant source of genetic YTHDC2 bind to RNA transcripts. However, the identity
variations for selection during evolution. Errors in meio- of the associated transcripts differs between two studies.9,10
sis are leading causes of genetic diseases including aneu- In the study by Abby et al., MEIOC/YTHDC2 binds to and
ploidy (such as Down syndrome, Klinefelter syndrome, and protects meiosis-specific transcripts upon meiotic entry.9
Turner syndrome), pregnancy loss, and infertility. Studies However, Soh et al. reported that MEIOC/YTHDC2 binds
from a variety of model organisms (yeast, fly, worm, plant, to mitotic cell cycle-associated transcripts but not meiosis-​
and mouse) have elucidated many conserved and divergent specific transcripts.10 Soh et al. hypothesizes that the
molecular processes in the regulation of meiosis. Genetic MEIOC/YTHDC2 complex represses mitosis-associated
and cell biological studies have identified a large number of transcripts to facilitate mitosis to meiosis transition and
proteins that are specifically involved in meiosis. This chap- to promote a meiotic cell cycle program. Despite this dis-
ter examines the major advances in the study of mamma- agreement, the MEIOC/YTHDC2 complex is required to
lian meiosis. maintain the lengthy meiotic prophase I program.

MEIOTIC INITIATION TELOMERE-LED MEIOTIC CHROMOSOME MOVEMENT


The timing of meiotic initiation differs between sexes. In During the meiotic prophase I, chromosomes undergo
males, spermatogonia (diploid germ cells) enter meiosis at rapid movement. Chromosome movement facilitates pair-
puberty, whereas in females, oogonia enter meiosis shortly ing of homologous chromosomes and resolves entangle-
after sex determination at the embryonic stage. Retinoid ments between nonhomologs. Chromosome movement is
signaling regulates the timing of meiotic entry in mice.1,2 led by telomeres and driven by the cytoplasmic microtu-
Retinoic acid induces the expression of Stra8. Stra8 is bules.12,13 Chromosomal ends (telomeres) are attached to
required for meiotic entry in female germ cells at the fetal the nuclear envelope through the SUN1-KASH5 nuclear
stage and in male germ cells at puberty.3,4 Rec8, encod- membrane proteins (Figure 8.1 and Table 8.1). SUN1, an
ing a meiosis-specific cohesin, is induced during premei- inner nuclear membrane protein, interacts with KASH5,
otic S phase.5 The expression of Rec8 is Stra8-independent. an outer nuclear membrane protein.14 KASH5 binds to
Retinoic acid induces the expression of both Stra8 and cytoplasmic dynein proteins, which are microtubule-
Rec8, demonstrating that retinoic acid activates at least based motors.15 Three meiosis-specific proteins, MAJIN,
two molecular pathways in meiosis.6 Fetal male germ cells TERB1, and TERB2, form a meiosis-specific telomere
do not enter meiosis but instead enter mitotic arrest. It cap.16 TERB1 binds to telomeres by forming a heterocom-
turns out that the enzyme CYP26B1 degrades retinoids in plex with TRF1, a canonical telomere protein, and also
fetal male germ cells to prevent precocious meiotic entry interacts with SUN1.17 MAJIN is a DNA-binding trans-
until puberty.2 FGF9 further inhibits meiotic initiation in membrane protein. Therefore, MAJIN-TERB1/2 complex

106
Chromosomal synapsis  107

Chromosomal synapsis

Chromosome movement SC/cohesins


KASH5, SUN1, MAJIN, SYCP1, SYCP2, SYCP3, SYCE1,
Meiotic entry
TERB1, TERB2, SPDYA, SYCE2, SYCE3, TEX12, REC8,
STRA8, CYP26B1 CDK2, Lamin C2 RAD21L, SMC1B, STAG3
DAZL, MAX
Meiotic recombination

Hotspot DSB formation DSB repair Crossover


PRDM9 SPO11, TOPOVIBL, DMC1, MND1, HOP2, MLH1, MLH3, RNF212
HORMAD1, IHO1, MEIOB, SPATA22, HEI10, CNTD1, MSH4,
REC114, MEI1, MEI4 MCMDC2, TEX15, MSH5, TEX11, EXO1,
TRIP13 HFM1, MUS81, BLM,
FANCJ

Additional meiosis proteins: DMRTB1, FMRP, HSPA2, MDC1, MEIOC, NXF2, RNF8, SCAI, SCML2, SETX, SOHLH2,
TET1, TEX14, TEX19, TH2A/TH2B, YTHDC2, ZRP318

Figure 8.1  A summary of proteins involved in the regulation of mammalian meiosis. The function of these proteins in meio-
sis have been demonstrated by studies of knockout mice. Additional meiosis proteins include proteins that cannot be definitely
placed into these categories or span more than one category. Description of genes is shown in Table 8.1. Most proteins in the piRNA
pathway are essential for male meiosis but are not shown here. The piRNA pathway proteins have been reviewed elsewhere.118,119
Developmentally essential genes with meiotic defects in germ cell specific conditional knockout mice are not listed.

physically links telomeres to the nuclear envelope.16 At imaging further demonstrate that the CE is a bilayered
the leptotene-zygotene transition, telomeres are clustered helical structure.24,25
at one pole of the nucleus (referred to as bouquet forma- The prophase I is further divided into substages largely
tion) to ensure accurate homolog pairing and synapsis. based on the morphology of the synaptonemal complex:
Both Speedy A (SPDYA) and CDK2 localize to telomeres leptotene, zygotene, pachytene, and diplotene. At the lep-
in meiotic germ cells.18–20 Speedy A is a noncanonical acti- totene stage, synaptonemal complex proteins form axial
vator of cyclin-dependent kinases (CDK) and binds to elements (AEs) along chromosomal axes but synapsis is
CDK2. CDK2 phosphorylates SUN1 in vitro. Therefore, not yet formed. At the zygotene stage, chromosomal syn-
the Speedy A-CDK2 binding regulates the initial telomere-​ apsis is initiated but incomplete. At this stage, CE and TF
nuclear envelope attachment. Loss of any of these proteins components connect the two AEs and thus only localize to
(SUN1, KASH5, MAJIN, TERB1, TERB2, Speedy A, and the synapsed regions. AEs are called LEs in synapsis. The
CDK2) causes failures in chromosome movement, homo- subsequent pachytene stage is characterized by full synap-
log pairing, and chromosomal synapsis, demonstrating sis along the entire length of all pairs of homologous chro-
the requirement of telomere-led bouquet formation for mosomes except for the X-Y chromosomes in male germ
meiosis. cells. At the diplotene stage, CE and TF components begin
to disassemble from SCs to cause desynapsis of homolo-
CHROMOSOMAL SYNAPSIS gous chromosomes.
During the extended prophase of meiosis I, homologous The tripartite structure of SC is highly conserved across
chromosomes are physically juxtaposed in a process species from yeast to human. However, the SC proteins are
called synapsis. The synaptonemal complex (SC) provides not conserved at the protein sequence level across species
the physical connection between homologous chromo- but all contain the coiled-coil secondary structure, which
somes during synapsis.21,22 The synaptonemal complex is is a protein–protein interaction domain.21,22 Synaptonemal
a 200-nm-wide tripartite protein structure consisting of complex proteins have been identified (Figure 8.1 and
two lateral elements (LEs) and one central element (CE). Table 8.1). In mammals, SYCP2 and SYCP3 are LE com-
One LE is formed along the sister chromatids. Transverse ponents.26–29 Four CE components are known: SYCE1,30
elements (TFs) connect the lateral elements and the central SYCE2,31 SYCE3,32 and TEX12.33 SYCP1 constitutes the
element. Under transmission electron microscopy, the tri- transverse filaments, with the N- and C-termini located at
partite structure of SC is shown by the two electron-dense CE and LEs, respectively.34 Crystal structures of several SC
LEs and one electron-dense CE in between.23 The LEs and proteins have been reported: C-terminal coiled-coil domain
CE of the SC cannot be resolved as individual structures of SYCP1,35 SYCP3,36 and SYCE3.37 Based on these structure
under conventional light microscopy. Superresolution studies, SYCP1 C-terminal region and SYCE3 form homodi-
imaging has not only revealed the tripartite structure mers, respectively, whereas SYCP3 forms a tetramer. In addi-
but also the organization of SC as a twisted helical struc- tion, SYCP3 tetramer interacts with DNA and self-assembles
ture.24 Immunoelectron microscopy and superresolution into filamentous structures that resemble LEs.36 In genetic
108  Genetics of mammalian meiosis

Table 8.1  Mouse genes involved in meiosis. Table 8.1 (Continued)  Mouse genes involved in
meiosis.
Gene
symbol Gene
(alias) Description symbol
Blm Bloom syndrome RecQ-like helicase (alias) Description
cdk2 Cyclin-dependent kinase 2 Rad21l RAD21 cohesin complex component like 1
Cntd1 Cyclin N-terminal domain containing 1 Rec114 REC114 meiotic recombination protein
CYP26B1 Cytochrome P450 family 26 subfamily B Rec8 REC8 meiotic recombination protein
member 1 Rnf8 Ring finger protein 8
Dazl Deleted in azoospermia like Rnf212 Ring finger protein 212
Dmc1 DNA meiotic recombinase 1 Scai Suppressor of cancer cell invasion
Dmrtb1 DMRT-like family B with proline-rich Scml2 Scm polycomb group protein like 2
(Dmrt6) C-terminal, 1 Setx Senataxin
Exo1 Exonuclease 1 Smc1b Structural maintenance of chromosomes 1B
Brip1 (FancJ) BRCA1 interacting protein C-terminal Sohlh2 Spermatogenesis and oogenesis specific
helicase 1 basic helix-loop-helix 2
Fmr1 (Fmrp) Fragile X mental retardation syndrome 1 Spata22 Spermatogenesis associated 22
Ccnb1ip1 Cyclin B1 interacting protein 1 Spdya Speedy/RINGO cell cycle regulator family
(Hei10) member A
Hfm1 HFM1, ATP-dependent DNA helicase Spo11 SPO11 meiotic protein covalently bound to
homolog DSB
Psmc3ip Proteasome (prosome, macropain) 26S Stag3 Stromal antigen 3
(Hop2) subunit, ATPase 3, interacting protein Stra8 Stimulated by retinoic acid gene 8
Hormad1 HORMA domain containing 1 Sun1 Sad1 and UNC84 domain containing 1
Hspa2 Heat shock protein family A (Hsp70) Syce1 Synaptonemal complex central element
member 2 protein 1
Iho1 Interactor of HORMAD1 Syce2 Synaptonemal complex central element
Ccdc155 Coiled-coil domain containing 155 protein 2
(Kash5) Syce3 Synaptonemal complex central element
Lmna (Lamin Lamin A protein 3
C2) Sycp1 Synaptonemal complex protein 1
Majin Membrane anchored junction protein Sycp2 Synaptonemal complex protein 2
Max MYC associated factor X Sycp3 Synaptonemal complex protein 3
Mcmdc2 Minichromosome maintenance domain Terb1 Telomere repeat binding bouquet formation
containing 2 protein 1
Mdc1 Mediator of DNA damage checkpoint 1 Terb2 Telomere repeat binding bouquet formation
Mei1 Meiotic double-stranded break formation protein 2
protein 1 Tet1 tet methylcytosine dioxygenase 1
Mei4 Meiotic double-stranded break formation Tex11 Testis expressed gene 11
protein 4 Tex12 Testis expressed gene 12
Meiob Meiosis specific with OB domains Tex14 Testis expressed gene 14
Meioc Meiosis specific with coiled-coil domain Tex15 Testis expressed gene 15
Mlh1 mutL homolog 1 Tex19 Testis expressed gene 19
Mlh3 mutL homolog 3 Hist1h2aa Histone cluster 1, H2aa
Mnd1 Meiotic nuclear divisions 1 (Th2a)
Msh4 mutS homolog 4 Hist1h2ba Histone cluster 1, H2ba
Msh5 mutS homolog 5 (Th2b)
Mus81 MUS81 structure-specific endonuclease TopoVIbl TopoVI DNA topoisomerase subunit B like
subunit Trip13 Thyroid hormone receptor interactor 13
Nxf2 Nuclear RNA export factor 2 Ythdc2 YTH domain containing 2
Prdm9 PR domain containing 9 Zfp318 Zinc finger protein 318
(Continued )
Meiotic recombination  109

studies, loss of any of SC proteins results in a failure in the SC unsynapsed. Sex chromosomes are subjected to silenc-
assembly and thus a block or defect in meiotic progression. ing at the pachytene and diplotene stages in a phenom-
In addition to SC proteins, cohesins are essential com- enon termed meiotic sex chromatin silencing (MSCI).
ponents of the axial/lateral elements. The cohesin complex Transcriptional silencing of sex chromosomes is a special
is required for sister chromatid cohesion in both mitosis case of a general meiotic silencing response to chromo-
and meiosis. The mammalian mitotic cohesin complex somal unsynapsis. Much progress has been made in under-
consists of two structural maintenance of chromosome standing the regulation and function of MSCI. For further
(SMC) proteins (SMC1 and SMC3) and two non-SMC interest in MSCI, please refer to a review by Turner.54
subunits (RAD21 and STAG1 or STAG2). Four meiosis-
specific paralogues of mitotic cohesins have been identi- MEIOTIC RECOMBINATION
fied in mammals: REC8,5,38 RAD21L,39–41 SMC1B,42 and In many species including mouse, chromosomal synapsis
STAG3.43 REC8 and RAD21L are homologous to RAD21. and meiotic recombination are interdependent. These two
SMC1B bears sequence similarity with SMC1. STAG3 processes are tightly coordinated. The synaptonemal com-
replaces STAG1/STAG2 in meiotic cohesin complexes. plex provides a structural framework for meiotic recom-
All these meiosis-specific cohesin proteins localize to the bination. Meiotic recombination initiates and stabilizes
axial elements. Strikingly, absence of REC8 leads to syn- chromosomal synapsis. Meiotic recombination leads to
apsis between sister chromatids, suggesting that one of the reciprocal exchange of genetic material between homolo-
main functions of REC8 is to limit synapsis to homologous gous chromosomes (more specifically, between nonsister
chromosomes.38,44 Genetic and cell biological analyses chromatids). In addition, formation of chiasmata resulting
have shown that these meiosis-specific cohesins regulate from recombination is essential for faithful segregation
the axial element formation, axis length, sister chromatid of homologous chromosomes during the first meiotic cell
cohesion, and meiotic recombination.45,46 division. Meiotic recombination involves a large number
While the identification and characterization of these of proteins at several distinct stages, which are discussed
meiosis-specific structural proteins have elucidated their separately.
role in SC formation, the mechanism regulating SC dis-
assembly is poorly understood. In budding yeast, polo- Programmed meiotic DSB formation
like kinase Cdc5 promotes SC disassembly at the end of Meiotic recombination begins with the formation of DNA
the pachytene stage.47 In mouse spermatocytes, polo-like double-strand breaks (DSBs). In mouse, about 300 DSBs
kinase 1 (PLK1) localizes to the SC.48 PLK1 phosphory- are generated in each meiotic germ cell. Formation of
lates SYCP1 and TEX12. Treatment of spermatocytes with DSBs is catalyzed by SPO11, a homologue of TopoVI DNA
the PLK1 inhibitor prevents removal of SYCP1 and TEX12 topoisomerase subunit A (TopoVIA).55–57 The activity of
from the SC.48 In addition, PLK1 promotes dissociation SPO11 requires TOPOVIBL, which displays ­ structural
of REC8 and RAD21L from SC.39 Although a Plk1 mouse similarity to the TopoVI DNA topoisomerase subunit B.58
mutant is needed to address its in vivo requirement, the TOPOVIBL interacts with SPO11 and is required for
PLK1 function in SC disassembly appears to be conserved DSB formation during meiosis. Additional proteins are
between yeast and mouse. required for DSB formation: MEI1,59 MEI4,60 REC114,60
The dynamic association of the cohesin complex with HORMAD1,61,62 and IHO1.63 MEI4 and REC114 interact
chromatin during the mitotic cell cycle is controlled by directly.60 HORMAD1 localizes to unsynapsed chromo-
a number of regulatory proteins in the loading, mainte- somal axis and recruits IHO1 through direct interaction.
nance, and release of cohesins: NIPBL, Sororin, and WAPL. IHO1 recruits MEI4-REC114 to unsynapsed chromosomal
While NIPBL is a cohesin loading factor, WAPL is a cohe- axis. The formation of this complex (HORMAD1-IHO1-
sin releasing factor. WAPL interacts with PDS5, a cohesin- REC114-MEI4) activates the SPO11-TOPOVIBL com-
binding protein, to remove cohesins. Sororin maintains plex.63 MEI1 is also important for recruitment of MEI4.64
cohesion by antagonizing WAPL. Mechanistically, Sororin In conclusion, meiotic DSB formation is genetically pro-
binds to PDS5B to prevent association of PDS5B with grammed and meticulously executed (Figure 8.1).
WAPL and thus inhibit the activity of WAPL.49 In meiotic
germ cells, all these factors (NIPBL, Sororin, PDS5, and DSB repair
WAPL) localize to the synaptonemal complex, suggesting Formation of DSBs entails a DNA damage response, lead-
the conserved functions of these regulatory factors in both ing to phosphorylation of histone H2AX by ATM.65,66
mitosis and meiosis.50–52 Furthermore, in meiosis, NIMA- Following generation of DSBs, SPO11 is covalently
like kinase-1 (NEK1) phosphorylates protein phosphatase attached to the 5’ strands of both ends of the DSB. DSB
1 gamma (PP1γ), which dephosphorylates WAPL to facili- breaks undergo end resection to produce 3’ single-stranded
tate cohesin removal.52 DNA (ssDNA) ends, releasing SPO11-oligonucleotide
Chromosomal synapsis is sexually dimorphic.53 In complexes.67 RPA, an ssDNA-binding complex, coats the
female meiosis, the two X chromosomes, like autosomes, ssDNA to protect it from degradation and prevent sec-
are fully synapsed. However, in male meiosis, the X and ondary structure formation.68 RAD51 and DMC1 are
Y chromosomes only form synapsis in the pseudoauto- also ssDNA-binding proteins and exhibit recombinase
somal regions, leaving the majority of sex chromosomes activities. RAD51/DMC1 recombinase is loaded onto
110  Genetics of mammalian meiosis

ssDNA, displaces RPA, and directs strand invasion into the MLH1/MLH3-dependent class I pathway. The class
the duplex of the homologous chromosomes, resulting in II pathway is minor and independent of MLH1/MLH3
the formation of displacement loop (D-loop).69 DMC1 is but requires MUS81.92 These two crossover pathways are
a meiosis-specific paralogue of RAD51.70 DMC1 favors coordinated because the class I pathway is upregulated as
repair of meiotic DSBs through a nonsister chromatid shown by the increased accumulation of MLH1 foci in the
rather than a sister chromatid. The MND1/HOP2 het- absence of MUS81.92
erodimer interacts with RAD51/DMC1 and stimulates
their recombinase activity to generate the D-loop recom- Meiotic recombination hotspot
bination intermediates.71 MCMDC2, a member of the Meiotic recombination occurs more frequently at cer-
minichromosome maintenance (MCM) helicase family, tain genomic locations (so-called hotspots) than oth-
is required for RAD51- and DMC1-directed strand inva- ers.93 The major advance in the study of recombination
sion or stabilization of recombination ­intermediates.72,73 hotspot is the identification of PRDM9. PRDM9 is a main
MEIOB is a ­meiosis-specific ssDNA-binding ­protein.74,75 determinant of meiotic recombination hotspots in mam-
MEIOB interacts and forms a heterodimer with SPATA22, mals.94–96 PRDM9 binds to the 13-mer hotspot consensus
which is also a meiosis-specific protein.74,76 The MEIOB/ DNA motif through its array of DNA-binding zinc fin-
SPATA22 complex interacts with the RPA complex and gers. PRDM9 catalyzes both trimethylation of lysine 4
localizes to DSB sites.77 The MEIOB/SPATA22 complex of histone H3 (H3K4me3)97 and trimethylation of lysine
has been hypothesized to function in the second end 36 of histone H3 (H3K36me3).98 Meiotic recombination
capture and maintenance of RAD51 foci.74,78 Second end hotspots are marked by both H3K4me3 and H3K36me3
capture results in the formation of the recombination modifications.99 H4K44 acetylation (H4K44ac) marks
intermediate–double Holliday junction (dHJ). Most pro- meiotic recombination hotspots in budding yeast but its
teins involved in meiotic DSB repair form foci at the DSB relationship with mammalian hotspots is unknown.100
sites on meiotic chromosomes. Since meiotic recombina- PRDM9 directs recombination away from functional
tion occurs in both male and female germ cells, loss of any genomic elements such as gene promoters.101 The Prdm9
of these recombination-related proteins leads to meiotic gene is rapidly evolving, especially in the zinc-finger cod-
arrest and thus sterility in both sexes. ing region.102 This rapid evolution is driven by the inherent
“self-destruct” property of recombination hotspots.95 A
Crossover formation recombination hotspot is favored for DSB formation and
Double Holliday junctions are resolved into either cross- thus is more likely to be replaced with a coldspot allele
overs or noncrossovers. In mouse meiotic germ cells, through homologous recombination. The 13-mer hotspot
a subset of DSBs are turned into ~25 crossovers per cell consensus motif varies in sequences among species. As
and the remaining DSBs result in noncrossovers. Each a new hotspot consensus emerges, the zinc-finger region
pair of homologous chromosomes must have at least one of PRMD9 evolves to bind to the new hotspots and vice
crossover, since crossover is required for proper chromo- versa. For example, humanizing the zinc-finger domain
some segregation during the first meiotic cell division. of PRDM9 in mice changes the positions of DSB hotspots
MSH4-MSH5 stabilizes dHJ structures and promotes in mouse.103 Prmd9 is the only known mammalian spe-
crossover formation (Figure 8.1).79 TEX11 promotes cross- ciation gene and is responsible for the hybrid sterility
over formation and interacts with the synaptonemal com- between mouse subspecies.104 How are H3K4me3-marked
plex protein SYCP2.80 MLH1-MLH3 is an endonuclease hotspots recognized for DSB formation? Recent studies
that is required for resolution of dHJs.81,82 MSH2-MSH3 provide insights into the connection of the hotspot to DSB.
stimulates the endonuclease activity of MLH1-MLH3.83 PRMD9 binding leads to nucleosome depletion at the
Exonuclease 1 (EXO1) is required for formation of chi- hotspot, a possible permissive environment for DSB for-
asmata even though the number of MLH1 foci is normal mation.105 PRDM9 interacts with CXXC1, which in turn
in its absence.84 The nuclease-dead Exo1 mutant mice are interacts with IHO1.106 IHO1 is a part of the protein com-
fertile, implying that EXO1 plays a structural but not cat- plex that activates SPO11. In addition to CXXC1, PRDM9
alytic role in meiosis.85 SUMO ligase RNF212 and ubiq- interacts with EWSR1, EHMT2, and CDYL.107 In testis,
uitin ligase HEI10 play antagonistic roles in crossover PRDM9 is complexed with REC8, SYCP1, and SYCP3.107
designation.86 RNF212 and HEI10 mediate localization of Therefore, the epigenetic and chromatin states specify the
SUMO and ubiquitin to the synaptonemal complex, which location of DSBs and PRDM9 is recruited through asso-
in turn recruits the proteasome.87 The proteasome regu- ciation with cohesin and SC.
lates homolog pairing and crossover formation in yeast.88
Cyclin-related protein CNTD1 regulates the association of IN VITRO MEIOSIS
HEI10, RNF212, and MSH4 with the recombination inter- In vitro spermatogenesis has been pursued for nearly
mediates.89 Interestingly, HEI10, RNF212, and TEX11 are a century. Many studies reported in vitro production of
dosage-sensitive regulators of crossover formation, sug- haploid male germ cells or oocytes from embryonic stem
gesting that they are rate-limiting factors.86,90 Importantly, cells. The common drawbacks are the extremely low effi-
RNF212 and TEX11 regulate genome-wide recombination ciency and the lack of proof of gamete function. One of
rates.90,91 The majority of crossovers are formed through the critical barriers is progression through meiosis in
References 111

Table 8.2  Meiosis-specific genes implicated in human infertility.


Gene Description Clinical phenotype Mutation Reference
MEIOB Meiosis specific with OB Meiotic arrest (NOA) Missense 120

domains
MSH4 mutS homolog 4 POF Splice site 121

PSMC3IP PSMC3 interacting protein Ovarian dysgenesis Deletion of one residue 122

(aka HOP2) (Glu201)


SYCE1 Synaptonemal complex POF Nonsense 123

central element protein 1 Meiotic arrest (NOA) Splice site 124

SYCP3 Synaptonemal complex NOA Heterozygous a for frame- 125

protein 3 shift mutation (1 bp deletion)


STAG3 Stromal antigen 3 POF Frame-shift deletion 126

TEX11 Testis expressed gene 11 Meiotic arrest (NOA) Deletion, nonsense, splice 90,117

site, missense
TEX14 Testis expressed gene 14 Meiotic arrest (NOA) Frame-shift deletion 120

TEX15 Testis expressed gene 15 NOA Nonsense 127,128

Abbreviations: NOA, nonobstructive azoospermia; POF, premature ovarian failure.

vitro.108 The prophase of meiosis I in mouse spans 14 days, To date, human genetic studies have identified causative
8 days out of which are spent on the pachytene stage.109 The mutations in nine meiosis-specific genes (Table 8.2). These
pachytene stage is critical since crossovers are matured at mutations cause either azoospermia in men or prema-
the mid-late pachytene stage. In previous testicular organ ture ovarian failure in women. Most of these mutations
cultures, germ cells were not able to progress beyond the were found in the offspring of consanguineous marriage
pachytene stage.110 A mouse testicular organ culture sys- and identified by exome sequencing of affected individu-
tem is successful for in vitro production of functional als. Homozygous mutations in MEIOB, PSMC3IP, STAG3,
sperm.111,112 The crucial factor for success in this system is SYCE1, TEX14, and TEX15 were identified in the offspring
the use of serum-free culture media. In this organ culture of consanguineous couples (Table 8.2). Sequencing of
system, mouse testis tissue is used, thus preserving the sporadic cases of infertile men reveals that mutations in
three-dimensional architecture of the testicular tubules. TEX11, an X-linked meiosis-specific gene, cause infertility
Using this organ culture system, sperm are also derived in ~1% of nonobstructive azoospermia.90,117 In the era of
from mouse spermatogonial stem cells.112,113 In a cocul- precision medicine, future genomic sequencing of infertile
ture system with neonatal testicular somatic cells, primor- patients is expected to identify causative mutations in more
dial germ cell-like cells derived from ES cells in vitro are meiosis-specific genes. Such information will be valuable
induced to complete meiosis to generate haploid spermatid- for diagnosis and genetic counseling in the treatment of
like cells.114 In this system, cultures were sequentially infertility in the upcoming era of precision medicine.
treated with retinoic acid, hormones, and other factors.114
Seminiferous tubules can be reconstructed from neonatal ACKNOWLEDGMENTS
testicular cells and cultured in vitro to support germ cell We would like to thank all our colleagues whose con-
differentiation but only up to the stage of pachytene.115 tributions in this topic have not been cited due to space
Retinoic acid is sufficient to induce mouse spermatogonial limitation. This work is supported by National Institutes
stem cells to enter meiosis and proceed up to the zygo- of Health/National Institute of General Medical Sciences
tene stage.116 These in vitro spermatogenesis systems will (R35GM118052 to P.J.W.).
facilitate mechanistic studies of meiosis. Optimization of
the existing in vitro meiosis systems and development of REFERENCES
new in vitro systems will likely revolutionize treatment
1. Koubova J, Menke DB, Zhou Q et al. Retinoic acid reg-
of human infertility.
ulates sex-specific timing of meiotic initiation in mice.
Proc Natl Acad Sci U S A. 2006;103(8):2474–2479.
IMPLICATIONS IN HUMAN INFERTILITY 2. Bowles J, Knight D, Smith C et al. Retinoid sig-
Genetic studies in mouse and other model organisms have naling determines germ cell fate in mice. Science.
identified many meiosis-specific proteins discussed here 2006;312(5773):596–600.
and elucidated the molecular mechanisms underlying their 3. Baltus AE, Menke DB, Hu YC et al. In germ cells
functions. These functional studies provide a strong ratio- of mouse embryonic ovaries, the decision to enter
nale for genetic studies in humans. Mutations in meiosis- meiosis precedes premeiotic DNA replication. Nat
specific genes are expected to be causes of human infertility. Genet. 2006;38(12):1430–1434.
112  Genetics of mammalian meiosis

4. Anderson EL, Baltus AE, Roepers-Gajadien HL 20. Viera A, Alsheimer M, Gomez R et al. CDK2 regu-
et al. Stra8 and its inducer, retinoic acid, regu- lates nuclear envelope protein dynamics and telo-
late meiotic initiation in both spermatogenesis mere attachment in mouse meiotic prophase. J Cell
and oogenesis in mice. Proc Natl Acad Sci U S A. Sci. 2015;128(1):88–99.
2008;105(39):14976–14980. 21. Yang F, Wang PJ. The Mammalian synaptonemal
5. Watanabe Y, Nurse P. Cohesin Rec8 is required for complex: A scaffold and beyond. Genome Dyn.
reductional chromosome segregation at meiosis. 2009;5:69–80.
Nature. 1999;400(6743):461–464. 22. Cahoon CK, Hawley RS. Regulating the construc-
6. Koubova J, Hu YC, Bhattacharyya T et al. Retinoic tion and demolition of the synaptonemal complex.
acid activates two pathways required for meiosis in Nat Struct Mol Biol. 2016;23(5):369–377.
mice. PLoS Genet. 2014;10(8):e1004541. 23. Moses MJ. Chromosomal structures in crayfish sper-
7. Bowles J, Feng CW, Spiller C et al. FGF9 suppresses matocytes. J Biophys Biochem Cytol. 1956;2(2):215–218.
meiosis and promotes male germ cell fate in mice. 24. Schucker K, Holm T, Franke C et al. Elucidation
Dev Cell. 2010;19(3):440–449. of synaptonemal complex organization by super-
8. Suzuki A, Hirasaki M, Hishida T et al. Loss of MAX resolution imaging with isotropic resolution. Proc
results in meiotic entry in mouse embryonic and Natl Acad Sci U S A. 2015;112(7):2029–2033.
germline stem cells. Nat Commun. 2016;7:11056. 25. Hernandez-Hernandez A, Masich S, Fukuda T et al.
9. Abby E, Tourpin S, Ribeiro J et al. Implementation The central element of the synaptonemal complex in
of meiosis prophase I programme requires a con- mice is organized as a bilayered junction structure.
served retinoid-independent stabilizer of meiotic J Cell Sci. 2016;129(11):2239–2249.
transcripts. Nat Commun. 2016;7:10324. 26. Schalk JA, Dietrich AJ, Vink AC et al. Localization
10. Soh YQS, Mikedis MM, Kojima M et al. Meioc main- of SCP2 and SCP3 protein molecules within syn-
tains an extended meiotic prophase I in mice. PLoS aptonemal complexes of the rat. Chromosoma.
Genet. 2017;13(4):e1006704. 1998;107(8):540–548.
11. Hsu PJ, Zhu Y, Ma H et al. YTHDC2 regulates sper- 27. Offenberg HH, Schalk JA, Meuwissen RL et al. SCP2:
matogenesis through promoting the translation of A major protein component of the axial elements of
N6-methyladenosine-modified RNA. FASEB J. 2017;​ synaptonemal complexes of the rat. Nucleic Acids
31:595–510. Res. 1998;26(11):2572–2579.
12. Reig-Viader R, Garcia-Caldes M, Ruiz-Herrera A. 28. Yang F, De La Fuente R, Leu NA et al. Mouse SYCP2
Telomere homeostasis in mammalian germ cells: A is required for synaptonemal complex assembly and
review. Chromosoma. 2016;125(2):337–351. chromosomal synapsis during male meiosis. J Cell
13. Lee CY, Horn HF, Stewart CL et al. Mechanism Biol. 2006;173(4):497–507.
and regulation of rapid telomere prophase move- 29. Yuan L, Liu JG, Zhao J et al. The murine SCP3 gene
ments in mouse meiotic chromosomes. Cell Rep. is required for synaptonemal complex assembly,
2015;11(4):551–563. chromosome synapsis, and male fertility. Mol Cell.
14. Ding X, Xu R, Yu J et al. SUN1 is required for telo- 2000;5(1):73–83.
mere attachment to nuclear envelope and gameto- 30. Bolcun-Filas E, Hall E, Speed R et al. Mutation of
genesis in mice. Dev Cell. 2007;12(6):863–872. the mouse Syce1 gene disrupts synapsis and sug-
15. Horn HF, Kim DI, Wright GD et al. A mammalian gests a link between synaptonemal complex struc-
KASH domain protein coupling meiotic chromosomes tural components and DNA repair. PLoS Genet.
to the cytoskeleton. J Cell Biol. 2013;202(7):​1023–1039. 2009;5(2):e1000393.
16. Shibuya H, Hernandez-Hernandez A, Morimoto A 31. Bolcun-Filas E, Costa Y, Speed R et al. SYCE2 is
et al. MAJIN links telomeric DNA to the nuclear required for synaptonemal complex assembly, dou-
membrane by exchanging telomere cap. Cell. ble strand break repair, and homologous recombina-
2015;163(5):1252–1266. tion. J Cell Biol. 2007;176(6):741–747.
17. Shibuya H, Ishiguro K, Watanabe Y. The TRF1- 32. Schramm S, Fraune J, Naumann R et al. A novel
binding protein TERB1 promotes chromosome mouse synaptonemal complex protein is essential for
movement and telomere rigidity in meiosis. Nat Cell loading of central element proteins, recombination,
Biol. 2014;16(2):145–156. and fertility. PLoS Genet. 2011;7(5):e1002088.
18. Tu Z, Bayazit MB, Liu H et al. Speedy A-Cdk2 33. Hamer G, Wang H, Bolcun-Filas E et al. Progression
binding mediates initial telomere-nuclear envelope of meiotic recombination requires structural matu-
attachment during meiotic prophase I indepen- ration of the central element of the synaptonemal
dent of Cdk2 activation. Proc Natl Acad Sci U S A. complex. J Cell Sci. 2008;121(Pt 15):2445–2451.
2017;114(3):592–597. 34. de Vries FA, de Boer E, van den Bosch M et al. Mouse
19. Mikolcevic P, Isoda M, Shibuya H et al. Essential role of Sycp1 functions in synaptonemal complex assembly,
the Cdk2 activator RingoA in meiotic telomere tether- meiotic recombination, and XY body formation.
ing to the nuclear envelope. Nat Commun. 2016;7:11084. Genes Dev. 2005;19(11):1376–1389.
References 113

35. Seo EK, Choi JY, Jeong JH et al. Crystal structure 51. Gomez R, Felipe-Medina N, Ruiz-Torres M et al.
of C-terminal coiled-coil domain of SYCP1 reveals Sororin loads to the synaptonemal complex central
non-canonical anti-parallel dimeric structure of region independently of meiotic cohesin complexes.
transverse filament at the synaptonemal complex. EMBO Rep. 2016;17(5):695–707.
PLoS One. 2016;11(8):e0161379. 52. Brieno-Enriquez MA, Moak SL, Toledo M et al.
36. Syrjanen JL, Pellegrini L, Davies OR. A molecular Cohesin removal along the chromosome arms dur-
model for the role of SYCP3 in meiotic chromosome ing the first meiotic division depends on a NEK1-
organisation. Elife. 2014;3:10.7554/eLife.02963. PP1gamma-WAPL axis in the mouse. Cell Rep. 2016;​
37. Lu J, Gu Y, Feng J et al. Structural insight into the 17(4):977–986.
central element assembly of the synaptonemal com- 53. Hunt PA, Hassold TJ. Sex matters in meiosis. Science.
plex. Sci Rep. 2014;4:7059. 2002;296(5576):2181–2183.
38. Xu H, Beasley MD, Warren WD et al. Absence of 54. Turner JM. Meiotic silencing in mammals. Annu Rev
mouse REC8 cohesin promotes synapsis of sister Genet. 2015;49:395–412.
chromatids in meiosis. Dev Cell. 2005;8(6):949–961. 55. Keeney S, Giroux CN, Kleckner N. Meiosis-specific
39. Ishiguro K, Kim J, Fujiyama-Nakamura S et al. A DNA double-strand breaks are catalyzed by Spo11,
new meiosis-specific cohesin complex implicated a member of a widely conserved protein family. Cell.
in the cohesin code for homologous pairing. EMBO 1997;88(3):375–384.
Rep. 2011;12(3):267–275. 56. Romanienko PJ, Camerini-Otero RD. The mouse
40. Lee J, Hirano T. RAD21L, a novel cohesin subunit Spo11 gene is required for meiotic chromosome syn-
implicated in linking homologous chromosomes in apsis. Mol Cell. 2000;6(5):975–987.
mammalian meiosis. J Cell Biol. 2011;192(2):263–276. 57. Baudat F, Manova K, Yuen JP et al. Chromosome synap-
41. Herran Y, Gutierrez-Caballero C, Sanchez-Martin sis defects and sexually dimorphic meiotic progression
M et al. The cohesin subunit RAD21L functions in in mice lacking Spo11. Mol Cell. 2000;6(5):989–998.
meiotic synapsis and exhibits sexual dimorphism in 58. Robert T, Nore A, Brun C et al. The TopoVIB-Like pro-
fertility. EMBO J. 2011;30(15):3091–3105. tein family is required for meiotic DNA double-strand
42. Revenkova E, Eijpe M, Heyting C et al. Cohesin SMC1 break formation. Science. 2016;351(6276):943–949.
beta is required for meiotic chromosome dynamics, 59. Libby BJ, Reinholdt LG, Schimenti JC. Positional
sister chromatid cohesion and DNA recombination. cloning and characterization of Mei1, a vertebrate-
Nat Cell Biol. 2004;6(6):555–562. specific gene required for normal meiotic chromo-
43. Prieto I, Suja JA, Pezzi N et al. Mammalian STAG3 is some synapsis in mice. Proc Natl Acad Sci U S A.
a cohesin specific to sister chromatid arms in meiosis 2003;100(26):15706–15711.
I. Nat Cell Biol. 2001;3(8):761–766. 60. Kumar R, Bourbon HM, de Massy B. Functional
44. Agostinho A, Manneberg O, van Schendel R et al. conservation of Mei4 for meiotic DNA double-
High density of REC8 constrains sister chromatid strand break formation from yeasts to mice. Genes
axes and prevents illegitimate synaptonemal com- Dev. 2010;24(12):1266–1280.
plex formation. EMBO Rep. 2016;17(6):901–913. 61. Shin YH, Choi Y, Erdin SU et al. Hormad1 mutation
45. Biswas U, Hempel K, Llano E et al. Distinct roles disrupts synaptonemal complex formation, recombi-
of meiosis-specific cohesin complexes in mam- nation, and chromosome segregation in mammalian
malian spermatogenesis. PLoS Genet. 2016;12(10):​ meiosis. PLoS Genet. 2010;6(11):e1001190.
e1006389. 62. Daniel K, Lange J, Hached K et al. Meiotic homo-
46. Ward A, Hopkins J, Mckay M et al. Genetic interac- logue alignment and its quality surveillance are
tions between the meiosis-specific cohesin compo- controlled by mouse HORMAD1. Nat Cell Biol.
nents, STAG3, REC8, and RAD21L. G3 (Bethesda). 2011;13(5):599–610.
2016;6(6):1713–1724. 63. Stanzione M, Baumann M, Papanikos F et al. Meiotic
47. Sourirajan A, Lichten M. Polo-like kinase Cdc5 DNA break formation requires the unsynapsed
drives exit from pachytene during budding yeast chromosome axis-binding protein IHO1 (CCDC36)
meiosis. Genes Dev. 2008;22(19):2627–2632. in mice. Nat Cell Biol. 2016;18(11):1208–1220.
48. Jordan PW, Karppinen J, Handel MA. Polo-like 64. Kumar R, Ghyselinck N, Ishiguro K et al. MEI4—A
kinase is required for synaptonemal complex dis- central player in the regulation of meiotic DNA double-
assembly and phosphorylation in mouse spermato- strand break formation in the mouse. J Cell Sci.
cytes. J Cell Sci. 2012;125(Pt 21):5061–5072. 2015;128(9):1800–1811.
49. Nishiyama T, Ladurner R, Schmitz J et al. Sororin 65. Burma S, Chen BP, Murphy M et al. ATM phosphory-
mediates sister chromatid cohesion by antagonizing lates histone H2AX in response to DNA double-strand
Wapl. Cell. 2010;143(5):737–749. breaks. J Biol Chem. 2001;276(45):42462–42467.
50. Kuleszewicz K, Fu X, Kudo NR. Cohesin loading 66. Lange J, Pan J, Cole F et al. ATM controls meiotic
factor Nipbl localizes to chromosome axes during double-strand-break formation. Nature. 2011;479​
mammalian meiotic prophase. Cell Div. 2013;8(1):12. (7372):​237–240.
114  Genetics of mammalian meiosis

67. Neale MJ, Pan J, Keeney S. Endonucleolytic process- 82. Baker SM, Plug AW, Prolla TA et al. Involvement of
ing of covalent protein-linked DNA double-strand mouse Mlh1 in DNA mismatch repair and meiotic
breaks. Nature. 2005;436(7053):1053–1057. crossing over. Nat Genet. 1996;13(3):336–342.
68. Wold MS. Replication protein A: A heterotrimeric, 83. Rogacheva MV, Manhart CM, Chen C et al. Mlh1-
single-stranded DNA-binding protein required for Mlh3, a meiotic crossover and DNA mismatch repair
eukaryotic DNA metabolism. Annu Rev Biochem. factor, is a Msh2-Msh3-stimulated endonuclease.
1997;66:61–92. J Biol Chem. 2014;289(9):5664–5673.
69. Brown MS, Bishop DK. DNA strand exchange and 84. Wei K, Clark AB, Wong E et al. Inactivation
RecA homologs in meiosis. Cold Spring Harb Perspect of Exonuclease 1 in mice results in DNA mis-
Biol. 2014;7(1):a016659. match repair defects, increased cancer suscepti-
70. Bishop DK, Park D, Xu L, Kleckner N. DMC1:  A bility, and male and female sterility. Genes Dev.
­meiosis-specific yeast homolog of E. coli recA required 2003;17(5):603–614.
for recombination, synaptonemal complex formation, 85. Schaetzlein S, Chahwan R, Avdievich E et al.
and cell cycle progression. Cell. 1992;69(3):439–456. Mammalian Exo1 encodes both structural and
71. Petukhova GV, Pezza RJ, Vanevski F et al. The Hop2 catalytic functions that play distinct roles in essen-
and Mnd1 proteins act in concert with Rad51 and tial biological processes. Proc Natl Acad Sci U S A.
Dmc1 in meiotic recombination. Nat Struct Mol Biol. 2013;110(27):E2470–E2479.
2005;12(5):449–453. 86. Qiao H, Prasada Rao HB, Yang Y et al. Antagonistic
72. Finsterbusch F, Ravindranathan R, Dereli I et al. roles of ubiquitin ligase HEI10 and SUMO ligase
Alignment of homologous chromosomes and effec- RNF212 regulate meiotic recombination. Nat Genet.
tive repair of programmed DNA double-strand breaks 2014;46(2):194–199.
during mouse meiosis require the minichromosome 87. Rao HB, Qiao H, Bhatt SK et al. A SUMO-ubiquitin
maintenance domain containing 2 (MCMDC2) pro- relay recruits proteasomes to chromosome axes to reg-
tein. PLoS Genet. 2016;12(10):e1006393. ulate meiotic recombination. Science. 2017;355(6323):​
73. McNairn AJ, Rinaldi VD, Schimenti JC. Repair of 403–407.
meiotic DNA breaks and homolog pairing in mouse 88. Ahuja JS, Sandhu R, Mainpal R et al. Control of meiotic
meiosis requires a minichromosome maintenance pairing and recombination by chromosomally teth-
(MCM) paralog. Genetics. 2017;205(2):529–537. ered 26S proteasome. Science. 2017;355(6323):408–411.
74. Luo M, Yang F, Leu NA et al. MEIOB exhibits single- 89. Holloway JK, Sun X, Yokoo R et al. Mammalian
stranded DNA-binding and exonuclease activi- CNTD1 is critical for meiotic crossover maturation
ties and is essential for meiotic recombination. Nat and deselection of excess precrossover sites. J Cell
Commun. 2013;4:2788. Biol. 2014;205(5):633–641.
75. Souquet B, Abby E, Herve R et al. MEIOB targets 90. Yang F, Silber S, Leu NA et al. TEX11 is mutated
single-strand DNA and is necessary for meiotic in infertile men with azoospermia and regulates
recombination. PLoS Genet. 2013;9(9):e1003784. genome-wide recombination rates in mouse. EMBO
76. La Salle S, Palmer K, O’Brien M et al. Spata22, a novel Mol Med. 2015;7(9):1198–1210.
vertebrate-specific gene, is required for meiotic prog- 91. Kong A, Thorleifsson G, Stefansson H et al. Sequence
ress in mouse germ cells. Biol Reprod. 2012;86(2):45. variants in the RNF212 gene associate with genome-
77. Xu Y, Greenberg RA, Schonbrunn E, Wang PJ. wide recombination rate. Science. 2008;319(5868):​
Meiosis-specific proteins MEIOB and SPATA22 1398–1401.
cooperatively associate with the ssDNA-binding 92. Holloway JK, Booth J, Edelmann W et al. MUS81
RPA complex and DNA double-strand breaks. Biol generates a subset of MLH1-MLH3-independent
Reprod. 2017;96(5):1096–1104. crossovers in mammalian meiosis. PLoS Genet.
78. Ishishita S, Matsuda Y, Kitada K. Genetic evidence 2008;4(9):e1000186.
suggests that Spata22 is required for the mainte- 93. Baudat F, Imai Y, de Massy B. Meiotic recombination
nance of Rad51 foci in mammalian meiosis. Sci Rep. in mammals: Localization and regulation. Nat Rev
2014;4:6148. Genet. 2013;14(11):794–806.
79. Snowden T, Acharya S, Butz C et al. hMSH4-hMSH5 94. Parvanov ED, Petkov PM, Paigen K. Prdm9 controls
recognizes Holliday junctions and forms a meiosis- activation of mammalian recombination hotspots.
specific sliding clamp that embraces homologous Science. 2010;327(5967):835.
chromosomes. Mol Cell. 2004;15(3):437–451. 95. Myers S, Bowden R, Tumian A et al. Drive
80. Yang F, Gell K, van der Heijden GW et al. Meiotic against hotspot motifs in primates implicates the
failure in male mice lacking an X-linked factor. PRDM9 gene in meiotic recombination. Science.
Genes Dev. 2008;22(5):682–691. 2010;327(5967):876–879.
81. Edelmann W, Cohen PE, Kane M et al. Meiotic 96. Baudat F, Buard J, Grey C et al. PRDM9 is a major
pachytene arrest in MLH1-deficient mice. Cell. determinant of meiotic recombination hotspots in
1996;85(7):1125–1134. humans and mice. Science. 2010;327(5967):836–840.
References 115

97. Hayashi K, Yoshida K, Matsui Y. A histone H3 meth- cell lines using an organ culture method. Nat Protoc.
yltransferase controls epigenetic events required for 2013;8(11):2098–2104.
meiotic prophase. Nature. 2005;438(7066):374–378. 114. Zhou Q, Wang M, Yuan Y et al. Complete meio-
98. Eram MS, Bustos SP, Lima-Fernandes E et al. sis from embryonic stem cell-derived germ cells in
Trimethylation of histone H3 lysine 36 by human vitro. Cell Stem Cell. 2016;18(3):330–340.
methyltransferase PRDM9 protein. J Biol Chem. 115. Yokonishi T, Sato T, Katagiri K et al. In vitro recon-
2014;289(17):12177–12188. struction of mouse seminiferous tubules supporting
99. Powers NR, Parvanov ED, Baker CL et al. The mei- germ cell differentiation. Biol Reprod. 2013;89(1):15.
otic recombination activator PRDM9 Trimethylates 116. Wang S, Wang X, Ma L et al. Retinoic acid is suf-
both H3K36 and H3K4 at recombination hotspots in ficient for the in vitro induction of mouse spermato-
vivo. PLoS Genet. 2016;12(6):e1006146. cytes. Stem Cell Rep. 2016;7(1):80–94.
100. Hu J, Donahue G, Dorsey J et al. H4K44 Acetylation 117. Yatsenko AN, Georgiadis AP, Ropke A et al. X-linked
facilitates chromatin accessibility during meiosis. TEX11 mutations, meiotic arrest, and azoospermia in
Cell. Rep 2015;13(9):1772–1780. infertile men. N Engl J Med. 2015;372(22):2097–2107.
101. Brick K, Smagulova F, Khil P et al. Genetic recom- 118. Fu Q, Wang PJ. Mammalian piRNAs: Biogenesis,
bination is directed away from functional genomic function, and mysteries. Spermatogenesis. 2014;​
elements in mice. Nature. 2012;485(7400):642–645. 4:e27889.
102. Ponting CP. What are the genomic drivers of 119. Siomi MC, Sato K, Pezic D, Aravin AA. PIWI-
the rapid evolution of PRDM9? Trends Genet. interacting small RNAs: The vanguard of genome
2011;27(5):165–171. defence. Nat Rev Mol Cell Biol. 2011;12(4):246–258.
103. Davies B, Hatton E, Altemose N et al. Re-engineering 120. Gershoni M, Hauser R, Yogev L et al. A familial study
the zinc fingers of PRDM9 reverses hybrid sterility in of azoospermic men identifies three novel causative
mice. Nature. 2016;530(7589):171–176. mutations in three new human azoospermia genes.
104. Mihola O, Trachtulec Z, Vlcek C et al. A mouse spe- Genet Med. 2017;19(9):998.
ciation gene encodes a meiotic histone H3 methyl- 121. Carlosama C, El Zaiat M, Patino LC et al. A homozy-
transferase. Science. 2009;323(5912):373–375. gous donor splice-site mutation in the meiotic gene
105. Baker CL, Walker M, Kajita S et al. PRDM9 binding MSH4 causes primary ovarian insufficiency. Hum
organizes hotspot nucleosomes and limits Holliday Mol Genet. 2017.
junction migration. Genome Res. 2014;24(5):​724–732. 122. Zangen D, Kaufman Y, Zeligson S et al. XX ovarian
106. Imai Y, Baudat F, Taillepierre M et al. The PRDM9 dysgenesis is caused by a PSMC3IP/HOP2 mutation
KRAB domain is required for meiosis and involved that abolishes coactivation of estrogen-driven tran-
in protein interactions. Chromosoma. 2017. scription. Am J Hum Genet. 2011;89(4):572–579.
107. Parvanov ED, Tian H, Billings T et al. PRDM9 inter- 123. de Vries L, Behar DM, Smirin-Yosef P et al. Exome
actions with other proteins provide a link between sequencing reveals SYCE1 mutation associated with
recombination hotspots and the chromosomal axis autosomal recessive primary ovarian insufficiency.
in meiosis. Mol Biol Cell. 2017;28(3):488–499. J Clin Endocrinol Metab. 2014;99(10):E2129–32.
108. Handel MA, Eppig JJ, Schimenti JC. Applying “gold 124. Maor-Sagie E, Cinnamon Y, Yaacov B et al.
standards” to in-vitro-derived germ cells. Cell. Deleterious mutation in SYCE1 is associated with
2014;157(6):1257–1261. non-obstructive azoospermia. J Assist Reprod Genet.
109. Russell LD, Ettlin RA, Sinha Hikim AP, Clegg ED. 2015;32(6):887–891.
Histological and histopathological evaluation of the 125. Miyamoto T, Hasuike S, Yogev L et al. Azoospermia
testis. Clearwater, FL: Cache River Press;1990. in patients heterozygous for a mutation in SYCP3.
110. Steinberger A, Steinberger E. Differentiation of rat Lancet. 2003;362(9397):1714–1719.
seminiferous epithelium in organ culture. J Reprod 126. Caburet S, Arboleda VA, Llano E et al. Mutant
Fertil. 1965;9:243–248. cohesin in premature ovarian failure. N Engl J Med.
111. Sato T, Katagiri K, Gohbara A et al. In vitro produc- 2014;370(10):943–949.
tion of functional sperm in cultured neonatal mouse 127. Okutman O, Muller J, Baert Y et al. Exome sequenc-
testes. Nature. 2011;471(7339):504–507. ing reveals a nonsense mutation in TEX15 causing
112. Sato T, Katagiri K, Yokonishi T et al. In vitro produc- spermatogenic failure in a Turkish family. Hum Mol
tion of fertile sperm from murine spermatogonial Genet. 2015;24(19):5581–5588.
stem cell lines. Nat Commun. 2011;2:472. 128. Colombo R, Pontoglio A, Bini M. Two novel TEX15
113. Sato T, Katagiri K, Kubota Y, Ogawa T. In vitro mutations in a family with nonobstructive azoosper-
sperm production from mouse spermatogonial stem mia. Gynecol Obstet Invest. 2017;82(3):283–286.
Roles of membrane and nuclear
estrogen receptors in spermatogenesis
9
PAUL S. COOKE, MANJUNATHA K. NANJAPPA, SERGEI G. TEVOSIAN,
and REX A. HESS

INTRODUCTION Other cells also contribute to the SSC niche through


Androgens are essential for spermatogenesis, and exten- production of growth factors that affect SSC proliferation
sive progress has been made in understanding androgen and differentiation. For example, SSCs are preferentially
signaling in different testicular cell types. In contrast, located near vascular elements outside the seminiferous
estrogen’s roles in the testis and spermatogenesis are less tubule,6 suggesting that testicular vasculature and/or asso-
clear. Estrogens are present systemically in males and tes- ciated Leydig cells also regulate SSCs. Furthermore, auto-
ticular estrogen concentrations are higher than peripheral crine actions of androgen in Leydig cells are required for
concentrations due to testicular estrogen production. In normal testicular function.7
addition, estrogen receptors are widely distributed in the Similar to androgens, estrogens affect male reproduc-
male reproductive tract. Despite extensive literature show- tion but the mechanisms of these effects are unclear.
ing deleterious effects of perinatal estrogen administration Estrogens are produced in the testis and estrogen recep-
on the testis and spermatogenesis, estrogen’s role in the tors (ERs) are present in many testicular cell types, but
normal testis and spermatogenesis, especially in humans, gaps remain in understanding estrogen’s role in spermato-
remains less clear. genesis. This chapter reviews testicular estrogen signaling
Before considering testicular estrogen effects, we briefly and also discusses how a lack of estrogen signaling affects
review androgen effects on various testicular cell types to other male reproductive organs, resulting in secondary
contrast the discrepancy in information regarding the two impairments in spermatogenesis and fertility.
areas. Androgens are obligatory for spermatogenesis, and
testicular androgen effects are mediated through many cell ESTROGEN SOURCES IN MALES
types. Sertoli cells are a major androgen target and adult Estrogen regulation of male reproduction is now well rec-
Sertoli cells express abundant androgen receptor (AR), but ognized. Estrogens were first reported in males (stallions)
Sertoli cells during development lack AR and only begin to more than 80 years ago.8 Decades later, radioimmuno-
show slight AR immunostaining at about day 12 in mouse assay methodologies established that men and males of
testis.1 In a conditional knockout of AR in Sertoli cells,2 other species had measureable circulating 17β-estradiol
spermatogenesis proceeded only up to meiosis, indicating (E2) concentrations. Testosterone (T) concentrations in
an obligatory dependence for androgen signaling through peripheral blood of men are typically two orders of mag-
Sertoli cells for all but the earliest spermatogenic stages. In nitude greater than E2 concentrations, indicating the pri-
these mice, there are increased adult Sertoli cell numbers, macy of T compared to E2 for male reproduction. However,
indicating that signaling through AR normally inhibits E2 concentrations of approximately 30–200 pmol/L have
Sertoli cell proliferation.3 been reported in men,9 which are several-fold less than E2
In contrast to somatic cells, developing testicular germ concentrations reported in women during certain phases
cells lack AR and germ cell-specific AR knockout mice of the menstrual cycle but greater than those for ovariec-
are fertile with normal spermatogenesis.4 Thus, andro- tomized female rats, rats in diestrus, and postmenopausal
gen actions on spermatogenesis are mediated indirectly women.
through effects on Sertoli cells and other cells that have Ovaries are the major source of circulating estrogens in
paracrine effects on germ cells. females. However, in males, testes produce only about 20%
Peritubular myoid cells are separated from spermato- of circulating estrogens, with the remainder produced
gonial stem cells (SSCs) within the seminiferous tubule by by adipose, adrenals, brain, skin, bone, and other organs
only the basement membrane of the seminiferous epithe- expressing aromatase (Cyp19a1), a ubiquitous NADPH-
lium. This suggests they may communicate with SSCs as dependent cytochrome P450 reductase enzyme.10 Local
well as other germ and somatic cells. A peritubular cell- testicular estrogen production leads to estrogen concen-
specific AR knockout showed progressive spermatogonial trations in testis and seminiferous tubule fluid that exceed
loss beginning at weaning, resulting in disorganized semi- circulating systemic levels. In monkeys, E2 concentra-
niferous epithelium and adult infertility5 and supporting tions of 40–145 pmol/L have been reported in peripheral
the idea that androgen actions through peritubular cells blood.11 Concentrations in the testicular vein and rete tes-
regulate spermatogenesis. tis (RT) are even higher (up to 640 pmol/L), illustrating

116
Estrogen sources in males  117

both testicular E2 production and the potential for tes- originally known as G protein-coupled receptor 30) are
ticular cells to be exposed to high E2 concentrations.11 expressed in testis and other male reproductive organs.
Although data are not available on E2 concentrations in Although most ESR1 and ESR2 are cytoplasmic/nuclear,
the testicular vein and RT of men, there are reports of a small fraction of ESR1 and ESR2, as well as all of GPER,
E2 concentrations in human semen of up to 520 pmol/L are localized in the cell membrane, where these membrane
(reviewed in Cooke et al.12). Similar increases in testicular receptors mediate rapid estrogen actions.
vein E2 vs. systemic E2 concentrations have been observed Although there are species-specific differences, ESR1
in other species (reviewed in Cooke et al.12). Thus, cells of expression has typically been reported in Leydig and peri-
the testis and other organs that come into direct contact tubular cells in mice and other mammals.17 Expression
with seminiferous tubule fluid and the developing ejacu- of ESR1 mRNA and protein expression in Sertoli cells
late (RT, efferent ductules, epididymis, ductus deferens, has been reported, but it has not been universally con-
urethra) can be exposed to high estrogen concentrations. firmed.18–20 Testicular ESR2 expression is more wide-
In the testis, aromatase is necessary for estrogen pro- spread, with expression in Leydig, peritubular, germ, and
duction and is expressed in Leydig (Figure 9.1) and imma- Sertoli cells.21,22 Efferent ductules express both ESR1 and
ture Sertoli cells.13,14 Sertoli cells were initially considered ESR2, with ESR1 being especially abundant. Likewise,
the sole source of aromatase/estrogen in seminiferous epi- epididymis and ductus deferens express ESR1 and ESR2,
thelia, but aromatase was identified in round spermatids but as in other organs, expression patterns are unique and
and in elongating and late spermatids in the early 1990s.15 regionally variable.
Germ cell aromatase expression, originally described in The pattern of ER expression differs in primates com-
rodents,15 was subsequently reported in men as well as pared to other animals (reviewed in Cooke et al.12) fur-
other mammalian and avian species.12 Germ cells appear ther complicating understanding ER’s role in the testis and
to be the major testicular source or aromatase and thus male tract. For example, ESR1 expression was minimal in
testicular E2.16 There are reports of epididymal aromatase primate compared to rodent testis. Varying patterns of
expression, which could contribute estrogen to the devel- ESR1 and ESR2 expression in other species have also been
oping ejaculate. Thus, in many species estrogens produced reported (reviewed in Cooke et al.12), which in addition to
in testis and epididymis can signal through estrogen actual differences in ER expression may reflect differences
receptor 1 (ESR1) in the male tract. in antibody specificity, fixation, and animal ages as well as
species variations.
ER expression in male reproductive organs Expression of GPER in various testicular and other
Various types of ERs, including ESR1, estrogen receptor 2 reproductive cell types, such as Sertoli cells, germ cells,
(ESR2), and G protein-coupled estrogen receptor (GPER, epididymis, and sperm, is well documented. Consistent
Testis
Lymphatics Efferent ducts
ESR1
ESR2
PM ESR2 GPER
GPER
ESR1
GPER GC ? NC CC
Arom
Blood
? T E2 E2
T E2
Arom ? AC
SC
LC ESR1
ESR1 ESR2 BC
ESR2 GPER
Epididymis
GPER
ESR1
ESR2
GPER
CL

Figure 9.1  Estrogen production and estrogen receptors in the male reproductive tract. In testis, Leydig cells and germ cells
(including sperm) express aromatase (pink) and convert testosterone (T) to 17β-estradiol (E2). ESR1 expression is low in testicular
cells, but ESR2 expression is higher and more ubiquitous. Both nuclear and membrane ERs are expressed throughout the male tract.
Specific targets for testicular E2 have not been established, but germ cell synthesis of E2 continues in the lumen of the reproductive
tract, where E2 affects the efferent ductules and epididymis as fluid transits through. The major target appears to be the efferent
ductules, where ESR1 concentrations are threefold greater than any other male or female tissue and both ciliated and nonciliated
cells are E2 targets. LC, Leydig cell; PM, peritubular myoid; SC, Sertoli cell; GC, germ cell; NC, nonciliated cell; CC, ciliated cell; AC, api-
cal cell (narrow); BC, basal cell; CL, clear cell; Arom, P450 aromatase.
118  Roles of membrane and nuclear estrogen receptors in spermatogenesis

with this, estrogen actions mediated partially or entirely nuclear receptors goes back to the 1950s, for years the iden-
through GPER are known.22–26 tity of these membrane receptors and their mechanism of
action remained unknown. A crucial step forward was the
Effects of early estrogen treatment in males demonstration that transfecting a plasmid encoding ESR1
Beginning in the 1970s, early treatment of mice with the into ESR1-deficient cells led to expression of both nuclear
potent synthetic estrogen diethylstilbestrol (DES) was and membrane ESR1.38,39 Furthermore, ESR1 antibodies
used to produce adult reproductive abnormalities. These showed both nuclear and cell membrane staining.39 These
included impaired spermatogenesis and other testicu- findings led to the conclusion that membrane estrogen
lar abnormalities, cryptorchid testes, epididymal cysts, receptors40 were identical to classical nuclear ESR1.
inflammatory orchitis and epididymitis, and accessory These findings raised questions concerning how ESR1
organ abnormalities, frequently accompanied by infertil- localized to cell membranes and signaled. Membrane
ity.27 Recent studies suggested that Sertoli cells may be a localization of ESR1 was found to require ESR1 pal-
major target for early DES effects on spermatogenesis.28 mitoylation at cysteine 451 in mice,41 which conferred
Studies using DES in animal models were partially driven hydrophobicity and allowed cell membrane localization.
by reports of a rare cancer in young women whose moth- Extensive work with various steroid hormone membrane
ers had taken DES during pregnancy.29 receptors established that they activate the phosphati-
dylinositol-3-kinase (PI3K) and mitogen-activated protein
ROLE OF E2/ESR1 IN MALE REPRODUCTION
kinase (MAPK) pathways41 and have other actions such as
Loss of ESR1 causes extensive male reproductive increasing cAMP and cytosolic Ca++.
defects Understanding the mechanism by which membrane
Despite E2 and ER presence in males and known deleteri- ESR1 (mESR1) localized to the membrane led to work
ous effects of early DES treatment, it was unclear for years involving cells that expressed an altered ESR1 in which
whether E2/ER signaling was important in normal male alanine was substituted for cysteine 451 (C451A) in mouse
reproduction or reproductive pathologies. Over the past 20 ESR1. Unlike cysteine, alanine cannot be palmitoylated.
years, development of transgenic mouse models in which This precludes cell membrane localization of ESR1 and
E2/ESR1 signaling was disrupted revealed that estrogens leads to a lack of mESR1 despite continued nuclear ESR1
are critical for normal development and function of male (nESR1) expression.37
reproductive and nonreproductive organs. Recently, this altered ESR1 molecule was used to
The first steroid hormone receptor knockout d ­ eveloped develop transgenic nuclear-only estrogen receptor 1
was the Esr1 knockout (Esr1KO) mouse.30 This mouse (NOER) mice.41,42 Mice with the C451A mutation in ESR1
and its subsequent iterations31 provided important infor- express normal amounts of functional nESR1, but mESR1
mation regarding E2/ESR1 signaling in male and female was essentially eliminated because ESR1 was not palmi-
reproduction. The infertility originally reported in toylated. The lack of mESR1 signaling resulted in female
Esr1KO males32 was subsequently shown to result in large infertility and other reproductive abnormalities.41,42 We
part from impaired efferent ductule and epididymal func- recently reported that male reproductive function was
tion.33,34 Specifically, impaired fluid resorption and pro- also altered dramatically in NOER mice.43 Reproductive
nounced luminal dilation in efferent ductules caused fluid abnormalities in NOER males include enlarged RT and
accumulation in RT and seminiferous tubules.33,34 Mice dilated efferent ductules with reduced epithelial height.
lacking aromatase and thus estrogens (aromatase knock- In addition, there were increased numbers of abnormal
out, Cyp19KO) showed progressive infertility with degen- seminiferous tubules and degeneration in the seminifer-
eration and apoptosis of round spermatids and Leydig ous tubule epithelium. Sperm production was decreased
cell hyperplasia,35 confirming that impaired E2/ESR1 85% by 8 months of age and sperm motility was also dras-
signaling led to reproductive abnormalities and infertil- tically reduced. Strikingly, over 95% of cauda epididymal
ity. However, Cyp19KO mice do not totally replicate the sperm had structural abnormalities. These abnormalities
Esr1KO phenotype. This suggests ligand-independent ER were absent in sperm maturing in the seminiferous epithe-
activation by dietary phytoestrogens or androgen metab- lium but were present when sperm reached the RT, indi-
olites may be sufficient to mitigate or delay pathological cating that sperm abnormalities arise following exposure
changes in Cyp19KO males. Although not conclusive, clin- to altered fluid environments in the seminiferous tubule
ical literature describing men lacking ESR1 or aromatase lumen and downstream. The NOER males also showed a
(reviewed in Cooke et al.12) largely confirmed results from progressive infertility. These findings revealed that loss of
mouse models. mESR1, despite functioning nESR1, caused severe struc-
tural and functional sperm abnormalities and infertility.
Loss of membrane ESR1 expression causes male Changes in NOER males were similar to those in Esr1KO
reproductive abnormalities males, and indicate that mESR1 expression is necessary
Expression of ESR1 is predominately cytoplasmic and for male43 as well as female41,42 fertility.
nuclear. However, approximately 5% of ESR1 localizes to In both NOER and Esr1KO males, impaired efferent
cell membranes.36,37 Although literature describing steroid ductule function leads to fluid accumulation and back-
hormone effects too rapid to be mediated through classical pressure, causing testicular and seminiferous epithelial
Role of E2/ESR1 in male reproduction  119

abnormalities. However, a critical question that remains is While pH changes may contribute to sperm defects in
whether lack of normal E2 signaling causes direct testicu- NOER mice,43 we also noted decreased Wnt4 expression
lar effects that contribute to Esr1KO and NOER testicular (Tevosian et al., unpublished). This is in agreement with
phenotypes, or whether changes in organs such as efferent previous results that showed Wnt signaling is required for
ductules are entirely responsible for Esr1KO and NOER epididymal sperm maturation (Koch et al.45; reviewed in
testicular phenotypes. De Robertis and Ploper46), but the epididymal role of Wnt4
Maintenance of epithelial morphological organization has not been explored.
and function in efferent ductules and fluid reabsorption in
the caput epididymis are ESR1-dependent, and a molecular Knockouts of ESR2 or GPER
mechanism to explain estrogen’s role in fluid reabsorption Despite widespread GPER expression in male reproduc-
and regulation of bicarbonate and pH balance in the male tive tracts, male fertility was not impaired in the four
reproductive tract was proposed.44 Briefly, Esr1KO efferent GPER knockouts reported (reviewed in Cooke et al.12),
ducts have reduced expression of proteins regulating fluid- suggesting that GPER is dispensible for fertility. Similar
ion dynamics, including the sodium/hydrogen exchanger to GPER, ESR2 is widely distributed in the testis and
3 (NHE3; SLC9A3), carbonic anhydrase II (CAR2), and male tract, but initial Esr2 knockouts (Esr2KOs) lacked
two water channels, aquaporin 1 and 9 (AQP1 and AQP9). the dramatic male reproductive defects of Esr1KO mice,47
RNA expression analysis in Esr1KO mice demonstrated although changes in Leydig cell numbers and germ cells
that ESR1 has a primary effect on Sls9a3 transcription were reported. Critically, Esr2KO mice were fertile,31
while decreased expression of Car2, Aqp1, and Aqp9 could confirming that ESR1 is the functionally dominant ER
be secondary to efferent ductule epithelium morphologi- in males, although important ESR2 actions in the semi-
cal defects.44 It was also determined that ESR1 is expressed niferous epithelium were recently described.48 Although
in the proximal epididymis and has a similar effect there. a new Esr2KO mouse developed to avoid complications
Thus, dysregulation of proteins that regulate pH balance arising from alternatively spliced ESR2 transcripts in the
results in a failure of Esr1KO epididymis to properly acid- original Esr2KO mouse showed no apparent reproductive/
ify the luminal milieu and causes defects in sperm intra- sperm changes, these mice were reported to have mating
cellular pH, maturation and motility.45 defects,49 although this remains controversial. Lack of
In agreement with previous data, we observed a strong severe reproductive abnormalities in Esr2KO males sug-
(~14-fold) downregulation of Slc9a3 RNA in NOER epidid- gests that ESR2, like GPER, has male reproductive actions,
ymis (Figure 9.2, Tevosian et al., unpublished) confirming but may not be essential for male reproductive function.
that mRNA signaling is required for Slc9a3 expression. This has led to work in this area preferentially focusing on
Aqp1 was also downregulated while expression of the ESR1 rather than GPER or ESR2.
ubiquitous Aqp4 was unchanged. Carbonic anhydrase 14
(Car14, CA-XIV), the primary carbonic anhydrase present Can environmental estrogens impact human
on the luminal surface in the proximal epididymis, was testicular development?
unchanged, and expression of Na + / HCO3− cotransporter Transgenic mice have allowed normal E2 functions in
SLC4A4 was slightly elevated (Figure 9.2). Collectively, males to be identified. This work has also shown that
these findings suggest that, similar to Esr1KO mice, disrupted E2 signaling secondarily affects spermatogen-
reduced expression of Slc9A3 and Aqp1 could be involved esis, as described above. There is also extensive litera-
in NOER male reproductive abnormalities. ture documenting effects of early DES exposure on male
reproductive tract development in animal models and
humans.27–29 Over the past two decades, extensive work
5
has documented estrogenic effects of many man-made
Fold-change relative to control

and natural chemicals in the environment. Given known


4 effects of xenoestrogens such as DES on male reproduc-
tion and the critical role of estrogen signaling in male
3 reproductive tracts, one important question is whether
exposure of men and males of other species to estrogenic
2
environmental chemicals significantly impacts male
1 reproduction.
Work by Niels Skakkebaek’s laboratory in the early
0 1990s suggested that sperm counts in men had dropped
4 3 4 4 1 4 50% over the previous half-century.50 This had worrisome
4a 9a r1 nt Aq
p
Aq
p
Slc Slc Ca W
implications for human fertility and led to studies world-
wide to further test this hypothesis. Subsequent work
Figure 9.2  Quantitative PCR analysis of gene expression largely confirmed the original observations, although
(Slc4a4, Slc9a3, Car14, Wnt4, Aqp1, and Aqp4) in adult NOER epi- regional variations exist.51
didymis. Results are mean ± SEM of the fold-change relative to Shortly after the suggestion of declining human sperm
the value in the WT control (set to 1) from at least n = 3 replicates. counts, Skakkebaek and Richard Sharpe published a
120  Roles of membrane and nuclear estrogen receptors in spermatogenesis

seminal paper expanding the initial hypothesis and offer- organs outside the testis, and loss of this E2/ESR1 signaling
ing a potential mechanistic explanation.52 These authors can produce secondary changes in the testis and spermato-
noted that in addition to falling sperm counts, reproduc- genesis. This field has undergone a total transformation in
tive problems such as cryptorchidism, hypospadias, and the past quarter-century, and while estrogen’s role in both
testicular cancer had increased in men. This suggested an the testis and other reproductive organs is now established,
environmental cause for these reproductive problems, an many critical questions remain in this evolving area.
idea supported by strikingly increased reproductive disor-
ders in Danish compared to Finnish men despite minimal ACKNOWLEDGMENTS
racial/ethnic differences in these two populations. This work was supported by a New Florida Scholar Boost
Based on these observations, they proposed the testicu- Award, a grant from the University of Florida, and NIH
lar dysgenesis syndrome (TDS) theory, which postulated grant HD087528 to PSC.
that abnormal early testis development was causing fall-
ing sperm counts and testicular pathologies. Furthermore, REFERENCES
these authors suggested that these abnormalities could 1. Tan KA, Turner KJ, Saunders PT et al. Androgen
involve Leydig and/or Sertoli cells and that environmental regulation of stage-dependent cyclin D2 expres-
estrogen exposure in male fetuses to environmental estro- sion in Sertoli cells suggests a role in modulating
gens could contribute to subsequent testicular abnormali- androgen action on spermatogenesis. Biol Reprod.
ties and decreased sperm production. 2005;72:1151–1160.
The TDS theory was problematical to test experimen- 2. De Gendt K, Swinnen JV, Saunders PT et al. A Sertoli
tally in humans, although long-term administration cell-selective knockout of the androgen receptor
of estrogen to adult males produces adverse testicular causes spermatogenic arrest in meiosis. Proc Natl
changes.53 Rat models showed that prenatal exposure to Acad Sci U S A. 2004;101:1327–1332.
environmental chemicals such as phthalates induced 3. O’Shaughnessy PJ, Monteiro A, Abel M. Testicular
abnormalities consistent with the TDS theory (increased development in mice lacking receptors for follicle
cryptorchidism and hypospadias, impaired spermatogen- stimulating hormone and androgen. PLoS One.
esis).54 These studies also suggested that impaired andro- 2012;7:e35136.
gen production might be one factor driving TDS.55 4. Tsai MY, Yeh SD, Wang RS et al. Differential effects
In contrast, a recent report that human fetal testes xeno- of spermatogenesis and fertility in mice lacking
grafted into nude mice did not show reduced T produc- androgen receptor in individual testis cells. Proc Natl
tion in response to DES administered to the host mouse56 Acad Sci U S A. 2006;103:18975–18980.
failed to provide data supportive of the TDS hypothesis. 5. Welsh M, Saunders PT, Atanassova N, Sharpe RM,
Furthermore, this same study suggested that human Smith LB. Androgen action via testicular peritubu-
Leydig cells lacked full-length ESR1 and thus would not lar myoid cells is essential for male fertility. FASEB J.
be direct targets of environmental estrogen exposure. 2009;23:4218–4230.
Although some earlier work reported that ESR1 was absent 6. Yoshida S, Sukeno M, Nabeshima Y. A vasculature-
in human testis, other groups reported ESR1 expression associated niche for undifferentiated spermatogonia
in Leydig and germ cells.57,58 More than two decades after in the mouse testis. Science. 2007;317:1722–1726.
original reports on falling human sperm counts and the 7. O’Hara L, McInnes K, Simitsidellis I et al. Autocrine
TDS hypothesis, the role of environmental estrogens in androgen action is essential for Leydig cell matura-
various reproductive pathologies remains unresolved. tion and function, and protects against late-onset
In summary, ESR1 is present in low concentrations in Leydig cell apoptosis in both mice and men. FASEB
human testis and varies significantly with age.59 Other spe- J. 2015;29:894–910.
cies show greater ESR1 expression. In addition to direct 8. Zondek B. Mass excretion of oestrogenic hormone in
estrogen effects on several testicular cell types, the potential the urine of the stallion. Nature. 1934;133:209–210.
for cross-talk between AR and ESR1 and 2 also increases 9. Sartorius G, Spasevska S, Idan A et al. Serum tes-
the complexity of this topic. This is illustrated by work with tosterone, dihydrotestosterone and estradiol con­
Sertoli cell lines in vitro, where low concentrations of E2 centrations in older men self-reporting very good
increase ESR2 but decrease AR, while T increased AR but health: The healthy man study. Clin Endocrinol (Oxf).
had no effect on ESR2.60 In efferent ductules, E2 induces 2012;77:755–763.
loss of ESR1 as well as AR but has no effect on ESR2; in 10. Vermeulen A, Kaufman JM, Goemaere S, van
contrast, T increases ESR1 and AR but has little effect on Pottelberg I. Estradiol in elderly men. Aging Male.
ESR2.61 Thus, the balance of estrogen/androgen and their 2002;5:98–102.
respective receptors appears to play a greater role in the 11. Waites GM, Einer-Jensen N. Collection and analysis
overall male reproductive physiology, which depends not of rete testis fluid from macaque monkeys. J Reprod
only on absolute levels of ER expression but also on devel- Fertil. 1974;41:505–508.
opmental stage. In addition to direct effects on spermato- 12. Cooke P, Nanjappa M, Ko C, Prins G, Hess R.
genesis, extensive literature documents estrogen effects on Estro­gens in male physiology. Physiol Rev. 2017; In Press.
References 121

13. Carreau S, Wolczynski S, Galeraud-Denis I. Aroma­ 28. Sharpe RM, Atanassova N, McKinnell C et al.
tase, oestrogens and human male reproduction. Philos Abnormalities in functional development of the
Trans R Soc Lond B Biol Sci. 2010;365:1571–1579. Sertoli cells in rats treated neonatally with diethyl-
14. Levallet J, Carreau S. In vitro gene expression of stilbestrol: A possible role for estrogens in Sertoli cell
aromatase in rat testicular cells. C R Acad Sci III. development. Biol Reprod. 1998;59:1084–1094.
1997;320(2):123–129. 29. Herbst AL, Ulfelder H, Poskanzer DC. Adenocar­
15. Nitta H, Bunick D, Hess RA et al. Germ cells of the cinoma of the vagina. Association of maternal stil-
mouse testis express P450 aromatase. Endocrinology. bestrol therapy with tumor appearance in young
1993;132:1396–1401. women. N Engl J Med. 1971;284:878–881.
16. Levallet J, Mittre H, Delarue B, Carreau S. Alternative 30. Lubahn DB, Moyer JS, Golding TS, Couse JF, Korach
splicing events in the coding region of the cyto- KS, Smithies O. Alteration of reproductive function
chrome P450 aromatase gene in male rat germ cells. but not prenatal sexual development after insertional
J Mol Endocrinol. 1998;20:305–312. disruption of the mouse estrogen receptor gene. Proc
17. Zhou Q, Nie R, Prins GS, Saunders PT, Natl Acad Sci U S A. 1993;90:11162–11166.
Katzenellenbogen BS, Hess RA. Localization of 31. Dupont S, Krust A, Gansmuller A, Dierich A,
androgen and estrogen receptors in adult male Chambon P, Mark M. Effect of single and compound
mouse reproductive tract. J Androl. 2002;23:870–881. knockouts of estrogen receptors alpha (ERalpha) and
18. Lin J, Zhu J, Li X et al. Expression of genomic func- beta (ERbeta) on mouse reproductive phenotypes.
tional estrogen receptor 1 in mouse Sertoli cells. Development. 2000;127:4277–4291.
Reprod Sci. 2014;21:1411–1422. 32. Eddy EM, Washburn TF, Bunch DO et al. Targeted
19. Lucas TF, Lazari MF, Porto CS. Differential role of disruption of the estrogen receptor gene in male
the estrogen receptors ESR1 and ESR2 on the regula- mice causes alteration of spermatogenesis and infer-
tion of proteins involved with proliferation and dif- tility. Endocrinology. 1996;137:4796–4805.
ferentiation of Sertoli cells from 15-day-old rats. Mol 33. Hess RA, Bunick D, Lee KH et al. A role for oes-
Cell Endocrinol. 2014;382:84–96. trogens in the male reproductive system. Nature.
20. Gunawan A, Kaewmala K, Uddin MJ et al. 1997;390:509–512.
Association study and expression analysis of porcine 34. Hess RA, Bunick D, Lubahn DB, Zhou Q, Bouma J.
ESR1 as a candidate gene for boar fertility and sperm Morphologic changes in efferent ductules and epi-
quality. Anim Reprod Sci. 2011;128:11–21. didymis in estrogen receptor-alpha knockout mice.
21. Saunders PT, Fisher JS, Sharpe RM, Millar MR. J Androl. 2000;21:107–121.
Expression of oestrogen receptor beta (ER beta) 35. Robertson KM, O’Donnell L, Jones ME et al.
occurs in multiple cell types, including some germ Impairment of spermatogenesis in mice lacking a
cells, in the rat testis. J Endocrinol. 1998;156:R13–R17. functional aromatase (cyp 19) gene. Proc Natl Acad
22. Hess RA, Fernandes SA, Gomes GR, Oliveira Sci U S A. 1999;96:7986–7991.
CA, Lazari MF, Porto CS. Estrogen and its recep- 36. Acconcia F, Ascenzi P, Fabozzi G, Visca P, Marino M.
tors in efferent ductules and epididymis. J Androl. S-palmitoylation modulates human estrogen receptor-­
2011;32:600–613. alpha functions. Biochem Biophys Res Commun.
23. Carreau S, Hess RA. Oestrogens and spermatogenesis. 2004;316:878–883.
Philos Trans R Soc Lond B Biol Sci. 2010;365:1517–1535. 37. Pedram A, Razandi M, Sainson RC, Kim JK, Hughes
24. Lucas TF, Royer C, Siu ER, Lazari MF, Porto CS. CC, Levin ER. A conserved mechanism for steroid
Expression and signaling of G protein-coupled receptor translocation to the plasma membrane.
estrogen receptor 1 (GPER) in rat Sertoli cells. Biol J Biol Chem. 2007;282:22278–22288.
Reprod. 2010;83:307–317. 38. Levin ER. Extranuclear estrogen receptor’s roles in
25. Chimento A, Sirianni R, Delalande C et al. 17beta- physiology: Lessons from mouse models. Am J Physiol
estradiol activates rapid signaling pathways Endocrinol Metab. 2014;307:E133–E140.
involved in rat pachytene spermatocytes apoptosis 39. Razandi M, Pedram A, Greene GL, Levin ER. Cell
through GPR30 and ERalpha. Mol Cell Endocrinol. membrane and nuclear estrogen receptors (ERs)
2010;320:136–144. originate from a single transcript: Studies of ERalpha
26. Gautier C, Barrier-Battut I, Guenon I, Goux D, and ERbeta expressed in Chinese hamster ovary
Delalande C, Bouraima-Lelong H. Implication of cells. Mol Endocrinol. 1999;13:307–319.
the estrogen receptors GPER, ESR1, ESR2 in post-­ 40. Szego CM, Davis JS. Adenosine 3’,5’-monophosphate
testicular maturations of equine spermatozoa. Gen in rat uterus: Acute elevation by estrogen. Proc Natl
Comp Endocrinol. 2016;233:100–108. Acad Sci U S A. 1967;58:1711–1718.
27. Bullock BC, Newbold RR, McLachlan JA. Lesions 41. Pedram A, Razandi M, Lewis M, Hammes S, Levin
of testis and epididymis associated with prenatal ER. Membrane-localized estrogen receptor α is
diethylstilbestrol exposure. Environ Health Perspect. required for normal organ development and func-
1988;77:29–31. tion. Dev Cell. 2014;29:482–490.
122  Roles of membrane and nuclear estrogen receptors in spermatogenesis

42. Adlanmerini M, Solinhac R, Abot A et al. Mutation 52. Sharpe RM, Skakkebaek NE. Are oestrogens involved
of the palmitoylation site of estrogen receptor α in in falling sperm counts and disorders of the male
vivo reveals tissue-specific roles for membrane versus reproductive tract? Lancet. 1993;341:1392–1395.
nuclear actions. Proc Natl Acad Sci U S A. 2014;111:​ 53. Leavy M, Trottmann M, Liedl B et al. Effects of ele-
E283–E290. vated beta-estradiol levels on the functional morphol-
43. Nanjappa MK, Hess RA, Medrano TI, Locker SH, ogy of the testis—New insights. Sci Rep. 2017;​7:39931.
Levin ER, Cooke PS. Membrane-localized estrogen 54. Fisher JS, Macpherson S, Marchetti N, Sharpe RM.
receptor 1 is required for normal male reproductive Human ‘testicular dysgenesis syndrome’: A possible
development and function in mice. Endocrinology. model using in-utero exposure of the rat to dibutyl
2016;157:2909–2919. phthalate. Hum Reprod. 2003;18:1383–1394.
44. Zhou Q, Clarke L, Nie R et al. Estrogen action 55. Sharpe RM, Skakkebaek NE. Testicular dysgenesis
and male fertility: Roles of the sodium/hydrogen syndrome: Mechanistic insights and potential new
exchanger-3 and fluid reabsorption in reproductive downstream effects. Fertil Steril. 2008;89:e33–e38.
tract function. Proc Natl Acad Sci U S A. 2001;98:​ 56. Mitchell RT, Sharpe RM, Anderson RA et al.
14132–14137. Diethylstilboestrol exposure does not reduce testos-
45. Koch S, Acebron SP, Herbst J, Hatiboglu G, Niehrs C. terone production in human fetal testis xenografts.
Post-transcriptional Wnt signaling governs epididy- PLoS One. 2013;8:e61726.
mal sperm maturation. Cell. 2015;163:​1225–1236. 57. Fietz D, Ratzenbock C, Hartmann K et al. Expression
46. De Robertis EM, Ploper D. Sperm motility requires pattern of estrogen receptors alpha and beta and
Wnt/GSK3 stabilization of proteins. Dev Cell. 2015;​ G-protein-coupled estrogen receptor 1 in the human
35:401–402. testis. Histochem Cell Biol. 2014;142:421–432.
47. Krege JH, Hodgin JB, Couse JF et al. Generation 58. Pelletier G, El-Alfy M. Immunocytochemical local-
and reproductive phenotypes of mice lacking ization of estrogen receptors alpha and beta in the
estrogen receptor beta. Proc Natl Acad Sci U S A. human reproductive organs. J Clin Endocrinol Metab.
1998;95:15677–15682. 2000;​85:4835–4840.
48. Kumar A, Dumasia K, Deshpande S, Balasinor NH. 59. Lucas TF, Siu ER, Esteves CA et al. 17beta-estradiol
Direct regulation of genes involved in sperm release induces the translocation of the estrogen receptors
by estrogen and androgen through their recep- ESR1 and ESR2 to the cell membrane, MAPK3/1
tors and coregulators. J Steroid Biochem Mol Biol. phosphorylation and proliferation of cultured imma-
2017;171:66–74. ture rat Sertoli cells. Biol Reprod. 2008;78:101–114.
49. Antal MC, Krust A, Chambon P, Mark M. Sterility 60. Sneddon SF, Walther N, Saunders PT. Expression
and absence of histopathological defects in nonre- of androgen and estrogen receptors in Sertoli
productive organs of a mouse ERbeta-null mutant. cells: Studies using the mouse SK11 cell line.
Proc Natl Acad Sci U S A. 2008;105:2433–2438. Endocrinology. 2005;146:5304–5312.
50. Carlsen E, Giwercman A, Keiding N, Skakkebaek 61. Oliveira CA, Mahecha GA, Carnes K, Prins GS,
NE. Evidence for decreasing quality of semen during Saunders PT, Franca LR et al. Differential hormonal
past 50 years. BMJ. 1992;305:609–613. regulation of estrogen receptors ERalpha and ERbeta
51. Swan SH, Elkin EP, Fenster L. The question of declin- and androgen receptor expression in rat efferent
ing sperm density revisited: An analysis of 101 stud- ductules. Reproduction. 2004;128:73–86.
ies published 1934–1996. Environ Health Perspect.
2000;108:961–966.
Regulation of fertility and infertility
in humans
10
NAHID PUNJANI, RYAN FLANNIGAN, and PETER N. SCHLEGEL

INTRODUCTION level in 12 years and second lowest in the last 30 years.6


Regulation of male fertility begins with the creation of The rates of infertility are difficult to assess but there are
primordial germ cells (PGCs) during embryonic devel- approximately 7%–18% of couples in the United States
opment. These PGCs enter a quiescent state and eventu- who fail to conceive within 12 months of attempting.7,8
ally differentiate into spermatogonia between birth and Internationally, an estimated 60–80 million couples
6 months of age. It is not until puberty where the pro- according to World Health Organization (WHO) suffer-
cess of spermatogenesis occurs to produce spermatozoa ing from infertility worldwide. These rates of infertility do
through a highly coordinated and evolutionarily con- not appear to have changed in recent decades9,10; however,
served process. This is the basis on which male fertility male reproductive health has been suggested to be decreas-
can be achieved. The spermatozoa must then be capable ing among European and Western men over the past few
of fertilizing the oocyte and transmitting its genetic mate- decades.11–14 Male factor infertility is present in 50% of
rial to create a zygote and enable appropriate embryonic these cases and affects 10% of North American men.10,15
and fetal development. Many intrinsic and extrinsic fac- These men have most often been characterized by semen
tors affect spermatogenesis and fertility. In this chapter, we parameters reflecting reduced concentration (oligozoo-
will provide a brief overview of the history and epidemiol- spermia or azoospermia), poor motility (asthenozoosper-
ogy of male fertility, discuss the process and regulation of mia), and abnormal morphology (teratozoospermia).10
spermatogenesis and spermiogenesis, and discuss clinical Interestingly, recent data and studies have suggested that
requirements, conditions, and treatments surrounding sperm counts are actually decreasing worldwide.16 While
male fertility. this has previously been somewhat controversial, recent
data have shown that since the 1970s, there has been a
History of fertility 50%–60% decline in North America, Europe, Australia,
Fertility requires gamete production in both male and and New Zealand.17 The etiology of this decline remains
females, fertilization, and successful pregnancy to term. unclear and is an area for further study.
Male gamete production occurs via spermatogenesis, ulti-
mately producing functionally competent spermatozoa. Spermatogenesis and spermiogenesis
Many anatomical and physiological factors are required During embryologic development, PGCs arise from the
for spermatogenesis, including seminiferous tubules con- extraembryonic tissues adjacent to the yolk sac. Migration
taining both somatic cells and germ cells that eventually to the gonadal ridge occurs at 3 to 5 weeks, and differen-
give rise to spermatozoa. Spermatozoa were first identi- tiation to gonocytes occurs via inducible factors mediated
fied to have an essential role for fertility in the 1600s, as by Sertoli cells (SCs).18,19 Gonocytes remain mitotically
proposed by van Leeuwenhoek.1 Sertoli cells are integral inactive until birth, where they differentiate into sper-
in coordinating spermatogenesis and the production of matogonia (SPG) within the seminiferous tubules over
sperm was later discovered by Enrico Sertoli in 1865.2 the following 6 months. This is mediated by a number of
Spermatogenesis was identified to be under hormonal factors, including stromal cell derived factor 1 (SDF-1),
regulation in the twentieth century when testosterone was activator protein-2 (AP-2), stem cell factor (SCF), growth
identified to influence sperm production3 as well as the and differentiation factor 3 (GDF3), retinoic acid, estra-
hypothalamic-pituitary-gonadal axis. This axis was rec- diol, and DNA methylation changes.20–29 SPGs remain qui-
ognized to play a pivotal role regulating spermatogenesis escent until 5–7 years of age, where proliferation occurs
via two key hormones—the follicle-stimulating hormone followed by a combination of proliferation and differentia-
(FSH) and luteinizing hormone (LH)—and was discov- tion during puberty with the onset of spermatogenesis.30,31
ered in the early 1900s.4 Over time, as forms of infertil- Spermatogenesis occurs in the seminiferous tubules
ity have been recognized, corresponding treatments have of the testes. Several cells are involved in the production
evolved as well, including the in vitro fertilization (IVF) of spermatozoa, including somatic cells (Leydig cells,
procedure first performed in humans in the 1970s.5 Sertoli cells, and peritubular myoid cells [PTMCs]), as
well as germ cells (spermatogonial stem cells [SSCs], type
Epidemiology of pregnancy and infertility A dark and pale spematogonia, type B spermatogonia,
Presently, in the United States, there are approximately primary and secondary spermatocytes, round and elon-
102.1 pregnancies per 1000 women aged 15–44, the lowest gating spermatids, and spermatozoa). This process begins

123
124  Regulation of fertility and infertility in humans

with the proliferation and differentiation of SSCs.32 SSCs where it combines with other secretions to form the ejacu-
first undergo a proliferative phase located adjacent to the late and is expelled through the urethral meatus during
basement membrane of seminiferous tubules.33 Here, ejaculation.46
there is initial division of SSC that produces two cells, one
of which functions to renew the SSC population of cells Role of SCs
(Adark spermatogonia) and the other (Apale spermato- SCs are somatic cells of the testis and are critical to the
gonia) commits to a path of differentiation transforming process of spermatogenesis.47 During spermatogenesis,
into a type B spermatogonia.34 This process is mediated SCs function differently depending on the stage of the
by SC factors, including glial-derived neurotrophic fac- maturing germ cell and spermatogenic cycle.48 SCs secrete
tor (GDNF) that promotes proliferation and self-renewal, numerous factors into the tubular fluid to interact with
while bone morphogenetic protein 4 (BMP4), retinoic acid, developing germ cells. These include androgen bind-
and Notch1/Jagged2 signaling promote differentiation.35–37 ing protein, GDNF, Transforming Growth Factor-alpha
This B spermatogonia undergoes a series of mitotic events (TGF-α), Transforming Growth Factor- beta (TGF-β),
to give rise to preleptotene spermatocytes (phase of pri- transferrin, Interleukin-1alpha (IL-1α), ceruloplasmin,
mary spermatocytes). These spermatocytes subsequently plasminogen activator, insulin-like growth factor, inhibin
enter meiosis.34 These preleptotene spermatocytes detach B, and anti-Mullerian hormone.29,35,49–56 The number of
from the basement membrane, cross the blood-testis bar- SCs is an important determinant of the number of sper-
rier (BTB) to the adluminal compartment, and undergo matozoon created.47 Each SC can support only a finite
the remaining phases of prophase I and subsequent com- number of germ cells in the order of 50–70 cells. FSH is the
pletion of meiosis I forming secondary spermatocytes. primary hormonal regulator of SC function.57 Numerous
Secondary spermatocytes then undergo meiosis II to form studies have illustrated that suppression of FSH will cause
round spermatids.38 reduced SC function and spermatogenesis production but
Spermiogenesis is the process where round spermatids is rescued upon readministration.57 FSH has also been
develop into a final product, spermatozoa that has an acro- shown to increase mitotic and meiotic DNA synthesis
some on the anterior surface of the head (enzyme storage in SPG and preleptotene spermatocytes.32 Evidence also
for penetration), as well as a midpiece, and tail.39 These exists that thyroid stimulating hormone may influence SC
cells lack cytoplasm and associated organelles.39 During function.58 SCs along with germ cells play a significant role
spermiogenesis, spermatids undergo acrosomal develop- in determination of testicular size, and SC replication may
ment from the golgi apparatus, nuclear condensation, tail continue through the pubertal period.59 The number of
elongation from the centriole, and an ultimate reduction SCs in adult men ranges from 25–900 × 10^6 per testis.60
of cytoplasm eventually transforming from round sper- Therefore, the efficiency of sperm production is closely
matids to elongating spermatids and finally into sper- tied to the number and function of SCs.57 SCs form tight
matozoa.40 Spermatozoa are then ready for release from junctions among themselves, forming two compartments
the apex of the SC into the lumen of the tubule once they and the blood-testis barrier.38 This barrier is essential
are mature. During this process the SCs engulf excess for progression of germ cells through meiosis and sper-
cytoplasm and in many instances phagocytize develop- miogenesis.38 The inner lumen, where there is secretion
ing germ cells.40 Peritubular myoid cells located on the of seminiferous tubule fluid, is essential for nutrient and
interstitial surface of the basement membrane encircling juxtacrine signaling to germ cells.61 SC response to FSH
the seminiferous tubules function to contract and propel then subsequently begins to decline during early puberty
sperm toward the rete testis and epididymis.38 PTMCs and responsiveness to testosterone begins to increase.62
also have additional roles including communication with FSH signaling is regulated by inhibin, which is produced
SCs and expression of GDNF supporting spermatogenesis by SCs acting centrally at the hypothalamus to negatively
and SSCs.41,42 Furthermore, PTMCs may have a role in regulate FSH production; activin, which is produced by the
immunity, interacting with immune cells such as macro- SCs acts centrally to positively regulate FSH production.63
phages (majority), dendritic cells, lymphocytes, and mast
cells located in the interstitium.43 Role of Leydig cells
Once in the epididymis, sperm undergo biochemical, Leydig cells are polygonal cells found adjacent to semi-
physiological, and morphological changes during epidid- niferous tubules in the interstitial space of the testis
ymal transit where they acquire motility and the ability and primarily produce testosterone.64 Leydig cells are
to undergo fertilization.44 As sperm travel from proximal primarily stimulated by LH; however, FSH can also
(caput) to distal (cauda) in the epididymis, their capac- indirectly regulate Leydig cells via effects on the SCs.64
ity for motility is enhanced by low pH and modulated by Other stimulators of Leydig cells include gonadotropin-
concentrations of sodium (low) and potassium (high).45 releasing hormone, melatonin, epidermal growth factor,
During this transit there are numerous changes to the insulin-like growth factor 1, atrial natriuretic peptide,
sperm head, such as a decrease in length:width ratio and ghrelin, and gamma-aminobutyric acid (GABA).65–72
reduction in cytoplasmic volume.45 Emission then occurs Leydig cell function is central to spermatogenesis because
with transport of sperm from distal epididymal and vas testosterone is a primary driver of spermatogenesis. Tes­
deferens to the ejaculatory ducts and posterior urethra tosterone produced by Leydig cells has the ability to
Azoospermia 125

initiate spermatogenesis in humans and functions to in men with secondary hypogonadism, also known as cen-
maintain qualitative levels but not quantitative levels tral hypogonadism.89–91
of sperm production.73 Leydig cells do not store testos-
terone but rapidly secrete it into the interstitial space, Requirements for fertilization
capillaries, and seminiferous tubules where it stimulates Fertilization is the process whereby there is an interac-
both SCs and germ cells. 38 Testosterone concentrations tion between sperm and an egg to form a zygote, which
are 100–1000 times greater intratesticular compared continues to grow forming an embryo and then a fetus.92
to systemic levels.74 Testosterone stimulates differen- Oocytes are stored in female primordial follicles, where
tiation of type A SPG to type B SPG,75 proliferation of they then mature, forming an extracellular matrix known
SPG,76,77 survival of spermatocytes and spermatids via as the zona pellucida, which secretes glycoproteins, fol-
antiapoptotic mechanisms,78 and helps to facilitate sper- lowed by release of eggs during ovulation that travel to
miogenesis.75,79–81 Testosterone negatively feeds back to the ampulla of the oviduct to await fertilization.92 Human
downregulate central LH production.82 In addition to sperm produced in seminiferous tubules are released dur-
testosterone, Leydig cells have been shown to release ing ejaculation during intercourse, and after deposition
substances that activate EGF receptors, a known catalyst in the vaginal canal travel to and through cervical mucus
for spermatogenesis.83 via the cervical os toward the uterus and fallopian tubes.93
Throughout this process, sperm capacitation occurs in
Hormonal regulation preparation for the acrosome reaction.93 Upon completion
Hormonal regulation of spermatogenesis occurs primar- of the acrosome reaction, the spermatozoon can penetrate
ily through intratesticular testosterone production. This the zona pellucida of the oocyte and eventual fusion to
occurs via the hypothalamic-pituitary-testis axis where form a zygote.
the hypothalamus releases gonadotropin releasing hor- Natural fertilization from a male perspective requires
mone (GnRH), stimulating the anterior pituitary via the adequate sperm parameters in order to complete the pro-
hypophyseal portal system into the adenohypophysis to cess described above. Precise thresholds for the lowest
release FSH and LH. FSH subsequently stimulates SCs sperm parameters possible to achieve fertilization are not
to drive spermatogenesis and LH stimulates Leydig cells known. However, the World Health Organization (WHO)
to stimulate testosterone production.84,85 has created normal values based on a population of men
Additional hormones such as inhibin are produced by who have previously demonstrated fertility. These values
SCs, and in conjunction with testosterone from Leydig are based on a properly collected sample, obtained after
cells function to negatively regulate GnRH production in 2–7 days of sexual abstinence.94 Table 10.1 reports the
the hypothalamus. Inhibin is a glycoprotein heterodimer WHO guidelines values that represent the lowest 5th per-
secreted from the base of SCs into interstitial fluid and into centile per each respective parameter among a cohort of
seminiferous tubule fluid via FSH stimulation of SCs.86 men that have previously demonstrated fertility.95
This process overall is stimulated by androgens.63 Inhibin
also acts to inhibit activin, another glycoprotein heterodi- AZOOSPERMIA
mer also released by SCs, which normally enhances FSH Azoospermia is the absence of sperm in the ejaculate after
synthesis and secretion.87 It is the intricate balance of these 2 centrifugations of semen, and can be regarded as nonob-
hormones that regulate spermatogenesis and maintain structive azoospermia (NOA) or obstructive azoospermia
homeostasis. (OA). OA represents 40% of cases.96,97 NOA can be divided
These hormonal pathways also permit opportunities into pretesticular and testicular. Pretesticular azoosper-
for manipulation in the presence of deficiencies and/or mia is often caused by an inadequate production of FSH
abnormalities. In the presence of testosterone deficiency, and LH secondary to a central deficiency in the pituitary
clinicians must remember to avoid treatment of men or hypothalamus. Etiologies of pretesticular NOA include
desiring fertility with exogenous testosterone, since exog-
enous testosterone will negatively feedback and inhibit LH Table 10.1  World Health Organization 2010 semen
production and subsequent intratesticular testosterone analysis lower limits of normal per respective parameter.
production, impairing spermatogenesis.88 Medications
such as selective estrogen receptor modulators (SERMs) WHO 2010 cutoff
(i.e., clomiphene citrate) may be used to block negative Semen analysis parameter (5th percentile)95
feedback of estrogen on the hypothalamic-pituitary-axis Volume (mL) 1.5
and ultimately increase testosterone production and pro- Concentration (million sperm/mL) 15
mote spermatogenesis.89 Aromatase inhibitors (AIs), (i.e., Total sperm count (million) 39
anastrozole) decrease conversion of peripheral testoster- Total motility (%) 40
one to estrogen, thus increasing circulating testosterone Progressive motility (%) 32
and decreasing estrogen levels. Other medications, such
Normal morphology (%) 4
as recombinant FSH, human menopausal gonadotropins
Vitality (%) 58
(hMGs), human chorionic gonadotropin (hCG) or exog-
Leukocyte count (million/mL) <1.0
enous GnRH have been used in the treatment of infertility
126  Regulation of fertility and infertility in humans

congenital disease such as Kallman syndrome, or acquired Generally, natural fertility is poor with oligozoospermia
through exogenous androgens and surgical extirpation and therefore treatment options include correcting the
or radiation of the pituitary.98 Testicular NOA occurs underlying cause, vitamins and antioxidants, varicocele
due to impaired spermatogenesis despite adequately pro- treatment if present, and lifestyle modification such as
duced FSH. Etiologies include genetic conditions such smoking cessation and avoidance of drugs or excessive
as Klinefelter syndrome, Y-chromosome microdeletion alcohol.107 Assisted reproductive techniques may be neces-
(AZFa, AZFb, AZFc), or acquired through infection, che- sary in some instances where natural fertility is not suc-
motherapy, or radiation.99 However, nearly 70% of NOA cessful despite treatment of modifiable factors.109
cases remain idiopathic without a defined explanation.
Impaired spermatogenesis may present with complete Asthenozoospermia
absence of germ cells termed Sertoli cell only (SCO), matu- Sperm motility is accomplished via microtubular struc-
ration arrest where germ cells stop differentiation beyond tures powered by ATP via dynein arms.110 Their move-
primary spermatocytes (early) or secondary spermato- ment is explained by the 9+2 configuration with motility
cytes (late), or hypospermatogenesis where very limited maturation in the epididymis.111 Asthenozoospermia is
spermatogenesis occurs. OA is due to mechanical obstruc- defined as less than 40% motility or less than 32% of pro-
tion such as congenital bilateral absence of the vas deferens gressively motile sperm based on the 2010 WHO semen
(CBAVD) due to a defect in the cystic fibrosis transmem- analysis parameters.112 A recent meta-analysis determined
brane conductance regulator (CFTR) gene, seminal vesical that the combination of total sperm number and motil-
atresia, ejaculatory duct cysts or obstruction, vasectomy, ity, termed total motile count (TMC), is more predictive
or idiopathic- or infectious-related primary epididymal of spontaneous pregnancy rate than either parameter
obstruction. Other posttesticular etiologies include ejacu- independently.113
latory dysfunction or failure of emission due to retroperi- Etiologies of asthenozoospermia include varicocele,
toneal lymph node dissection (RPLND), pelvic surgery, prolonged periods of anejaculation, leukocytospermia,
spinal cord injury, or diabetes.99,100 genital infections, oxidative stress, antisperm antibod-
Treatment: Sperm retrieval for OA includes microsur- ies, metabolic disorders affecting ATP production, partial
gical epididymal sperm aspiration (MESA) and percutane- ejaculatory duct obstruction, partial epididymal obstruc-
ous epididymal sperm aspiration (PESA). MESA utilizes tion or sperm (USDs).114,115 Antisperm antibodies (ASAs)
a scrotal incision whereby the testis is delivered and the have been found in 5%–15% of infertile men versus low
epididymis is observed using an operating microscope. rates of 1%–2% in fertile men and are often caused by
Dilated epididymal tubules are aspirated at the caput of the trauma, infection, malignancy, vasectomy, or sources of
testis.101 This is the gold standard surgical sperm retrieval inflammation.116 ASAs are variable in that they can act
technique for OA, with success rates of 96%–100%, yield- against different immunoglobulins with the majority
ing 15–95 million sperm.102,103 PESA is performed trans- against immunoglobulin G (IgG) and immunoglobulin A
cutaneously with a needle inserted directly into caput (IgA) but also immunoglobulin M (IgM).117 ASAs interfere
epididymis and sperm is aspirated, with success rates of with various elements of sperm including the head (inter-
61%–96%.104,105 Surgical sperm retrieval for NOA occurs fering with acrosome reaction), tail (alter sperm motil-
via microdissection testicular sperm extraction (micro- ity), and midpiece.116 These patients can be treated with
TESE). This is performed through delivering the testis, Assisted reproductive techniques (ARTs) with reasonable
making a tunical incision, bivalving the testis, and exam- success as meta-analyses have demonstrated that ASAs are
ining for the seminiferous tubules under an operating not related to pregnancy rates with IVF and intracytoplas-
microscope for dilated tubules. MicroTESE has successful mic sperm injection (ICSI).118 USDs may occur in axone-
sperm retrieval rates on average of 45%–63%.106 Patients mal or periaxonemal structures of the flagellum.115 These
with testicular NOA may also be treated with hormonal typically occur in men with viable but severely astheno-
therapy such as clomiphene citrate or aromatase inhibitors zoospermic sperm with motility less than 10%. Primary
in the setting of low testosterone or high estradiol levels, immotile-cilia syndrome is an uncommon autosomal
respectively.107 Patients with central NOA require either recessive disorder with altered ciliary structure, known as
GnRH, hCG, recombinant FSH, or hMG to stimulate Kartagener’s syndrome, diagnosed by transmission elec-
spermatogenesis. tron microscopy illustrating dynein arm absence.119 Other
defects include the axonemal 9+0 defect lacking the cen-
Oligozoospermia tral microtubular structure that can be associated with
Oligozoospermia is defined as low sperm concentration autosomal dominant polycystic kidney disease.115 USDs
(<15 million/mL semen) in the ejaculate.108 Etiologies can may be treated with IVF/ICSI, although the efficiency of
be related to primary testicular failure (idiopathic sper- ICSI appears to be lower in men with USDs. Treatment
matogenic failure, testicular damage such as varicocele, options for asthenozoospermia should focus on any
drugs and toxins, chromosomal abnormalities, and genet- identifiable etiology such as varicocele repair, transure-
ics), mixed primary and secondary failure (nonrepro- thral resection of ejaculatory ducts if partial obstruction,
ductive illness), and secondary testicular failure (partial removal of external environmental stressor, and regular
GnRH deficiency or partial gonadotropin deficiency).107 intercourse. Medical treatments are limited; however,
Genetic factors  127

studies examining L-carnitine have been explored in an and fluorescence in situ hybridization (FISH).128 Studies
attempt to improve sperm motility rates with positive have shown comparable correlations in DNA measure
results.120 If infertility persists, assisted reproductive tech- damage between these assays.127 The results of these assays
niques should be considered. have been promising as a recent meta-analysis revealed
that couples are more likely to achieve pregnancy if DNA
Teratozoospermia fragmentation index was low, illustrating its significant
Normal sperm morphology is a controversial require- predictive value for pregnancy success.129 Since only neat
ment for fertility due to its subjectivity in its assessment semen DNA fragmentation best predicts IVF/ICSI suc-
and reproducibility.121 Kruger et al. have emphasized the cess, it is not clear if “selection” of undamaged sperm from
importance of standardized criteria, which includes a nor- an abnormal sample improves the chance of pregnancy for
mal sperm possessing a head with a smooth, oval configu- men with abnormal sperm DNA fragmentation. Patients
ration with well-defined acrosome (40%–70%), length of with varicoceles have increased levels of DNA fragmen-
head to be 5–6 μm, and no visible defect in the neck, mid- tation.130 Treatment options include varicocele repair,
piece, or tail.122 Teratozoospermia is the term used to iden- antioxidants, and reduction of sexual abstinence.131–133 Of
tify abnormal sperm morphology that inhibits the ability note, sperm directly retrieved from the testis reliably have
for normal fertilization. Etiologies of teratozoospermia lower sperm DNA fragmentation than that in the ejacu-
may include genetic disorders leading to globoozospermia, late of men with abnormal sperm DNA fragmentation.
macrocephaly, decapitated sperm syndrome, and fibrous Antioxidants are naturally found in seminal fluid and play
sheath dysplasia, and potentially environmental factors a key role due to the lack of cytoplasmic fluid in sperm,
such as tobacco, cannabis, temperature, obesity, testicular which illustrates that antioxidant supplementation could
cancer, varicocele, or infection. However, the origin of the play a key role in protecting against DNA damage and frag-
majority of defects remains unclear.123 Treatment options mentation.134 A recent review of literature illustrated that
include correcting underlying etiologies if possible or use antioxidant therapy also improved outcomes for patients
of ARTs such as IVF/ICSI to select for morphologically undergoing ICSI with elevated DNA fragmentation levels,
normal sperm.123 and which had improved clinical pregnancy and implan-
tation rates.135 Antioxidants that have been studied include
Acrosome reaction and capacitation vitamin C, vitamin E, glutathione, L-carnitine, coenzyme
q10, vitamin B9, selenium, and vitamin B12, which have
Successful sperm fertilization of an egg relies on numer- shown varying levels of success, suggesting that these sub-
ous sperm parameters, described above, with the goal of stances may serve to curb DNA fragmentation, but further
penetrating the zona pellucida and fusion with an oocyte adequately powered prospective studies are required.135
membrane, which is known as the acrosome reaction.124
Capacitation is the process that permits the acrosome GENETIC FACTORS
reaction to occur and creates biochemical modifications to Genetic factors are identifiable in 15%–30% of male infertil-
the acrosome of the sperm head, allowing penetration of ity, while the majority (70%) remain idiopathic and under
the outer layer, and changes in the tail permitting greater investigation.136 Klinefelter syndrome (KS) is the most com-
mobility. These are ultimately facilitated by removal of mon genetic cause of male infertility with a prevalence 10%
sterols, glycoproteins, and increased permeability and in men with NOA and 5% in men with severe oligozoosper-
influx of calcium.125 The acrosome is structurally located mia and an incidence of 0.1%–0.2% in the general popula-
on the anterior half of the sperm head. Acrosome reaction tion.137–139 It is characterized by a karyotype of 47, XXY in
involves the penetration of the corona radiata followed 80% of cases and mosaic patterns in the remaining 20%.140,141
by the release of hyaluronidase and exposure of acrosin, Clinically, men with KS have a variable phenotype but are
which then digests the zona pellucida of the oocyte.126 generally characterized as tall, with gynecomastia, gynoid
These reactions are required for fertilization and defects hips, cognitive abnormalities, sparse hair distribution, and
in these steps have negative implications on fertility. having progressive hyalinization-associated testicular fail-
ure resulting in infertility, primary hypogonadism, and
DNA fragmentation small firm testes.142 Another sex chromosome anomaly
Sperm DNA fragmentation has emerged as another con- includes 47,XYY, which carries an incidence of 0.1%. These
dition associated with infertility and numerous assays men will have variable sperm concentrations but typically
have been developed to assess this, with the theory that present with severe oligozoospermia or azoospermia.143
the extent of DNA fragmentation is inversely related to Y-chromosome microdeletions are characterized by 1 of 3
sperm quality.127 Numerous techniques have been devel- clinically relevant and described deletions, AZFa, AZFb,
oped to assess DNA fragmentation, each with their own and AZFc. These deletions occur within the azoosper-
advantages and disadvantages, and include sperm chro- mia factor (AZF) region of the euchromatin long arm of
matin dispersion (SCD), sperm chromatin structure assay the Y chromosome.144,145 This locus holds 14 protein cod-
(SCSA), terminal deoxynucleotidyl transferase-mediated ing genes that are required for spermatogenesis. AZFa and
dUTP nick end labeling (TUNEL), single-cell gel electro- AZFb deletions result in azoospermia and complete dele-
phoresis (COMET), DNA ladder, DNA-break detection, tions do not have any reports of successful surgical sperm
128  Regulation of fertility and infertility in humans

retrieval.146 AZFc deletions result in severe oligozoospermia VARICOCELES


in 70% of men with total sperm concentrations typically less Varicoceles are abnormally dilated internal spermatic
than 1 million sperm per milliliter and azoospermia in the veins in the scrotum. Clinically relevant varicoceles are
remainder of men.147 Kallman syndrome is characterized those that can be palpated with Valsalva (grade 1), pal-
by hypogonadotropic hypogonadism and anosmia. Other pated without Valsalva (grade 2), or visualized through
clinical features include cleft palate abnormalities, cryptor- the scrotal wall at rest (grade 3). They occur in 15% of the
chidism, unilateral renal agenesis, neurogenic deafness and general population and 35% of men presenting with infer-
infertility due to lack of GnRH production and subsequent tility. They are also the most common cause of second-
gonadotropins, testosterone, and spermatogenesis.148 This ary infertility, accounting for 70%–80% of these men.168
may be the result from abnormalities in the KAL1 or FGFR1 Varicoceles may lead to increased sperm DNA frag-
gene.149 Mutations in the testis-expressed 11 (TEX11) gene mentation,169 decreased sperm concentration, motility,
may lead to maturation arrest and azoospermia.150 TEX11 is and abnormal morphology independently or altogether,
located on the X chromosome Xq13.2 and encodes a protein which is termed oligoastheoteratozoospermia (OAT).170
that functions to repair double-strand DNA breaks, homol- Varicocele repairs have been associated with improved
ogous chromosome synapses, and recombination.151 CFTR semen analysis parameters,171 DNA fragmentation,172 nat-
gene mutations may lead to CBAVD. The most commonly ural pregnancy rates, increased fertilization during IVF173
mutated alleles implicated in CBAVD include F508del and intrauterine insemination (IUI),174 and decreased
(17%), R117H (3%), and 5T (25%). This carries a prevalence miscarriage rates.175 Several theories exist regarding the
of 1% among infertile men and 25% of men with primary mechanism of varicoceles on male infertility; however,
obstructive azoospermia. These men require surgical sperm increased scrotal temperature and oxidative stress are
retrieval from the epididymis for use with assisted repro- thought to be central to the mechanisms. This may con-
ductive techniques. They also require genetic counseling, tribute to germ cell apoptosis, impaired Leydig cell func-
abdominal ultrasound to assess for renal agenesis, and tion, and testosterone production,176,177 as well as damage
CFTR testing of their partner to assess risk of cystic fibrosis to the BTB with associated autoimmunity and impaired
in their offspring.152 cleavage of sperm cytoplasm.178–180 Seminal proteomes also
More recent research has been uncovering regulatory differ in men with varicoceles; expression of semenogelins
mechanisms of spermatogenesis exerted by small and long I and II are increased and are thought to inhibit sperm
noncoding RNAs. Small RNAs comprise three different motility and prevent premature sperm hyperactivation
classes of RNA: microRNA (miRNA), small interfering and capacitation.181,182 As such, varicocele repair for pal-
RNA (siRNA), and PIWI-interacting RNA (piRNA).153 pable varicoceles may be of benefit to the infertile male.
piRNAs play a vital role in spermatogenesis during both However, it is important to consider both male and female
PGC development and during meiotic prophase 1, termed factors, such as age, when choosing the appropriate course
prepachytene piRNAs, and pachytene piRNAs, respec- of therapy since the effect of varicocele repair takes 3–6
tively. PiRNAs function through repression of transposons months to see potential improvements.
and elimination of mRNAs during spermatid forma-
tion.154–160 Further research is required to identify mutated Fertility as a function of age
and aberrant functioning piRNAs among infertile males. In our present-day society, patients are seeking fertility
Numerous microRNA, however, have repeatedly demon- later in life, and this impacts outcomes both for females
strated important roles during spermatogenesis, including and males with respect to fertility potential.183 Fertility
proliferation of PGCs, meiotic prophase I, and spermatid as a function of age is commonly assessed, especially in
elongation.161–164 MicroRNA function via associating with the female population. Women are born with a set num-
argonaute proteins (AGO) and silencing translation of ber of primordial follicles.184 As a consequence, increased
mRNA targets. Several differentially expressed microRNAs age among women results in increased rates of infertility
have been identified in men with NOA and are summa- particularly after age 35.185 Men, on the other hand, con-
rized by Neto et al. 2016. These include downregulation tinue active sperm production throughout life but do lose
of miR-34 family, miR-122, miR-181a, miR-146b, miR- function with age.184 However, albeit controversial, some
513a-5p, miR-509-5p, miR-274b, and miR-202-5p, and studies have identified an association of increased paternal
upregulation of miR19b, Let-7a, miR-429, miR-141, and age with increased sperm DNA fragmentation, epigenetic
miR-7-1-3p.29 Long noncoding RNA (lncRNA) interact changes, reduced fertility beyond late 30s, and decreased
with chromatin binding proteins, RNA binding proteins, ART successes.186 Additionally, paternal age greater than
and mRNA protection or degradation, and act as a miRNA 45 is associated with risk of late fetal death.187 Increased
decoy and function to activate or repress translation.165 paternal age has also been associated with cleft lip and pal-
Expression of lncRNAs begins during early pachytene ate and some childhood malignancies such as retinoblas-
spermatocytes, increases in late pachytene spermatocytes, toma188 and childhood acute lymphoblastic leukaemia.189
and tapers off in spermatids. Several lncRNAs have been Schizophrenia, bipolar affective disorder, and autism
identified to play an important role in spermatogenesis have also been associated with increased paternal age.186
and male sex development,166,167 but further research is Effects of advanced paternal age are thoroughly reviewed
needed to identify candidate lncRNAs in male fertility. in Sharma et al. 2015 for further reading.186
Environmental chemicals  129

OBESITY AND DIABETES MELLITUS macrolides result in impaired sperm motility and via-
Obesity is in epidemic proportions, particularly in North bility,228,229 aminoglycosides lead to impaired sperm
America, where the prevalence is 35% based on the defini- motility,230,231 subtilosin results in impaired sperm motil-
tion of a body mass index (BMI) >30.190 Obesity has a neg- ity,232 and ketoconazole results in impaired testosterone
ative association with sperm concentration, total sperm production.209,233
count,191–193 morphology, motility, and DNA fragmenta- Chemotherapeutic agents generally have significantly
tion.194–198 A central hypothesis for the effect of obesity negative effects on germ cells, Leydig cells, and SCs,
on abnormal sperm production includes the state of high resulting in temporary or permanently impaired sper-
estradiol noted in obese men due to peripheral conversion matogenesis;234 this topic is reviewed in depth by Wallace
of testosterone to estrogen through aromatase enzymes et al. 2005.235 High-risk agents include cyclophosphamide,
in adipose tissue.199,200 Estradiol negatively regulates the ifosfamide, chlorambucil, melphalan, busulfan, dacarba-
hypothalamus-pituitary-testis axis, serving to decrease zine, procarbazine, and nitrogen-mustard, vincristine,
LH and FSH production, resulting in less testosterone procarbazine, prednisone (MOPP). Intermediate risk
production and less SC stimulation. Evidence to support agents include carboplatin, cisplatin, doxorubicin, and
this includes low levels of inhibin B in obese men. 201,202 adriamycin, bleomycin, vinblastine, dacarbazine (ABVD),
Furthermore, ERα receptors are found in Leydig cells and and bleomycin, etoposide, cisplatin (BEP). Low-risk agents
SCs, and ERβ receptors are present in nearly all intersti- include methotrexate, vincristine, vinblastine, dactinomy-
tial and tubular cells of the testis.203–206 Other factors likely cine, mercaptopurine, and bleomycin. Imatinib has been
contributing to impaired semen production include sed- demonstrated to induce Leydig cell dysfunction with
entary lifestyle, diet, and epigenetic changes to the germ subsequently impaired testosterone production.235,236 It
cells.191,207 is essential for clinicians to discuss fertility preservation
The incidence of infertility among diabetic men is much with men and couples prior to undergoing chemotherapy
higher than the average population and ranges from 35% to afford the ability for sperm cryopreservation as many
to 51%. Men with diabetes mellitus (DM) are found to have men may suffer from irreversible infertility following
decreased testosterone levels, increased sperm DNA frag- therapy.
mentation, and variable reports on semen analysis param-
Fertility and pollution
eters, epididymal damage and impaired sperm transport,
neuropathy and abnormalities of emission, increased oxi- There are many reported factors affecting male infertil-
dative stress and associated damage to sperm nuclear and ity, which include cigarette smoking, alcohol consump-
mitochondrial DNA, and potential intratesticular autoim- tion, and environmental hazards.237,238 Air pollution is
mune damage.208 an increasing problem in large populations living in large
urban settings and its effects on fertility are apparent but
MEDICATIONS remain unclear.239 Studies have demonstrated that in both
Several medications that are used among men in repro- animal and human studies, air pollution may alter game-
ductive years may negatively impact spermatogenesis and togenesis and therefore alter reproductive ability.240 Other
fertility. This topic is reviewed in depth by Samplaski and studies have shown increases in miscarriage rate in IVF.239
Nangia 2015.209 Hormonal medications such as exogenous Numerous components of air pollution are being exam-
testosterone, anabolic steroids, and androgen deprivation ined, including nitrogen, sulfur, ozone, and particulate
suppress endogenous testosterone production, resulting matter concentrations.241
in suppression of spermatogenesis.210 Medications used to
treat lower urinary tract symptoms such as alpha blockers ENVIRONMENTAL CHEMICALS
may cause impaired emission or retrograde ejaculation, Numerous environmental chemicals have demonstrated
reduced sperm counts and motility,211,212 while 5 alpha- potentially harmful effects on spermatogenesis and infer-
reductase inhibitors result in reduced sperm counts among tility (reviewed in depth by Neto et al.29). Lead (paint)
5% of men, and rarely, sexual dysfunction.213,214 Psychiatric- has been demonstrated to disrupt chromatin during
related medications such as selective 5-hydroxytryptamine spermatogenesis and damage mannose receptors.242
reuptake inhibitors (i.e., SSRIs, SNRIs, TCAs, MAOIs) Cadmium (batteries and pigments) disrupts tight junc-
result in sperm DNA fragmentation and erectile and ejac- tions of the BTB and causes failure of spermiation.243,244
ulatory dysfunction, likely as a consequence of impaired Ethylene dibromide (gasoline) has been shown to bind
sperm transport;215–218 lithium has been shown to reduce histones, disrupting the ability of DNA packaging,
sperm viability.219 Antihypertensives such as beta block- resulting in reducing sperm counts, impaired motil-
ers may lead to sexual dysfunction and impaired sperm ity, and abnormal morphology.245,246 1,2-Dibromo-3-
motility;220,221 diuretics may also cause sexual dysfunction, chloropropane (synthesis of fire retardants) results in
impaired sperm motility and concentration;222,223 calcium increased reactive oxygen species among germ cells
channel blockers result in decreased sperm concentration, with spermatogonial apoptosis and decreased sperm
motility, and acrosome reaction.224–226 Antibiotics have counts.247,248 Tetrachlorodibenzo-p-dioxin (herbicide
varying effects: nitrofurantoin has been shown to lead to and pesticide synthesis) impairs BTB tight junctions and
spermatogenic arrest and reduced sperm concentration,227 results in impaired semen parameters.249–251 Phthalates
130  Regulation of fertility and infertility in humans

(plastics) result in decreased testosterone production due REFERENCES


to activation of peroxisome proliferator-activated recep- 1. Parker V. Antony Van Leeuwenhoek. Bull Med Libr
tors (PPARs) and also impairs the function of both germ Assoc. 1965;53:442–447.
cells and SCs.252,253 Bisphenol A (BPA) (plastics and res- 2. Ge R, Chen G, Hardy MP. The role of the Leydig
ins) binds to the androgen receptor (AR), ERβ and ERα, cell in spermatogenic function. Adv Exp Med Biol.
resulting in impaired androgen and estrogen signaling 2008;636:255–269.
and associated poor semen parameters.254,255 It is unclear 3. Morales A. The long and tortuous history of
what degree of exposure is required in humans to see the discovery of testosterone and its clinical
negative effects for many of these substances; however, application. The Journal of Sexual Medicine.
among men wishing to conceive, the best recommenda- 2013;10(4):1178–1183.
tion is to avoid potential exposures when possible. 4. Smith PE. Maintenance and restoration of spermato-
genesis in hypophysectomized rhesus monkeys by
Assisted reproductive techniques androgen administration. The Yale Journal of Biology
ARTs work to treat infertility in the presence of male or and Medicine. 1944;17(1):281–287.
female factor infertility. Methods for ART include IUI, 5. Wang J, Sauer MV. In vitro fertilization (IVF): A
IVF, and ICSI. 256 Table 10.2 highlights the sperm counts review of 3 decades of clinical innovation and tech-
required for successful fertilization using assisted repro- nological advancement. Therapeutics and Clinical
ductive techniques. IUI can be performed in as low as Risk Management. 2006;2(4):355–364.
a 500,000 motile sperm count, but optimal results are 6. Curtin SC, Abma JC, Ventura SJ, Henshaw SK.
generally reported with at least 5 million sperm. 257,258 Pregnancy rates for U.S. women continue to drop.
IVF typically requires 50,000 to 100,000 sperm per NCHS Data Brief. 2013(136):1–8.
oocyte.259,260 ICSI on the other hand can be performed 7. Thoma ME, McLain AC, Louis JF et al. Prevalence
with very few sperm present in the ejaculate or even of infertility in the United States as estimated by the
a single sperm from the ejaculate or when harvested current duration approach and a traditional con-
using methods such as MESA, PESA, or microTESE. 261 structed approach. Fertil Steril. 2013;99(5):1324–1331
Other methods include cryopreservation of sperm or e1321.
oocytes, which are techniques that have been used 8. Smith JF, Eisenberg ML, Millstein SG et al. Fertility
successfully. 262,263 treatments and outcomes among couples seek-
ing fertility care: Data from a prospective fertility
cohort in the United States. Fertility and Sterility.
CONCLUSION
2011;95(1):79–84.
Male fertility relies first on adequate spermatogenesis 9. Mascarenhas MN, Flaxman SR, Boerma T, Vanderpoel
and spermiogenesis producing viable spermatozoa capable S, Stevens GA. National, regional, and global trends
of fertilization. This process involves critical regulatory in infertility prevalence since 1990: A system-
steps that are both spatially and temporally important. atic analysis of 277 health surveys. PLoS Medicine.
Abnormalities during these processes may lead to poor 2012;9(12):e1001356.
sperm production and subsequent infertility. Extrinsic 10. Kumar N, Singh AK. Trends of male factor infer-
factors such as medications, environmental pollution, and tility, an important cause of infertility: A review of
lifestyle factors may also impact the efficiency of spermato- literature. Journal of Human Reproductive Sciences.
genesis or the fertilization potential of the spermatozoa. 2015;8(4):191–196.
Thus, it is important for the clinician to understand the 11. Jorgensen N, Andersen AG, Eustache F et al. Regional
multitude of factors that are both necessary for optimal differences in semen quality in Europe. Hum Reprod.
fertility as well as those that may contribute to infertility 2001;16(5):1012–1019.
to ensure each patient may be optimized for a successful 12. Jorgensen N, Carlsen E, Nermoen I et al. East-West
pregnancy. gradient in semen quality in the Nordic-Baltic
area: A study of men from the general population
in Denmark, Norway, Estonia and Finland. Hum
Table 10.2  Minimal motile sperm counts required for
Reprod. 2002;17(8):2199–2208.
respective type of assisted reproductive technique.
13. Fernandez MF, Duran I, Olea N et al. Semen qual-
Assisted reproductive ity and reproductive hormone levels in men from
techniques (ART) Motile sperm count Southern Spain. Int J Androl. 2012;35(1):1–10.
IUI (intrauterine 5 million264 (range: 500,000 to 14. Nordkap L, Joensen UN, Blomberg Jensen M,
Insemination) 10 million257,258) Jorgensen N. Regional differences and temporal
IVF (in vitro fertilization) 50,000 to 100,000 motile trends in male reproductive health disorders: Semen
sperm/oocyte quality may be a sensitive marker of environmen-
tal exposures. Mol Cell Endocrinol. 2012;355(2):​
ICSI (intracytoplasmic Minimum single sperm261
221–230.
sperm injection)
References 131

15. Agarwal A, Mulgund A, Hamada A, Chyatte MR. 32. de Kretser DM, Loveland KL, Meinhardt A,
A unique view on male infertility around the globe. Simorangkir D, Wreford N. Spermatogenesis. Hum
Reprod Biol Endocrinol. 2015;13:37. Reprod. 1998;13 Suppl 1:1–8.
16. Merzenich H, Zeeb H, Blettner M. Decreasing sperm 33. Phillips BT, Gassei K, Orwig KE. Spermatogonial
quality: A global problem? BMC Public Health. stem cell regulation and spermatogenesis. Philos Trans
2010;10:24. R Soc Lond B Biol Sci. 2010;365(1546):1663–1678.
17. Pastuszak AW, Lamb DJ. Counting your sperm 34. Walker WH. Non-classical actions of testosterone
before they fertilize: Are sperm counts really declin- and spermatogenesis. Philos Trans R Soc Lond B Biol
ing? Asian J Androl. 2013;15(2):179–183. Sci. 2010;365(1546):1557–1569.
18. Heller CH, Clermont Y. Kinetics of the germi- 35. Hai Y, Hou J, Liu Y et al. The roles and regulation of
nal epithelium in man. Recent Prog Horm Res. Sertoli cells in fate determinations of spermatogonial
1964;20:545–575. stem cells and spermatogenesis. Semin Cell Dev Biol.
19. Clermont Y. The cycle of the seminiferous epithe- 2014;29:66–75.
lium in man. Am J Anat. 1963;112:35–51. 36. Hayashi T, Yamada T, Kageyama Y, Kihara K.
20. Robinson LL, Gaskell TL, Saunders PT, Anderson RA. Expression failure of the notch signaling system is
Germ cell specific expression of c-kit in the human associated with the pathogenesis of testicular germ
fetal gonad. Mol Hum Reprod. 2001;7(9):845–852. cell tumor. Tumour Biol. 2004;25(3):99–105.
21. del Mazo J, Garcia-Lopez J, Weber M. Epigenetic 37. van den Driesche S, Sharpe RM, Saunders PT,
traits of testicular cancer: From primordial germ Mitchell RT. Regulation of the germ stem cell niche
cells to germ cell tumors. Epigenomics. 2014;6(3):​ as the foundation for adult spermatogenesis: A role
253–255. for miRNAs? Semin Cell Dev Biol. 2014;29:76–83.
22. Figueira MI, Cardoso HJ, Correia S, Maia CJ, 38. Smith LB, Walker WH. The regulation of sper-
Socorro S. Hormonal regulation of c-KIT receptor matogenesis by androgens. Semin Cell Dev Biol.
and its ligand: Implications for human infertility? 2014;30:2–13.
Prog Histochem Cytochem. 2014;49(1–3):1–19. 39. O’Donnell L. Mechanisms of spermiogenesis and
23. Gilbert DC, Chandler I, McIntyre A et al. ­spermiation and how they are disturbed. Spermato­
Clinical and biological significance of CXCL12 genesis. 2014;4(2):e979623.
and CXCR4 expression in adult testes and germ 40. Cheng CY, Mruk DD. Regulation of spermiogenesis,
cell tumours of adults and adolescents. J Pathol. spermiation and blood-testis barrier dynamics: Novel
2009;217(1):94–102. insights from studies on Eps8 and Arp3. Biochem J.
24. Pauls K, Jager R, Weber S et al. Transcription fac- 2011;435(3):553–562.
tor AP-2gamma, a novel marker of gonocytes and 41. Chen LY, Brown PR, Willis WB, Eddy EM. Peritubular
seminomatous germ cell tumors. Int J Cancer. myoid cells participate in male mouse spermato-
2005;115(3):470–477. gonial stem cell maintenance. Endocrinology.
25. Levine AJ, Brivanlou AH. GDF3, a BMP inhibitor, 2014;155(12):4964–4974.
regulates cell fate in stem cells and early embryos. 42. Nurmio M, Kallio J, Adam M, Mayerhofer A, Toppari
Development. 2006;133(2):209–216. J, Jahnukainen K. Peritubular myoid cells have a role
26. Pentikainen V, Erkkila K, Suomalainen L, Parvinen in postnatal testicular growth. Spermatogenesis.
M, Dunkel L. Estradiol acts as a germ cell survival 2012;2(2):79–87.
factor in the human testis in vitro. J Clin Endocrinol 43. Zhao S, Zhu W, Xue S, Han D. Testicular defense sys-
Metab. 2000;85(5):2057–2067. tems: Immune privilege and innate immunity. Cell
27. Lambrot R, Coffigny H, Pairault C et al. Use of organ Mol Immunol. 2014;11(5):428–437.
culture to study the human fetal testis development: 44. van Der Horst G, Seier JV, Spinks AC, Hendricks S.
Effect of retinoic acid. J Clin Endocrinol Metab. The maturation of sperm motility in the epididymis
2006;91(7):2696–2703. and vas deferens of the vervet monkey, Cercopithecus
28. Culty M. Gonocytes, the forgotten cells of the germ aethiops. Int J Androl. 1999;22(3):197–207.
cell lineage. Birth Defects Res C Embryo Today. 45. Cooper TG. The epididymis, cytoplasmic droplets
2009;87(1):1–26. and male fertility. Asian J Androl. 2011;13(1):130–138.
29. Neto FT, Bach PV, Najari BB, Li PS, Goldstein M. 46. Roberts M, Jarvi K. Steps in the investigation and
Spermatogenesis in humans and its affecting factors. management of low semen volume in the infertile
Semin Cell Dev Biol. 2016;59:10–26. man. Can Urol Assoc J. 2009;3(6):479–485.
30. Print CG, Loveland KL. Germ cell suicide: New 47. Griswold MD. The central role of Sertoli cells in sper-
insights into apoptosis during spermatogenesis. matogenesis. Semin Cell Dev Biol. 1998;9(4):411–416.
Bioessays. 2000;22(5):423–430. 48. Kerr JB, Millar M, Maddocks S, Sharpe RM. Stage-
31. Paniagua R, Nistal M. Morphological and histo- dependent changes in spermatogenesis and Sertoli
metric study of human spermatogonia from birth cells in relation to the onset of spermatogenic fail-
to the onset of puberty. J Anat. 1984;139(Pt 3):​ ure following withdrawal of testosterone. Anat Rec.
535–552. 1993;235(4):547–559.
132  Regulation of fertility and infertility in humans

49. Robinson R, Fritz IB. Metabolism of glucose by Sertoli 66. Sharpe RM, Fraser HM, Cooper I, Rommerts FF.
cells in culture. Biol Reprod. 1981;24(5):1032–1041. Sertoli-Leydig cell communication via an LHRH-
50. Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli- like factor. Nature. 1981;290(5809):785–787.
germ cell interactions and their significance in germ 67. Rossi SP, Windschuettl S, Matzkin ME et al.
cell movement in the seminiferous epithelium during Melatonin in testes of infertile men: Evidence for
spermatogenesis. Endocr Rev. 2004;25(5):747–806. anti-proliferative and anti-oxidant effects on local
51. Hogarth CA, Griswold MD. The key role of vitamin A macrophage and mast cell populations. Andrology.
in spermatogenesis. J Clin Invest. 2010;120(4):956–962. 2014;2(3):436–449.
52. Boussouar F, Benahmed M. Lactate and energy 68. Rossi SP, Matzkin ME, Terradas C et al. New insights
metabolism in male germ cells. Trends Endocrinol into melatonin/CRH signaling in hamster Leydig
Metab. 2004;15(7):345–350. cells. Gen Comp Endocrinol. 2012;178(1):153–163.
53. Rato L, Alves MG, Socorro S, Duarte AI, Cavaco JE, 69. Syed V, Khan SA, Nieschlag E. Epidermal growth
Oliveira PF. Metabolic regulation is important for factor stimulates testosterone production of
spermatogenesis. Nat Rev Urol. 2012;9(6):330–338. human Leydig cells in vitro. J Endocrinol Invest.
54. Alves MG, Socorro S, Silva J et al. In vitro cultured 1991;14(2):93–97.
human Sertoli cells secrete high amounts of acetate 70. Ishikawa T, Fujioka H, Ishimura T, Takenaka
that is stimulated by 17beta-estradiol and suppressed A, Fujisawa M. Ghrelin expression in human
by insulin deprivation. Biochim Biophys Acta. testis and serum testosterone level. J Androl.
2012;1823(8):1389–1394. 2007;28(2):320–324.
55. Meinhardt A, Hedger MP. Immunological, paracrine 71. Geigerseder C, Doepner R, Thalhammer A et al.
and endocrine aspects of testicular immune privi- Evidence for a GABAergic system in rodent and
lege. Mol Cell Endocrinol. 2011;335(1):60–68. human testis: Local GABA production and GABA
56. Cudicini C, Lejeune H, Gomez E et al. Human Leydig receptors. Neuroendocrinology. 2003;77(5):314–323.
cells and Sertoli cells are producers of i­ nterleukins-1 72. Geigerseder C, Doepner RF, Thalhammer A, Krieger
and -6. J Clin Endocrinol Metab. 1997;82(5):​ A, Mayerhofer A. Stimulation of TM3 Leydig cell
1426–1433. proliferation via GABA(A) receptors: A new role for
57. Simoni M, Weinbauer GF, Gromoll J, Nieschlag testicular GABA. Reprod Biol Endocrinol. 2004;2:13.
E. Role of FSH in male gonadal function. Ann 73. Ambiye VR, Langade D, Dongre S, Aptikar P,
Endocrinol (Paris). 1999;60(2):102–106. Kulkarni M, Dongre A. Clinical evaluation of
58. Wagner MS, Wajner SM, Maia AL. The role of thy- the spermatogenic activity of the root extract of
roid hormone in testicular development and func- Ashwagandha (Withania somnifera) in oligosper-
tion. J Endocrinol. 2008;199(3):351–365. mic males: A pilot study. Evid Based Complement
59. Pintus E, Ros-Santaella JL, Garde JJ. Beyond tes- Alternative Med. 2013;2013:571420.
tis size: Links between spermatogenesis and sperm 74. Jarow JP, Chen H, Rosner TW, Trentacoste S,
traits in a seasonal breeding mammal. PloS One. Zirkin BR. Assessment of the androgen environ-
2015;10(10):e0139240. ment within the human testis: Minimally invasive
60. Petersen PM, Seieroe K, Pakkenberg B. The total method to obtain intratesticular fluid. J Androl.
number of Leydig and Sertoli cells in the testes of 2001;22(4):640–645.
men across various age groups – a stereological 75. McLachlan RI, O’Donnell L, Meachem SJ et al.
study. J Anat. 2015;226(2):175–179. Identification of specific sites of hormonal regula-
61. Richburg JH, Redenbach DM, Boekelheide K. tion in spermatogenesis in rats, monkeys, and man.
Seminiferous tubule fluid secretion is a Sertoli Recent Prog Horm Res. 2002;57:149–179.
cell microtubule-dependent process inhibited by 76. Haywood M, Spaliviero J, Jimemez M, King NJ,
2,5-hexanedione exposure. Toxicol Appl Pharmacol. Handelsman DJ, Allan CM. Sertoli and germ cell
1994;128(2):302–309. development in hypogonadal (hpg) mice express-
62. Walker WH, Cheng J. FSH and testosterone signal- ing transgenic follicle-stimulating hormone alone
ing in Sertoli cells. Reproduction. 2005;130(1):15–28. or in combination with testosterone. Endocrinology.
63. Meachem SJ, Nieschlag E, Simoni M. Inhibin B in 2003;144(2):509–517.
male reproduction: Pathophysiology and clinical rel- 77. Holdcraft RW, Braun RE. Hormonal regulation of
evance. Eur J Endocrinol. 2001;145(5):561–571. spermatogenesis. Int J Androl. 2004;27(6):335–342.
64. O’Donnell L, Stanton P, de Kretser DM. Endocrinology 78. Ruwanpura SM, McLachlan RI, Meachem SJ.
of the male reproductive system and spermatogene- Hormonal regulation of male germ cell develop-
sis. In De Groot LJ, Chrousos G, Dungan K et al., eds. ment. J Endocrinol. 2010;205(2):117–131.
Endotext [Internet]. South Dartmouth, MA: MDText​ 79. Singh J, O’Neill C, Handelsman DJ. Induction of sper-
.com. matogenesis by androgens in gonadotropin-deficient
65. White RB, Eisen JA, Kasten TL, Fernald RD. Second (hpg) mice. Endocrinology. 1995;136(12):5311–5321.
gene for gonadotropin-releasing hormone in humans. 80. Allan CM, Garcia A, Spaliviero J et al.
Proc Natl Acad Sci U S A. 1998;95(1):305–309. Complete Sertoli cell proliferation induced by
References 133

follicle-stimulating hormone (FSH) independently 96. Jarvi K, Lo K, Grober E et al. The workup and man-
of luteinizing hormone activity: Evidence from agement of azoospermic males. Can Urol Assoc J.
genetic models of isolated FSH action. Endocrinology. 2015;9(7–8):229–235.
2004;145(4):1587–1593. 97. Jarow JP, Espeland MA, Lipshultz LI. Evaluation of
81. Matthiesson KL, McLachlan RI, O’Donnell L et al. the azoospermic patient. J Urol. 1989;142(1):62–65.
The relative roles of follicle-stimulating hormone 98. Esteves SC. Clinical management of infertile men
and luteinizing hormone in maintaining spermato- with nonobstructive azoospermia. Asian J Androl.
gonial maturation and spermiation in normal men. 2015;17(3):459–470.
J Clin Endocrinol Metab. 2006;91(10):3962–3969. 99. Jarvi K, Lo K, Fischer A et al. CUA Guideline: The
82. Pitteloud N, Dwyer AA, DeCruz S et al. Inhibition workup of azoospermic males. Can Urol Assoc J.
of luteinizing hormone secretion by testosterone in 2010;4(3):163–167.
men requires aromatization for its pituitary but not 100. Wosnitzer M, Goldstein M, Hardy MP. Review of
its hypothalamic effects: Evidence from the tan- azoospermia. Spermatogenesis. 2014;4:e28218.
dem study of normal and gonadotropin-releasing 101. Tournaye H, Devroey P, Liu J, Nagy Z, Lissens W,
hormone-deficient men. J Clin Endocrinol Metab. Van Steirteghem A. Microsurgical epididymal sperm
2008;93(3):784–791. aspiration and intracytoplasmic sperm injection:
83. Shiraishi K, Matsuyama H. Local expression of epi- A new effective approach to infertility as a result of
dermal growth factor-like growth factors in human congenital bilateral absence of the vas deferens. Fertil
testis and its role in spermatogenesis. J Androl. Steril. 1994;61(6):1045–1051.
2012;33(1):66–73. 102. Schlegel PN, Palermo GD, Alikani M et al.
84. Thackray VG, Mellon PL, Coss D. Hormones in syn- Micropuncture retrieval of epididymal sperm with in
ergy: Regulation of the pituitary gonadotropin genes. vitro fertilization: Importance of in vitro microma-
Mol Cell Endocrinol. 2010;314(2):192–203. nipulation techniques. Urology. 1995;46(2):238–241.
85. Araujo AB, Wittert GA. Endocrinology of the 103. Bernie AM, Ramasamy R, Stember DS, Stahl PJ.
aging male. Best Pract Res Clin Endocrinol Metab. Microsurgical epididymal sperm aspiration: Indica­
2011;25(2):303–319. tions, techniques and outcomes. Asian J Androl. 2013;​
86. Franchimont P, Hazee-Hagelstein MT, Jaspar JM, 15(1):40–43.
Charlet-Renard C, Demoulin A. Inhibin and related 104. Meniru GI, Gorgy A, Batha S, Clarke RJ, Podsiadly
peptides. Mechanisms of action and regulation BT, Craft IL. Studies of percutaneous epididymal
of secretion. Rev Fr Gynecol Obstet. 1988;83(10):​ sperm aspiration (PESA) and intracytoplasmic sperm
607–611. injection. Hum Reprod Update. 1998;4(1):57–71.
87. Deffieux X, Antoine JM. [Inhibins, activins and anti- 105. Lin YM, Hsu CC, Kuo TC, Lin JS, Wang ST, Huang
Mullerian hormone: Structure, signalling pathways, KE. Percutaneous epididymal sperm aspiration versus
roles and predictive value in reproductive medicine]. microsurgical epididymal sperm aspiration for irrep-
Gynecol Obstet Fertil. 2003;31(11):900–911. arable obstructive azoospermia—Experience with
88. Crosnoe LE, Grober E, Ohl D, Kim ED. Exogenous 100 cases. J FormoS Med Assoc. 2000;99(6):459–465.
testosterone: A preventable cause of male infertility. 106. Schlegel PN. Testicular sperm extraction:
Transl Androl Urol. 2013;2(2):106–113. Microdissection improves sperm yield with minimal
89. Rambhatla A, Mills JN, Rajfer J. The role of estrogen tissue excision. Hum Reprod. 1999;14(1):131–135.
modulators in male hypogonadism and infertility. 107. McLachlan RI. Approach to the patient with oligo-
Rev Urol. 2016;18(2):66–72. zoospermia. J Clin Endocrinol Metabol. 2013;98(3):​
90. Palermo GD, Kocent J, Monahan D, Neri QV, 873–880.
Rosenwaks Z. Treatment of male infertility. Methods 108. Milardi D, Grande G, Sacchini D et al. Male fertil-
Mol Biol. 2014;1154:385–405. ity and reduction in semen parameters: A single
91. Cocuzza M, Agarwal A. Nonsurgical treatment tertiary-care center experience. Int J Endocrinol.
­
of male infertility: Specific and empiric therapy. 2012;​2012:649149.
Biologics. 2007;1(3):259–269. 109. Bhasin S. Approach to the infertile man. J Clin
92. Okabe M. The cell biology of mammalian fertiliza- Endocrinol Metabol. 2007;92(6):1995–2004.
tion. Development. 2013;140(22):4471–4479. 110. Jewett MA, Greenspan MB, Shier RM, Howatson AF.
93. Georgadaki K, Khoury N, Spandidos DA, Necrospermia or immotile cilia syndrome as a cause
Zoumpourlis V. The molecular basis of fertilization of male infertility. J Urol. 1980;124(2):292–293.
(Review). Int J Mol Med. 2016;38(4):979–986. 111. Amelar RD, Dubin L, Schoenfeld C. Sperm motility.
94. Vasan SS. Semen analysis and sperm function tests: Fertil Steril. 1980;34(3):197–215.
How much to test? Indian J Urol. 2011;27(1):41–48. 112. Curi SM, Ariagno JI, Chenlo PH et al. Astheno­
95. Cooper TG, Noonan E, von Eckardstein S et al. zoospermia: Analysis of a large population. Arch
World Health Organization reference values for Androl. 2003;49(5):343–349.
human semen characteristics. Hum Reprod Update. 113. Hajder M, Hajder E, Husic A. The effects of total
2010;16(3):231–245. motile sperm count on spontaneous pregnancy rate
134  Regulation of fertility and infertility in humans

and pregnancy after IUI treatment in couples with 130. Bertolla RP, Cedenho AP, Hassun Filho PA, Lima
male factor and unexplained infertility. Med Arch. SB, Ortiz V, Srougi M. Sperm nuclear DNA fragmen-
2016;70(1):39–43. tation in adolescents with varicocele. Fertil Steril.
114. Smith RP, Coward RM, Lipshultz LI. The office visit. 2006;85(3):625–628.
Urol Clin North Am. 2014;41(1):19–37. 131. Agarwal A, Cho CL, Esteves SC. Should we evalu-
115. Ortega C, Verheyen G, Raick D, Camus M, Devroey ate and treat sperm DNA fragmentation? Curr Opin
P, Tournaye H. Absolute asthenozoospermia and Obstet Gynecol. 2016;28(3):164–171.
ICSI: What are the options? Hum Reprod Update. 132. Greco E, Iacobelli M, Rienzi L, Ubaldi F, Ferrero
2011;17(5):684–692. S, Tesarik J. Reduction of the incidence of sperm
116. Cui D, Han G, Shang Y et al. Antisperm antibodies DNA fragmentation by oral antioxidant treatment.
in infertile men and their effect on semen param- J Androl. 2005;26(3):349–353.
eters: A systematic review and meta-analysis. Clin 133. Pons I, Cercas R, Villas C, Brana C, Fernandez-Shaw
Chim Acta. 2015;444:29–36. S. One abstinence day decreases sperm DNA frag-
117. Yeh WR, Acosta AA, Seltman HJ, Doncel G. Impact mentation in 90% of selected patients. J Assist Reprod
of immunoglobulin isotype and sperm surface loca- Genet. 2013;30(9):1211–1218.
tion of antisperm antibodies on fertilization in vitro 134. Zini A, San Gabriel M, Baazeem A. Antioxidants and
in the human. Fertil Steril. 1995;63(6):1287–1292. sperm DNA damage: A clinical perspective. J Assist
118. Zini A, Fahmy N, Belzile E, Ciampi A, Al-Hathal Reprod Genet. 2009;26(8):427–432.
N, Kotb A. Antisperm antibodies are not asso- 135. Majzoub A, Agarwal A, Esteves SC. Antioxidants for
ciated with pregnancy rates after IVF and ICSI: elevated sperm DNA fragmentation: A mini review.
Systematic review and meta-analysis. Hum Reprod. Trans Androl Urol. 2017:1–5.
2011;26(6):1288–1295. 136. Walsh TJ, Pera RR, Turek PJ. The genetics of male
119. Mishra M, Kumar N, Jaiswal A, Verma AK, Kant S. infertility. Semin Reprod Med. 2009;27(2):124–136.
Kartagener’s syndrome: A case series. Lung India. 137. Suganya J, Kujur SB, Selvaraj K, Suruli MS, Haripriya
2012;29(4):366–369. G, Samuel CR. Chromosomal abnormalities in infer-
120. Wang YX, Yang SW, Qu CB et al. [L-carnitine: Safe tile men from Southern India. J Clin Diagn Res.
and effective for asthenozoospermia]. Zhonghua Nan 2015;9(7):GC05–10.
Ke Xue. 2010;16(5):420–422. 138. Abramsky L, Chapple J. 47,XXY (Klinefelter syn-
121. Szczygiel M, Kurpisz M. Teratozoospermia and its drome) and 47,XYY: Estimated rates of and indication
effect on male fertility potential. Andrologia. 1999;​ for postnatal diagnosis with implications for prena-
31(2):63–75. tal counselling. Prenat Diagn. 1997;17(4):363–368.
122. Kruger TF, Menkveld R, Stander FS et al. Sperm 139. Stahl PJ, Schlegel PN. Genetic evaluation of the azo-
morphologic features as a prognostic factor in in ospermic or severely oligozoospermic male. Curr
vitro fertilization. Fertil Steril. 1986;46(6):1118–1123. Opin Obstet Gynecol. 2012;24(4):221–228.
123. Gatimel N, Moreau J, Parinaud J, Leandri RD. Sperm 140. Wosnitzer MS, Paduch DA. Endocrinological issues
morphology: Assessment, pathophysiology, clinical and hormonal manipulation in children and men
relevance, and state of the art in 2017. Andrology. with Klinefelter syndrome. Am J Med Genet C Semin
2017. Med Genet. 2013;163C(1):16–26.
124. Brucker C, Lipford GB. The human sperm acrosome 141. Aksglaede L, Wikstrom AM, Rajpert-De Meyts E,
reaction: Physiology and regulatory mechanisms. An Dunkel L, Skakkebaek NE, Juul A. Natural history
update. Hum Reprod Update. 1995;1(1):51–62. of seminiferous tubule degeneration in Klinefelter
125. Zaneveld LJ, De Jonge CJ, Anderson RA, Mack SR. syndrome. Hum Reprod Update. 2006;12(1):39–48.
Human sperm capacitation and the acrosome reac- 142. Bonomi M, Rochira V, Pasquali D et al. Klinefelter syn-
tion. Hum rReprod. 1991;6(9):1265–1274. drome (KS): Genetics, clinical phenotype and hypo-
126. Baker HW, Liu DY, Garrett C, Martic M. The human gonadism. J Endocrinol Invest. 2017;40(2):123–134.
acrosome reaction. Asian J Androl. 2000;2(3):172–178. 143. Abdel-Razic MM, Abdel-Hamid IA, ElSobky ES.
127. Evgeni E, Charalabopoulos K, Asimakopoulos B. Nonmosaic 47,XYY syndrome presenting with male
Human sperm DNA fragmentation and its correla- infertility: Case series. Andrologia. 2012;44(3):200–204.
tion with conventional semen parameters. J Reprod 144. Vogt PH, Edelmann A, Kirsch S et al. Human Y
Infertil. 2014;15(1):2–14. chromosome azoospermia factors (AZF) mapped to
128. Ribas-Maynou J, Garcia-Peiro A, Fernandez-Encinas different subregions in Yq11. Hum Mol Genet. 1996;​
A et al. Comprehensive analysis of sperm DNA frag- 5(7):933–943.
mentation by five different assays: TUNEL assay, 145. Vogt PH. AZF deletions and Y chromosomal hap-
SCSA, SCD test and alkaline and neutral Comet logroups: History and update based on sequence.
assay. Andrology. 2013;1(5):715–722. Hum Reprod Update. 2005;11(4):319–336.
129. Evenson D, Wixon R. Meta-analysis of sperm DNA 146. Krausz C, Forti G, McElreavey K. The Y chromo-
fragmentation using the sperm chromatin structure some and male fertility and infertility. Int J Androl.
assay. Reprod Biomed Online. 2006;12(4):466–472. 2003;26(2):70–75.
References 135

147. Georgiou I, Syrrou M, Pardalidis N et al. Genetic and 164. Zimmermann C, Romero Y, Warnefors M et al.
epigenetic risks of intracytoplasmic sperm injection Germ cell-specific targeting of DICER or DGCR8
method. Asian J Androl. 2006;8(6):643–673. reveals a novel role for endo-siRNAs in the progres-
148. Silveira LF, MacColl GS, Bouloux PM. Hypo­ sion of mammalian spermatogenesis and male fertil-
gonadotropic hypogonadism. Semin Reprod Med. ity. PLoS One. 2014;9(9):e107023.
2002;20(4):327–338. 165. Zhang C, Gao L, Xu EY. LncRNA, a new compo-
149. Dode C, Levilliers J, Dupont JM et al. Loss-of- nent of expanding RNA-protein regulatory network
function mutations in FGFR1 cause autosomal dom- important for animal sperm development. Semin
inant Kallmann syndrome. Nat Genet. 2003;33(4):​ Cell Dev Biol. 2016;59:110–117.
463–465. 166. Dolci S, Grimaldi P, Geremia R, Pesce M, Rossi P.
150. Yatsenko AN, Georgiadis AP, Ropke A et al. X-linked Identification of a promoter region generating Sry
TEX11 mutations, meiotic arrest, and azoospermia in circular transcripts both in germ cells from male
infertile men. N Engl J Med. 2015;372(22):2097–2107. adult mice and in male mouse embryonal gonads.
151. Adelman CA, Petrini JH. ZIP4H (TEX11) deficiency Biol Reprod. 1997;57(5):1128–1135.
in the mouse impairs meiotic double strand break 167. Hansen TB, Jensen TI, Clausen BH et al. Natural
repair and the regulation of crossing over. PLoS RNA circles function as efficient microRNA sponges.
Genet. 2008;4(3):e1000042. Nature. 2013;495(7441):384–388.
152. McGinniss MJ, Chen C, Redman JB et al. Extensive 168. Gorelick JI, Goldstein M. Loss of fertility in men
sequencing of the CFTR gene: Lessons learned with varicocele. Fertil Steril. 1993;59(3):613–616.
from the first 157 patient samples. Hum Genet. 169. Wang YJ, Zhang RQ, Lin YJ, Zhang RG, Zhang WL.
2005;118(3–4):331–338. Relationship between varicocele and sperm DNA
153. Kim VN, Han J, Siomi MC. Biogenesis of small RNAs damage and the effect of varicocele repair: A meta-
in animals. Nat Rev Mol Cell Biol. 2009;10(2):126–139. analysis. Reprod Biomed Online. 2012;25(3):307–314.
154. Meikar O, Da Ros M, Korhonen H, Kotaja N. 170. Sedaghatpour D, Berookhim BM. The role of vari-
Chromatoid body and small RNAs in male germ cocele in male factor subfertility. Curr Urol Rep.
cells. Reproduction. 2011;142(2):195–209. 2017;18(9):73.
155. De Fazio S, Bartonicek N, Di Giacomo M et al. The 171. Schauer I, Madersbacher S, Jost R, Hubner WA, Imhof
endonuclease activity of Mili fuels piRNA ampli- M. The impact of varicocelectomy on sperm param-
fication that silences LINE1 elements. Nature. eters: A meta-analysis. J Urol. 2012;187(5):1540–1547.
2011;480(7376):259–263. 172. Wang Y, Zhang W, Li D. [High ligation of varico-
156. Kuramochi-Miyagawa S, Watanabe T, Gotoh K et al. cele improves sperm DNA integrity in patients with
DNA methylation of retrotransposon genes is regu- asthenospermia]. Zhong Nan Da Xue Xue Bao Yi Xue
lated by Piwi family members MILI and MIWI2 in Ban. 2012;37(12):1228–1232.
murine fetal testes. Genes Dev. 2008;22(7):908–917. 173. Guo TH, Tong XH, Luo LH, Luan HB, Zhou GX,
157. Siomi MC, Sato K, Pezic D, Aravin AA. PIWI- Wan YY. Value of microsurgical varicocelectomy for
interacting small RNAs: The vanguard of genome severe oligo-asthenospermia patients failed in fertil-
defence. Nat Rev Mol Cell Biol. 2011;12(4):246–258. ization assisted by in vitro fertilization. Eur Rev Med
158. Reuter M, Berninger P, Chuma S et al. Miwi catalysis is Pharmacol Sci. 2016;20(9):1669–1674.
required for piRNA amplification-independent LINE1 174. Daitch JA, Bedaiwy MA, Pasqualotto EB et al.
transposon silencing. Nature. 2011;480(7376):264–267. Varicocelectomy improves intrauterine insemina-
159. Di Giacomo M, Comazzetto S, Saini H et al. Multiple tion success rates in men with varicocele. J Urol.
epigenetic mechanisms and the piRNA pathway 2001;165(5):1510–1513.
enforce LINE1 silencing during adult spermatogen- 175. Mansour Ghanaie M, Asgari SA, Dadrass N,
esis. Mol Cell. 2013;50(4):601–608. Allahkhah A, Iran-Pour E, Safarinejad MR. Effects
160. Gou LT, Dai P, Yang JH et al. Pachytene piRNAs of varicocele repair on spontaneous first trimester
instruct massive mRNA elimination during late miscarriage: Arandomized clinical trial. Urol J. 2012;​
spermiogenesis. Cell Res. 2014;24(6):680–700. 9(2):505–513.
161. Greenlee AR, Shiao MS, Snyder E et al. Deregulated 176. Wang C, Cui YG, Wang XH et al. transient scro-
sex chromosome gene expression with male germ cell- tal hyperthermia and levonorgestrel enhance
specific loss of Dicer1. PLoS One. 2012;7(10):e46359. testosterone-­induced spermatogenesis suppression
162. Romero Y, Meikar O, Papaioannou MD et al. Dicer1 in men through increased germ cell apoptosis. J Clin
depletion in male germ cells leads to infertility due to Endocrinol Metab. 2007;92(8):3292–3304.
cumulative meiotic and spermiogenic defects. PLoS 177. Shiraishi K, Takihara H, Matsuyama H. Elevated
One. 2011;6(10):e25241. scrotal temperature, but not varicocele grade, reflects
163. Wu Q, Song R, Ortogero N et al. The RNase III testicular oxidative stress-mediated apoptosis. World
enzyme DROSHA is essential for microRNA produc- J Urol. 2010;28(3):359–364.
tion and spermatogenesis. J Biol Chem. 2012;287(30):​ 178. Camargo M, Intasqui P, Bertolla RP. Proteomic pro-
25173–25190. file of seminal plasma in adolescents and adults with
136  Regulation of fertility and infertility in humans

treated and untreated varicocele. Asian J Androl. 194. Kort HI, Massey JB, Elsner CW et al. Impact of body
2016;18(2):194–201. mass index values on sperm quantity and quality.
179. Drabovich AP, Saraon P, Jarvi K, Diamandis EP. J Androl. 2006;27(3):450–452.
Seminal plasma as a diagnostic fluid for male 195. La Vignera S, Condorelli RA, Vicari E, Calogero AE.
reproductive system disorders. Nat Rev Urol. Negative effect of increased body weight on sperm
2014;11(5):278–288. conventional and nonconventional flow cytometric
180. Cocuzza M, Athayde KS, Agarwal A et al. Impact sperm parameters. J Androl. 2012;33(1):53–58.
of clinical varicocele and testis size on seminal 196. Hofny ER, Ali ME, Abdel-Hafez HZ et al. Semen
reactive oxygen species levels in a fertile popula- parameters and hormonal profile in obese fertile and
tion: A prospective controlled study. Fertil Steril. infertile males. Fertil Steril. 2010;94(2):581–584.
2008;90(4):1103–1108. 197. Hammoud AO, Wilde N, Gibson M, Parks A, Carrell
181. Zylbersztejn DS, Andreoni C, Del Giudice PT et DT, Meikle AW. Male obesity and alteration in sperm
al. Proteomic analysis of seminal plasma in ado- parameters. Fertil Steril. 2008;90(6):2222–2225.
lescents with and without varicocele. Fertil Steril. 198. Chavarro JE, Toth TL, Wright DL, Meeker JD,
2013;99(1):92–98. Hauser R. Body mass index in relation to semen
182. Del Giudice PT, da Silva BF, Lo Turco EG et al. quality, sperm DNA integrity, and serum reproduc-
Changes in the seminal plasma proteome of adoles- tive hormone levels among men attending an infer-
cents before and after varicocelectomy. Fertil Steril. tility clinic. Fertil Steril. 2010;93(7):2222–2231.
2013;100(3):667–672. 199. de Boer H, Verschoor L, Ruinemans-Koerts J, Jansen
183. Lampinen R, Vehvilainen-Julkunen K, Kankkunen M. Letrozole normalizes serum testosterone in
P. A review of pregnancy in women over 35 years of severely obese men with hypogonadotropic hypogo-
age. Open Nurs J. 2009;3:33–38. nadism. Diabetes Obes Metab. 2005;7(3):211–215.
184. George K, Kamath MS. Fertility and age. J Hum 200. Burger HG. Physiological principles of endocrine
Reprod Scie. 2010;3(3):121–123. replacement: Estrogen. Horm Res. 2001;56 Suppl 1:​
185. Liu K, Case A; Reproductive endo­crinology and infer- 82–85.
tility committee. Advanced reproductive age and fer- 201. Giagulli VA, Kaufman JM, Vermeulen A. Pathogenesis
tility. J Obstet Gynaecol Can. 2011;33(11):1165–1175. of the decreased androgen levels in obese men. J Clin
186. Sharma R, Agarwal A, Rohra VK, Assidi M, Abu- Endocrinol Metab. 1994;79(4):997–1000.
Elmagd M, Turki RF. Effects of increased paternal 202. Gutorova NV, Kleshchyov MA, Tipisova EV,
age on sperm quality, reproductive outcome and Osadchuk LV. Effects of overweight and obesity on
associated epigenetic risks to offspring. Reprod Biol the spermogram values and levels of reproductive
Endocrinol. 2015;13:35. hormones in the male population of the European
187. Nybo Andersen AM, Hansen KD, Andersen PK, north of Russia. Bull Exp Biol Med. 2014;157(1):95–98.
Davey Smith G. Advanced paternal age and risk 203. Cavaco JE, Laurentino SS, Barros A, Sousa M,
of fetal death: A cohort study. Am J Epidemiol. Socorro S. Estrogen receptors alpha and beta in
2004;160(12):1214–1222. human testis: Both isoforms are expressed. Syst Biol
188. Dockerty JD, Draper G, Vincent T, Rowan SD, Reprod Med. 2009;55(4):137–144.
Bunch KJ. Case-control study of parental age, par- 204. Rodriguez JJ, Filipiak K, Maslyk M et al. Towards
ity and socioeconomic level in relation to childhood beta-selectivity in functional estrogen receptor antag-
cancers. Int J Epidemiol. 2001;30(6):1428–1437. onists. Org Biomol Chem. 2012;10(36):7334–7346.
189. Murray L, McCarron P, Bailie K et al. Association of 205. Rago V, Romeo F, Giordano F, Maggiolini M,
early life factors and acute lymphoblastic leukaemia Carpino A. Identification of the estrogen receptor
in childhood: Historical cohort study. Br J Cancer. GPER in neoplastic and non-neoplastic human tes-
2002;86(3):356–361. tes. Reprod Biol Endocrinol. 2011;9:135.
190. Ogden CL, Carroll MD, Kit BK, Flegal KM. 206. Sandner F, Welter H, Schwarzer JU, Kohn FM,
Prevalence  of childhood and adult obesity in the Urbanski HF, Mayerhofer A. Expression of the oes-
United States, 2011–2012. JAMA. 2014;311(8):​806–814. trogen receptor GPER by testicular peritubular cells
191. Hammoud AO, Gibson M, Peterson CM, Hamilton is linked to sexual maturation and male fertility.
BD, Carrell DT. Obesity and male reproductive Andrology. 2014;2(5):695–701.
potential. J Androl. 2006;27(5):619–626. 207. Rato L, Alves MG, Cavaco JE, Oliveira PF. High-
192. Koloszar S, Fejes I, Zavaczki Z, Daru J, Szollosi energy diets: A threat for male fertility? Obes Rev.
J, Pal A. Effect of body weight on sperm concen- 2014;15(12):996–1007.
tration in normozoospermic males. Arch Androl. 208. La Vignera S, Condorelli R, Vicari E, D’Agata R,
2005;51(4):299–304. Calogero AE. Diabetes mellitus and sperm param-
193. Jensen TK, Andersson AM, Jorgensen N et al. Body eters. J Androl. 2012;33(2):145–153.
mass index in relation to semen quality and repro- 209. Samplaski MK, Nangia AK. Adverse effects of com-
ductive hormones among 1,558 Danish men. Fertil mon medications on male fertility. Nat Rev Urol.
Steril. 2004;82(4):863–870. 2015;12(7):401–413.
References 137

210. Gonzalo IT, Swerdloff RS, Nelson AL et al. 224. Saha L, Bhargava VK, Garg SK, Majumdar S. Effect of
Levonorgestrel implants (Norplant II) for male con- nimodipine on male reproductive functions in rats.
traception clinical trials: Combination with trans- Indian J Physiol Pharmacol. 2000;44(4):449–455.
dermal and injectable testosterone. J Clin Endocrinol 225. Lee JH, Ahn HJ, Lee SJ, Gye MC, Min CK. Effects
Metab. 2002;87(8):3562–3572. of L- and T-type Ca(2)(+) channel blockers on sper-
211. Hellstrom WJ, Sikka SC. Effects of acute treatment matogenesis and steroidogenesis in the prepubertal
with tamsulosin versus alfuzosin on ejaculatory mouse testis. J Assist Reprod Genet. 2011;28(1):​23–30.
function in normal volunteers. J Urol. 2006;176(4 Pt 226. Feng HL, Han YB, Hershlag A, Zheng LJ. Impact of
1):1529–1533. Ca2+ flux inhibitors on acrosome reaction of ham-
212. Hellstrom WJ, Sikka SC. Effects of alfuzosin and ster spermatozoa. J Androl. 2007;28(4):561–564.
tamsulosin on sperm parameters in healthy men: 227. Nelson WO, Bunge RG. The effect of therapeutic dos-
Results of a short-term, randomized, double-blind, ages of nitrofurantoin (furadantin) upon spermato-
placebo-controlled, crossover study. J Androl. 2009;​ genesis in man. J Urol. 1957;77(2):275–281.
30(4):469–474. 228. Hargreaves CA, Rogers S, Hills F, Rahman F,
213. Amory JK, Wang C, Swerdloff RS et al. The effect Howell RJ, Homa ST. Effects of co-trimoxazole,
of 5alpha-reductase inhibition with dutasteride and erythromycin, amoxycillin, tetracycline and chlo-
finasteride on semen parameters and serum hor- roquine on sperm function in vitro. Hum Reprod.
mones in healthy men. J Clin Endocrinol Metab. 1998;13(7):1878–1886.
2007;92(5):1659–1665. 229. White IG. The toxicity of some antibacterials for bull,
214. Kaufman KD, Olsen EA, Whiting D et al. Finasteride ram, rabbit and human spermatozoa. Aust J Exp Biol
in the treatment of men with androgenetic alopecia. Med Sci. 1954;32(1):41–48.
Finasteride Male Pattern Hair Loss Study Group. 230. Timmermans L. Influence of antibiotics on sper-
J Am Acad Dermatol. 1998;39(4 Pt 1):578–589. matogenesis. J Urol. 1974;112(3):348–349.
215. Montejo AL, Llorca G, Izquierdo JA, Rico- 231. Schlegel PN, Chang TS, Marshall FF. Antibiotics:
Villademoros F. Incidence of sexual dysfunction Potential hazards to male fertility. Fertil Steril.
associated with antidepressant agents: A prospec- 1991;55(2):235–242.
tive multicenter study of 1022 outpatients. Spanish 232. Sutyak KE, Anderson RA, Dover SE et al. Spermicidal
Working Group for the Study of Psychotropic-Related activity of the safe natural antimicrobial peptide sub-
Sexual Dysfunction. J Clin Psychiatry. 2001;62 Suppl tilosin. Infect Dis Obstet Gynecol. 2008;2008:540758.
3:10–21. 233. Pont A, Williams PL, Azhar S et al. Ketoconazole
216. Tanrikut C, Schlegel PN. Antidepressant-associated blocks testosterone synthesis. Arch Intern Med. 1982;​
changes in semen parameters. Urology. 2007;69(1):185 142(12):2137–2140.
e185–187. 234. Boekelheide K. Mechanisms of toxic damage to sper-
217. Safarinejad MR. Sperm DNA damage and semen matogenesis. J Natl Cancer Inst Monogr. 2005(34):6–8.
quality impairment after treatment with selective 235. Wallace WH, Anderson RA, Irvine DS. Fertility
serotonin reuptake inhibitors detected using semen preservation for young patients with cancer: Who
analysis and sperm chromatin structure assay. J Urol. is at risk and what can be offered? Lancet Oncol.
2008;180(5):2124–2128. 2005;6(4):209–218.
218. Tanrikut C, Feldman AS, Altemus M, Paduch DA, 236. Gambacorti-Passerini C, Tornaghi L, Cavagnini F
Schlegel PN. Adverse effect of paroxetine on sperm. et al. Gynaecomastia in men with chronic myeloid
Fertil Steril. 2010;94(3):1021–1026. leukaemia after imatinib. Lancet. 2003;361(9373):​
219. Levin RM, Amsterdam JD, Winokur A, Wein AJ. 1954–1956.
Effects of psychotropic drugs on human sperm 237. Oliva A, Spira A, Multigner L. Contribution of envi-
motility. Fertil Steril. 1981;36(4):503–506. ronmental factors to the risk of male infertility.
220. Ko DT, Hebert PR, Coffey CS, Sedrakyan A, Curtis Human reproduction. 2001;16(8):1768–1776.
JP, Krumholz HM. Beta-blocker therapy and symp- 238. Chia SE, Lim ST, Tay SK, Lim ST. Factors associated
toms of depression, fatigue, and sexual dysfunction. with male infertility: A case-control study of 218 infer-
JAMA. 2002;288(3):351–357. tile and 240 fertile men. BJOG. 2000;107(1):55–61.
221. White DR, Clarkson JS, Ratnasooriya WD,  Aitken 239. Checa Vizcaino MA, Gonzalez-Comadran M,
RJ. Complementary effects of propranolol  and Jacquemin B. Outdoor air pollution and human
­nonoxynol-9 upon human sperm motility. Contra­ infertility: A systematic review. Fertil Steril. 2016;​106​
ception. 1995;52(4):241–247. (4):897–904 e891.
222. Clark E. Spironolactone therapy and gynecomastia. 240. Carre J, Gatimel N, Moreau J, Parinaud J, Leandri R.
JAMA. 1965;193:163–164. Does air pollution play a role in infertility?: A sys-
223. Caminos-Torres R, Ma L, Snyder PJ. Gynecomastia tematic review. Environ Health. 2017;16(1):82.
and semen abnormalities induced by spironolac- 241. Mahalingaiah S, Hart JE, Laden F et al. Adult air pol-
tone in normal men. J Clin Endocrinol Metab. 1977;​ lution exposure and risk of infertility in the Nurses’
45(2):255–260. Health Study II. Hum Reprod. 2016;31(3):638–647.
138  Regulation of fertility and infertility in humans

242. Quintanilla-Vega B, Hoover DJ, Bal W, Silbergeld EK, 253. Lucas B, Fields C, Hofmann MC. Signaling path-
Waalkes MP, Anderson LD. Lead interaction with ways in spermatogonial stem cells and their disrup-
human protamine (HP2) as a mechanism of male repro- tion by toxicants. Birth Defects Res C Embryo Today.
ductive toxicity. Chem Res Toxicol. 2000;13(7):​594–600. 2009;87(1):35–42.
243. Chung NP, Cheng CY. Is cadmium chloride-induced 254. Meeker JD, Ehrlich S, Toth TL et al. Semen qual-
inter-Sertoli tight junction permeability barrier dis- ity and sperm DNA damage in relation to urinary
ruption a suitable in vitro model to study the events bisphenol A among men from an infertility clinic.
of junction disassembly during spermatogenesis in Reprod Toxicol. 2010;30(4):532–539.
the rat testis? Endocrinology. 2001;​142(5):​1878–1888. 255. Lassen TH, Frederiksen H, Jensen TK et al. Urinary
244. Siu ER, Wong EW, Mruk DD, Sze KL, Porto CS, Cheng bisphenol A levels in young men: Association with
CY. An occludin-focal adhesion kinase protein com- reproductive hormones and semen quality. Environ
plex at the blood-testis barrier: A study using the cad- Health Perspect. 2014;122(5):478–484.
mium model. Endocrinology. 2009;150(7):​3336–3344. 256. Tournaye H. Male factor infertility and ART. Asian J
245. Ratcliffe JM, Schrader SM, Steenland K, Clapp DE, Androl. 2012;14(1):103–108.
Turner T, Hornung RW. Semen quality in papaya 257. Van Voorhis BJ, Barnett M, Sparks AE, Syrop CH,
workers with long term exposure to ethylene dibro- Rosenthal G, Dawson J. Effect of the total motile
mide. Br J Ind Med. 1987;44(5):317–326. sperm count on the efficacy and cost-effectiveness of
246. Schrader SM, Turner TW, Ratcliffe JM. The effects of intrauterine insemination and in vitro fertilization.
ethylene dibromide on semen quality: A comparison Fertil Steril. 2001;75(4):661–668.
of short-term and chronic exposure. Reprod Toxicol. 258. Merviel P, Heraud MH, Grenier N, Lourdel E,
1988;2(3–4):191–198. Sanguinet P, Copin H. Predictive factors for preg-
247. Potashnik G, Porath A. Dibromochloropropane nancy after intrauterine insemination (IUI): An
(DBCP): A 17-year reassessment of testicular func- analysis of 1038 cycles and a review of the literature.
tion and reproductive performance. J Occup Environ Fertil Steril. 2010;93(1):79–88.
Med. 1995;37(11):1287–1292. 259. Michelmann HW. Minimal criteria of sperm qual-
248. Easley CAt, Bradner JM, Moser A et al. Assessing ity for insemination and IVF therapy. Int J Androl.
reproductive toxicity of two environmental toxicants 1995;18 Suppl 2:81–87.
with a novel in vitro human spermatogenic model. 260. Lopez G, Lafuente R, Checa MA, Carreras R,
Stem Cell Res. 2015;14(3):347–355. Brassesco M. Diagnostic value of sperm DNA frag-
249. el-Sabeawy F, Wang S, Overstreet J, Miller M, Lasley mentation and sperm high-magnification for pre-
B, Enan E. Treatment of rats during pubertal devel- dicting outcome of assisted reproduction treatment.
opment with 2,3,7,8-tetrachlorodibenzo-p-dioxin Asian J Androl. 2013;15(6):790–794.
alters both signaling kinase activities and epidermal 261. Merchant R, Gandhi G, Allahbadia GN. In vitro
growth factor receptor binding in the testis and the fertilization/intracytoplasmic sperm injection for
­
motility and acrosomal reaction of sperm. Toxicol male infertility. Indian J Urol. 2011;27(1):121–132.
Appl Pharmacol. 1998;150(2):427–442. 262. Di Santo M, Tarozzi N, Nadalini M, Borini A.
250. Wong EW, Cheng CY. Impacts of environmental Human sperm cryopreservation: Update on tech-
toxicants on male reproductive dysfunction. Trends niques, effect on DNA integrity, and implications for
Pharmacol Sci. 2011;32(5):290–299. ART. Adv Urol. 2012;2012:854–837.
251. Van Waeleghem K, De Clercq N, Vermeulen L, 263. Argyle CE, Harper JC, Davies MC. Oocyte cryo-
Schoonjans F, Comhaire F. Deterioration of sperm preservation: Where are we now? Hum Rreprod
quality in young healthy Belgian men. Hum Reprod. Update. 2016;22(4):440–449.
1996;11(2):325–329. 264. Dickey RP, Pyrzak R, Lu PY, Taylor SN, Rye PH.
252. Ge RS, Chen GR, Tanrikut C, Hardy MP. Phthalate Comparison of the sperm quality necessary for suc-
ester toxicity in Leydig cells: Developmental timing cessful intrauterine insemination with World Health
and dosage considerations. Reprod Toxicol. 2007;​ Organization threshold values for normal sperm.
23(3):366–373. Fertil Steril. 1999;71(4):684–689.
Male infertility
Evaluation and treatment
11
RYAN FLANNIGAN and MARC GOLDSTEIN

INTRODUCTION a detailed list of key considerations to elicit on history. Here


Approximately 15% of couples seek evaluation for infer- it is important to consider that a full cycle of spermatogen-
tility.1 Male factors contribute to 50% of infertile couples esis and spermiation takes approximately 72 days; thus,
and are thought to be primarily responsible in 20%.2 Some exposures or illnesses within the past 3 months may impact
forms of infertility can be managed and result in natural present fertility and semen analyses. It is also important to
pregnancies; however, more severe forms of infertility may ensure that the female partner obtains an appropriate eval-
necessitate assisted reproductive technologies (ARTs), pri- uation, and the male specialist knows the female partner’s
marily in vitro fertilization (IVF) with intracytoplasmic age, past reproductive history, and abnormalities on gyne-
sperm injection (ICSI), genetic counseling, donor sperm, cologic evaluation. Beyond evaluating factors affecting fer-
or adoption. In 2014, nearly 170,000 IVF procedures were tility potential, it is essential to determine the couple’s goals
utilized in the United States.3 In this chapter, we will iden- of fertility, including the number of children desired and
tify appropriate evaluation and management options for the timeline they wish to achieve pregnancies.
men presenting with infertility.
Physical exam
EVALUATION All patients should receive a general physical exam.
The objective of evaluating men presenting with infertility is Particular focus should be made to body habitus, hair dis-
threefold: (1) identify modifiable factors to improve fertility tribution, secondary sex characteristics, and presence of
through behavioral change, medical, and/or surgical thera- gynecomastia, stature, and midline defects. An abdominal
pies, (2) identify nonmodifiable factors affecting fertility examination should be performed to assess for signs of pre-
potential, and (3) provide appropriate counseling, expecta- vious abdominal or inguinal surgical scars. Penile examina-
tions, and resources to men and their partners based on their tion should be performed to assess the location of the meatal
evaluation. Couples unable to achieve pregnancy within opening. Scrotal examination should be performed in a
12 months of regular unprotected intercourse should seek warm and comfortable environment to encourage relaxation
reproductive evaluation. Earlier evaluation is warranted of the scrotal dartos muscle, making examination easier and
after 6 months of regular intercourse among couples where more accurate. Locally, we use a heating pad on the scrotum
the female is greater than 35 years of age. Furthermore, men for 2–5 minutes to facilitate scrotal relaxation. The testicles
with medical comorbidities known to impact fertility or are palpated for size and consistency; we recommend using
men with concerns regarding future fertility also warrant an orchidometer for accurate assessment of testicular size.
evaluation. Nonreproductive specialists may conduct initial The epididymis is palpated for fullness, induration, and ten-
screening on history, physical exam, and semen analysis prior derness. The spermatic cord should be palpated while the
to referral to a specialist in the case of any abnormalities. patient is both supine and standing. The vas deferens are
All men undergoing fertility evaluation should initially palpated bilaterally and the cords are assessed for varico-
undergo a history, physical examination, and semen anal- celes with and without Valsalva upright. If the presence of
ysis. Abnormalities may be further assessed with special- varicocele is unclear, the patient should be placed supine and
ized investigations in the following sections. reexamined; if a varicocele is present, the cord will collapse
when supine and become distended when standing.
History
A male reproductive history involves evaluating the couple’s INVESTIGATIONS
experience in attempting fertility (i.e., history of present- Semen analysis
ing illness), their social history for behavioral and envi- Beyond history and physical examination, semen analy-
ronmental factors potentially affecting fertility potential, sis is the first investigation performed in evaluating a
past medical history for conditions predisposing one to man’s fertility status. The present reference values for
poor reproductive health or abnormal mechanics of inter- semen analyses arise from the World Health Organization
course (i.e., erectile dysfunction, intravaginal ejaculation), (WHO) 2010 iteration. It is important to recognize how
past pelvic, abdominal, inguinal, or scrotal surgical history, these values were derived and the limitations they pose.
medication history affecting spermatogenesis, emission and Here, the study population was defined as men able to con-
ejaculation or erectile function, family history of infertility ceive a pregnancy within 12 month of attempting regular
or predisposing conditions, and allergies. Table 11.1 outlines unprotected intercourse.6 One thousand nine hundred

139
140  Male infertility

Table 11.1  Key considerations to elicit on history.


Type of history Key considerations
History of presenting illness • Duration of regular unprotected intercourse
• Use of aids or devices for fertility (i.e., ovulation kits)
• Mechanics of intercourse (i.e., intravaginal ejaculation)
• Previous paternal or maternal pregnancies
• Erectile function
• Ejaculatory history
Social • Smoking
• Recreational drug use
• Anabolic steroids
• Exercise (i.e., sedentary or prolonged endurance activity)
• Dietary history
• Heat exposures (i.e., >20-min hot shower, hot tubs, laptop use on thighs)
• Tight underwear
Medical • Abdominal or pelvic radiation therapy4
• Germ cells are significantly more sensitive than somatic cells
• Doses as low as 0.1–1.2 Gy may impair spermatogenesis
• >4 Gy may cause permanent damage
• 20–30 Gy may cause Leydig cell dysfunction
• Diabetes
• Obesity
• Galactorrhea
• Visual field deficits
• Central hypothalamic or pituitary dysfunction
• i.e., Kallman syndrome, pituitary adenoma, prolactinoma
• Klinefelter syndrome
• Sinus and respiratory infections
• Undescended testicle
• Testicular torsion
• Orchitis or epididymitis
• Testicular or pelvic trauma
• Systemic illness or fever
• Age of puberty
• Malignancies
• Reviewed in depth in Wallace et al. 20054
Surgical • Previous abdominal surgery affecting erections or ejaculation
• i.e., retroperitoneal lymph node dissection, rectal surgery, radical
prostatectomy, transurethral resection, or ablation of prostate
• Inguinal surgery or scrotal surgery compromising vas deferens, epididymis,
and/or testicular blood supply
• .i.e., inguinal hernia repair, hydrocelectomy, spermatocelectomy,
orchidopexy, vasectomy
Medications *This topic is reviewed in depth in Samplaski and Nangia 20155
Hormonal medications
• Exogenous testosterone or anabolic steroids
• Suppression of spermatogenesis
• Supplement use
• Various effects
Prostate medications
• Alpha blockers
• Retrograde ejaculation
• 5 alpha-reductase inhibitors
• Reduce sperm counts in 5% of men, sexual dysfunction
(Continued )
Investigations 141

Table 11.1 (Continued)  Key considerations to elicit on history.


Type of history Key considerations
Psychiatry related
• Selective 5-hydroxytryptamine reuptake inhibitors (i.e., SSRIs, SNRIs, TCAs,
MAOIs)
• Sperm DNA fragmentation, erectile and ejaculatory dysfunction
• Lithium
• Reduced sperm viability
Antihypertensives
• Beta blockers
• Sexual dysfunction, impaired sperm motility
• Diuretics
• Sexual dysfunction, impaired sperm motility and concentration
• Calcium channel blockers
• Decreased sperm concentration, motility and acrosome reaction
• ACE inhibitors
Antibiotics
• Trimethoprim-sulfamethoxazole
• Nitrofurantoin
• Spermatogenic arrest, reduced sperm concentration
• Macrolides
• Impaired sperm motility and viability
• Aminoglycosides
• Impaired spermatogenesis
• Subtilosin
• Impaired sperm motility
• Ketoconazole
• Reduced testosterone production
Oncologic agents**
• Chemotherapy4
• Gonadotoxicity to germ cells, Leydig cells, or Sertoli cells
• High risk: chlorambucil, cyclophosphamide, ifosfamide, busulfan,
melphalan, procarbazine, dacarbazine, and nitrogen-mustard, vincristine,
procarbazine, prednisone (MOPP)
• Intermediate risk: cisplatin, carboplatin, doxorubicin and bleomycine,
etoposide, cisplatin (BEP), and adriamycin, bleomycin, vinblastine,
dacarbazine (ABVD)
• Low risk: vincristine, methotrexate, dactinomycine, bleomycin,
mercaptopurine, vinblastine
• Imatinib
• Leydig cell dysfunction and subsequent impaired testosterone production
Partner history • Female age
• Female evaluation
• Previous pregnancy history
Allergies • Medication allergies
• Latex allergy if considering procedure
Family history • Infertility
• Cystic fibrosis
Goals of fertility • Number of children desired
• Time course couple wishes to conceive
• Restrictions (religious or personal beliefs) of assisted reproductive techniques
* Adapted from Samplaski and Nangia.5
** Adapted from Wallace et al.4
142  Male infertility

Table 11.2  Semen analysis parameters derived from Cooper and colleagues (2010).
WHO 2010 cutoff
Semen analysis parameter (5th percentile)6 Mean values7
Volume (mL) 1.5 3.9 ± 1.7
Concentration (million sperm/mL) 15 93.2 ± 65
Total sperm count (million) 39 350.7 ± 279.8
Total motility (%) 40 –
Progressive motility (%) 32 48 ± 16
Normal morphology (%) 4 37 ± 24
Vitality (%) 58 80 ± 12
Leukocyte count (million/mL) <1.0 –
Source: Splingart, C et al., Int J Androl. 2012;35(3):467–474; Cooper TG et al., Hum Reprod Update.
2010;16(3):​231–245.
Note: Lower 5th percentile values are presented from 1852 semen analyses of men able to conceive
a pregnancy within 12 months of unprotected regular intercourse. Mean values are presented
from a study evaluating 1114 men donating sperm for preservation with a history of fathering
at least 1 child.

fifty-three semen samples were assessed among men from with intrauterine insemination (IUI) have demonstrated
eight countries on three different continents. The lowest heterogeneous results. A meta-analysis from 2001 favored
5th percentile was used as the reference value (Table 11.2).7 pregnancy rates among men with >4% normal sperm
It is also important to recognize that values within the morphology12; however, more recent reports have not
normal range do not guarantee fertility and values below found a difference.13 Most importantly, these data suggest
the 5th percentile do not necessarily prevent men from that men with poor morphology <4% are still candidates
obtaining pregnancies as significant overlap exists among for IUI. The literature surrounding IVF and sperm mor-
fertile and infertile men with respect to semen analyses. phology is heterogeneous. A recent meta-analysis from
WHO 5th percentile cutoff values are important to use 2011 did not identify normal sperm morphology >4% to
to direct further investigation or management targeting be associated with higher pregnancy rates for either IVF
potential sources of impaired fertility; however, mean val- or IVF-ICSI.14
ues are also of benefit to know (Table 11.2).
Motility and progressive motility
Specific semen analysis abnormalities may be defined
as poor sperm concentration (oligozoospermia), reduced The WHO classifies motility as progressive motility
semen volume, poorly motile sperm (asthenozoospermia), (PM), non-progressive motility (NPM), and immotility
abnormal morphology (teratozoospermia), dead sperm (IM). PM sperm motility refers to active movement of
(necrospermia), or absence of sperm (azoospermia). sperm in a linear or large circular direction. NPM refers
to any other movements with lack of progression and
Morphology IM is the complete absence of movement. The impact of
Kruger and colleagues introduced strict criteria for grad- PM on natural pregnancy is heterogeneous. Some stud-
ing sperm morphology in 1986 for purposes of prognos- ies suggest PM is important in achieving pregnancies15–17
ticating sperm for success in in vitro fertilization.8 The while recent studies did not find statistically significant
criteria were described in additional detail by Menkveld difference once controlled for other semen parameters.18
and colleagues in 1990.9 The normal cutoffs for morphol- Sperm motility is likely required for natural pregnan-
ogy by the WHO has been decreasing to less stringent cies; however, the proportion of PM or NPM sperm that
values with more recent iterations, thus emphasizing are required for a natural pregnancy has not yet been
less importance on the number of sperm required to be defined.
normal for adequate fertilizing potential. However, a
ASTHENOSPERMIA
recent study evaluated the receiver operating character-
istics (ROCs) of semen analyses among fertile men and Causes
those requiring IUI. Sperm morphology demonstrated Asthenozoospermia is present among 25% of men present-
the greatest area under the curve and thus was most pre- ing for infertility evaluation. The differential diagnosis
dictive of fertility.10 Little data exist evaluating the effect includes leukocytospermia, antisperm antibodies, partial
of severely abnormal morphology on natural pregnancy ejaculatory duct obstruction, prolonged abstinence, vari-
rate. One study reports natural pregnancy rates of 29.2% coceles, or sperm ultrastructural defects (USDs).19 USDs
in men with severe 0% normal forms compared to a con- may be of the axoneme or periaxonemal structures. These
trol cohort of 55.6%.11 Thus, the fertilizing potential is typically occur among samples with motility less than
likely less; however, natural pregnancy is in fact possible. 7%.20 Kartagener syndrome is a genetic cause of primary
The impact of sperm morphology on pregnancy rates ciliary dyskinesia. It is important to note that immotile
Azoospermia 143

sperm on a semen analysis must be tested with a viability Concentration and sperm count
assay to determine if they are living but immotile sperm or Eight percent of men presenting with infertility will have a
if they are dead sperm (necrospermia). sperm concentration less than 15 million/mL and be classi-
Management fied as asthenospermia.22 The minimal number of sperm to
achieve a natural pregnancy is unknown. Several studies have
Treatment of the underlying cause is always indicated if evaluated the effect of sperm concentration and time to preg-
identified (i.e., varicocele repair, transurethral resection of nancy. Results suggest that an increased trend is observed
the ejaculatory duct [TURED] for partial ejaculatory duct among sperm concentrations increasing to 40–55 million/
obstruction (EDO), and regular intercourse or ejaculation mL, but the effect levels out at greater concentrations.23,24
for prolonged abstinence). For nonmodifiable factors, IVF/ Thus, low sperm concentrations below 40 million/mL may
ICSI may be used with live sperm confirmed by the presence have an impact on time to natural pregnancy among couples.
of at least twitching tails after treatment with pentoxifylline. With respect to IUI, sperm concentration and total motile
Semen volume and pH count (motility × total sperm count) have been demonstrated
to be the strongest predictors of success. Total motile count of
Semen collection for a semen analysis requires 2–5 days 5 million or greater portends to greater IUI pregnancy rates.25
of abstinence, collection without the use of spermicidal or IVF results are best with 3 million total motile count.26
toxic lubricants, and processing typically within 1 hour of
collection. Semen should be maintained at room or body OLIGOSPERMIA
temperature prior to analysis. No upper limit of normal Causes
exists; however, as mentioned earlier, the lower limit of vol-
ume is 1.5 mL. The differential diagnosis for men present- Oligozoospermia is directly related to spermatogenesis. The
ing with low semen volume includes incomplete collection, differential diagnosis includes environmental factors, medi-
retrograde ejaculation, testosterone deficiency, obstructed cal conditions, and medications. Environmental factors
ejaculatory ducts, absent seminal vesicles, or congenital include heat exposures, pesticides and chemical exposures,
bilateral absence of the vas deferens. The latter is diagnosed high caffeine intake, diets high in saturated fat, tobacco use,
by physical examination of the scrotum where no vas def- chronic intense endurance exercise, stress, marijuana use,
erens are palpable. Here, hypoplastic seminal vesicles are opioid abuse, and excessive alcohol use.27 Medical conditions
often associated. Obstructed ejaculatory ducts is suspected include varicoceles, systemic illnesses, fever, cryptorchidism,
with low volume, acidic semen with low fructose, and obesity,28 primary or secondary hypogonadism, radiation
absent or few poorly motile sperm. Digital rectal examina- therapy,4 previous testicular trauma or orchitis, malignancy
tion may also be positive for a midline cyst at the level of the and genetic causes such as Y-chromosome microdeletions,
prostate. Transrectal ultrasound (TRUS) is confirmatory and karyotype abnormalities such as Klinefelter syndrome
with dilated seminal vesicles >1.5 cm in diameter and often (progressive testicular failure), and Kallman syndrome (cen-
a midline cystic structure. Management of ejaculatory duct tral hypogonadism). Medication causes include but are not
obstruction includes TURED with the guidance of TRUS. exclusive to the following: chemotherapy, nitrofurantoin,
Low testosterone is diagnosed by serum testing. Men found aminoglycosides, diuretic and calcium channel blocker anti-
to have low testosterone cannot be placed on exogenous tes- hypertensives, 5 alpha reductase inhibitors, and exogenous
tosterone. The exogenous testosterone will provide negative testosterone or anabolic steroid use.5
feedback to the anterior pituitary and hypothalamus, con-
Management
sequently downregulate the hypothalamic-pituitary-testes
(HPT) axis and inhibit release of luteinizing hormone Oligozoospermic men with counts less than 10 million/
(LH) and follicle-stimulating hormone (FSH). This sup- mL warrant a hormonal evaluation with serum FSH and
presses spermatogenesis. Alternatively, many practitioners testosterone.29,30 Men with Oligozoospermia less than
use selective estrogen receptor modulators (SERMs) such 5 million/mL warrant both hormonal evaluation and
as clomiphene to upregulate the HPT axis and endogenous genetic testing including karyotype and Y-chromosome
testosterone production. In central pituitary or hypotha- microdeletion.31 Patients may be counseled to improve
lamic abnormalities, human chorionic gonadotropins lifestyle or environmental exposures, consider alternative
(hCG), recombinant FSH, or human menopausal gonado- medications, or treat medical illnesses. Beyond reversible
tropins (hMGs) may be used. causes of oligospermia, IUI may be considered in patients
with a total motile count greater than 5 million, IVF for
Postejaculate urinalysis patients with a total motile count of 3 million, and IVF-
In men with either low semen volume or a medical con- ICSI for severely oligozoospermic patients.
dition that predisposes one to retrograde ejaculation,
a postejaculate urinalysis should be obtained to search AZOOSPERMIA
for the presence of sperm on microscopic assessment. Azoospermia is defined as the absence of spermatozoa
Retrograde ejaculation occurs when the bladder neck is after two semen specimens have been examined with
incompetent functionally or anatomically and is the path extended search after centrifugation. The etiology may
of least resistance for semen to travel. be due to a blockage within the male reproductive tract,
144  Male infertility

Table 11.3  Investigations to differentiate the etiology of azoospermia.


Post-testicular
Ejaculatory
Investigation Pretesticular Testicular Obstructive ­dysfunction
Testicular size ↓ N/↓ N N
Semen volume N N N/↓ ↓
Post ejaculate N N N N/+ for sperm
urinalysis
Semen pH ≥7.2 N N N/↓ N
FSH ↓ ↑/N N N
T ↓ ↓/N N N
Prolactin N/↑ N N N
TRUS N N N/Abn N
Karyotype N/Abn N/Abn N N
y-Microdeletion N N/Abn N N
CFTR N N N/Abn N
Note: NOA can be classified into pretesticular or testicular abnormalities. Pretesticular, also termed hypogonadal hypogonad-
ism, is due to a central deficiency in the pituitary or hypothalamus. Testicular dysfunction, also termed hypergonadal
hypogonadism, is due to failure of the testicle to produce testosterone and/or spermatogenesis. Post-testicular azoosper-
mia may be due to obstruction in the epididymis, vas deferens, or ejaculatory duct. Ejaculatory dysfunction is best diag-
nosed by history and may comprise the absence of ejaculation failure of emission; here, semen volume is low and a
postejaculate urinalysis must be performed to assess for retrograde ejaculation.

termed obstructive azoospermia (OA), or may be due to If the pituitary is intact, pulsatile GnRH 25 to 600 ng/KG
impaired spermatogenesis, classically termed nonobstruc- may be delivered subcutaneously, intra­venously, or by pump
tive azoospermia (NOA). every 120 minutes. Otherwise, hCG 1000 to 2500 interna-
NOA may be due to central hypothalamic or pituitary tional units (IUs) are injected intramuscularly or subcuta-
abnormalities (pretesticular azoospermia) or due to testic- neously twice per week and act to stimulate the Leydig and
ular failure. History, physical exam, serum hormonal pro- Sertoli cells, with or without hMG 75 to 150 IU three times
file (testosterone, estrogen, FSH +/− LH and prolactin), and per week; alternatively, recombinant FSH (rFSH) may be
genetic testing including karyotype and Y-chromosome used in place of hMG but is significantly more costly.33
microdeletion are part of a standard evaluation to deter-
mine the etiology (Table 11.3). NOA patients often have Surgical sperm retrieval
small testes on physical exam and the epididymis is flat Among patients with NOA, sperm is surgically retrieved
and nondilated. Histology of testicles from NOA patients from the testicle. Techniques include testicular sperm aspira-
are classified into four patterns: hypospermatogenesis, late tion (TESA) (Figure 11.1), testicular sperm extraction (TESE),
maturation arrest, early maturation arrest, and Sertoli-
only syndrome.

Pretesticular
Causes
The differential diagnosis for pretesticular causes of
azoospermia include congenital abnormalities, such as
Kallman syndrome, characterized by midline defects,
anosmia, and tall stature, or acquired abnormalities, such
as pituitary tumor where they may present with headache,
galactorrhea, or hemianopsia, cranial radiotherapy, or
surgical extirpation, or exogenous androgens.32

Management
Treatment of these patients entail two components: medi- Figure 11.1  Testicular sperm aspiration (TESA) for sperm
cal correction of their hormonal abnormalities, and surgical retrieval in men with obstructive azoospermia. This procedure
retrieval of testicular sperm if no sperm returns to the ejacu- is often performed in an outpatient setting with local anesthetic
late following hormonal modification. GnRH or gonado- to the scrotal skin and a spermatic cord block is performed. A
tropins may be used depending on the a­ natomical location 23-gauge needle is passed into the testis 20–30 times while
of the defect. Several regimens exist for hormonal therapy. aspirating. The fluid is then placed in sperm transport media.
Azoospermia 145

Figure 11.2  Microdissection testicular sperm extraction (mTESE) for non-obstructive azoospermia. This procedure is typically
performed under a general anesthetic. The testicle is delivered from the scrotum and bivalved in a transverse equatorial fashion
with a 15-degree microknife. The seminiferous tubules are searched under 15-20x magnification for dilated opalescent tubes. These
tubes are teased out of the surround tissue, placed in a petri dish and minced with sterile scissors, and filtered through a 24-gauge
angiocatheter-tipped syringe to further break down the tissue. The tissue is placed on a microscope slide with a small amount of
sperm transport media and examined for the presence of sperm on a bright field microscope in the surgical theater.

and microdissection testicular sperm extraction (mTESE) Testicular azoospermia


(Figure 11.2). TESA is performed in an outpatient setting Causes
with local anesthesia in the form of a cord block and skin
wheal. A 23-gauge needle is passed into the testis 20–30 Testicular azoospermia is the result of the testicles not
times while aspirating. Contents are placed into a sperm responding to appropriate levels of FSH and LH. The dif-
buffer. TESA is successful in 52%–100% of cases.34 TESE ferential diagnosis for testicular failure can be organized
involves a general anesthetic, delivery of the testicle, inci- into congenital and acquired causes. Congenital causes
sion in the tunica albuginea, and excision of seminifer- include Klinefelter syndrome (i.e., XXXY karyotype),
ous tubules. This technique has largely been replaced by Y-chromosome microdeletion (AZFa, AZFb, AZFc), and
mTESE, which involves the use of an operating micro- cryptorchidism. Acquired causes include orchitis, vari-
scope and was described in 1999 by Peter Schlegel.35 Here, coceles, significant heat exposures, malignancies, chemo-
seminiferous tubules are visualized under a 10–20 power therapy, or radiation therapy. Successful sperm retrievals
operating microscope; dilated tubules are identified and have not been reported in the literature for complete AZFa
selectively removed as they are more likely to contain and AZFb Y-chromosome microdeletions, but are reported
active spermatogenesis and yield sperm. This technique in 70% of men with AZFc deletions.42
results in sperm retrieval rates of 45%–63%, and 70-fold
less tissue is excised compared to conventional TESE.36 Management
Surgical sperm retrieval rates vary by histologic subtype Medical optimization prior to surgical sperm retrieval
of NOA: 73%–91% in hypospermatogenesis, 27%–86% in NOA patients with testicular failure has been lightly
in late maturation arrest, 27%–40% in early maturation studied. One multicenter study determined that use of
arrest, and 31%–73% in Sertoli-cell-only.37–40 MicroTESE clomiphene citrate, hCG, and hMG to elevate presurgical
is considered the gold standard for sperm retrieval of testosterone levels to 600–800 ng/dL and FSH to 1.5 times
NOA patients. It is important to note that a semen analysis baseline improved sperm retrieval rate from 34% in the
should be performed on all men the morning of a sched- control group to 57% in the medical intervention group.43
uled mTESE because 5%–10% of NOA patients will have Aromatase inhibitors such as anastrozole have also dem-
sperm in their ejaculate despite prior documentation of onstrated benefit in NOA patients with low testosterone
azoospermia, thus obviating the need for the surgical and elevated estrogen levels.44 These medications have lim-
procedure.41 ited evidence and are presently used off-label. Otherwise,
146  Male infertility

surgical sperm retrieval is performed in this patient popu- rectal injury may also be used. Upon relieving the obstruc-
lation as mentioned above with mTESE. tion, cloudy milky fluid is often released.46
Posttesticular Epididymal obstruction and vasal obstruction
Posttesticular Azoospermia is due to a mechanical is diagnosed by history, physical exam demonstrating nor-
obstruction in the male reproductive tract or secondary to mal volume testicles, and normal serum FSH and testos-
ejaculatory or emissary dysfunction. terone. Among patients presenting with a vasectomy and
Obstructive causes previously normal spermatogenesis, then the presence
of normal volume testicles, serum FSH, and testosterone
OA due to mechanical obstruction may be secondary to is acceptable. However, if history identifies risk factors
vasectomy at the level of the vas deferens or at the level of for secondary impaired spermatogenesis or the patient
the epididymis secondary to epididymal blowout in the is presenting with primary epididymal or vasal obstruc-
setting of vasectomy, previous infection causing epididy- tion, then confirmation of spermatogenesis must be per-
mal stricturing, iatrogenic damage to the epididymis or vas formed. The gold standard method for confirmation of
deferens at the time of hydrocelectomy, orchidopexy, sper- normal spermatogenesis is testicular biopsy, demonstrat-
matocelectomy, or inguinal hernia repair. These patients ing active spermatogenesis with at least 20 mature sper-
will present with normal volume azoospermia. Congenital matids per seminiferous tubule cross section.48 However,
bilateral absence of the vas deferens (CBAVD) occurs due use of serum antisperm antibodies is an effective means of
to one of several possible mutations of the cystic fibro- identifying active spermatogenesis in an OA patient; here
sis transmembrane regulatory (CFTR) protein gene and
the area under the curve (AUC) for IgG against sperm tails
results in absence of the vas deferens and an incompletely
has been shown to be 0.92 with a sensitivity of 85% and
formed epididymis on physical exam; these patients will
specificity of 97%, while IgA demonstrated the greatest
often have hypoplastic seminal vesicles and demonstrate
specificity of 99%, a positive predictive value of 99% and
low volume on their semen analysis. They require CFTR
a positive likelihood ratio of 70.49 Patients presenting with
testing for both the patient and partner to provide appro-
epididymal or vasal obstruction can be managed by sperm
priate counseling for the risk of cystic fibrosis in their
retrieval, microsurgical reconstruction, or a combination.
potential offspring. Ejaculatory duct obstruction is another
etiology and often presents with a low-volume, acidic Sperm retrieval in OA
semen analysis with absence of fructose. In the presence
Sperm may be retrieved from the epididymis in OA.
of low semen volume and a pH <7.2, a digital rectal exam
This may be performed percutaneously or microsurgi-
and TRUS ultrasound is indicated to evaluate the patient
cally. Percutaneous epididymal sperm aspiration (PESA)
for cystic dilatation of the ejaculatory duct or enlarged
involves transcutaneous insertion of a 21–26-gauge nee-
seminal vesicles with a diameter greater than 1.5 cm. In the
dle into the epididymis while withdrawing (Figure 11.3).
case of equivocal TRUS findings but strong clinical suspi-
cion, a T2-weighted magnetic resonance imaging with an
endorectal coil may provide enhanced tissue resolution and
enable detection of subtle abnormalities.45 The differential
diagnosis for EDO includes congenital and acquired etiolo-
gies. Congenital causes include ejaculatory duct atresia or
stenosis and cysts of the prostate or utricle. Acquired causes
include seminal vesicle calculi, stricturing postcatheteriza-
tion or instrumentation, prostatitis, procedures for benign
prostatic hyperplasia such as transurethral resection of
prostate, green light laser, or prostate cancer radiotherapy,
or focal therapies such as cryotherapy, or high-intensity
focused ultrasonography (HIFU).46

Management of obstructive azoospermia


Ejaculatory duct obstruction may be managed via laser
incision of the ejaculatory ducts, retrograde balloon Figure 11.3 Percutaneous epididymal sperm aspiration
dilatation, retrograde seminal vesiculoscopy using a 9F (PESA) for sperm retrieval in men with obstructive azoosper-
scope or transurethral resection of the ejaculatory duct mia. This procedure is often performed in an outpatient set-
(TURED).46,47 TURED is both a popular and effective ting with local anesthetic to the scrotal skin and a spermatic
technique. Here, a resectoscope is introduced into the cord block is performed. The testicle is secured in the surgeon’s
urethra and tissue is resected on the inferiolateral veru- nondominant hand and a 21-26-gauge needle is inserted into
montana. Care is taken to avoid injury to the bladder neck the epididymis while aspirating for fluid. It is good practice to
and external striated sphincter. In addition, intraoperative examine the fluid under bright field microscopy to confirm the
use of TRUS to guide depth of resection and avoidance of presence of motile sperm.
Ejaculatory dysfunction  147

Reconstruction
Epididymal and vasal obstruction are often differenti-
ated intraoperatively depending on characteristics of the
vasal fluid following history of vasectomy (Table 11.4).
These findings direct decision making for the appropri-
ate reconstruction required. Important concepts for both
types of reconstruction include ensuring sperm parts or
copious clear fluid is present on the testicular end of the
anastomosis and an unobstructed abdominal vas. This can
be tested by injecting saline through the vas deferens with
a 24-gauge angiocatheter or placing a 2-0 monofilament
suture into the abdominal vas to ensure it can be passed
freely beyond the inguinal region.
Epididymal obstruction is corrected via a vasoepidid-
ymostomy (VE). Several techniques have been described;
however, the technique with the greatest patency rate is
Figure 11.4  Microsurgical epididymal aspiration (MESA) the longitudinal intussusception vasoepididymostomy
for men with obstructive azoospermia. The testis is deliv- (LIVE) technique (Figure 11.5). This technique has dem-
ered from the scrotum and visualized through an operat- onstrated a patency rate of 90% in experienced hands.54,55
ing microscope under 15-25x magnification. A 15-degree Vasal obstruction is reconstructed by performing a
microknife is used to incise a dilated tubule and the fluid vasovasostomy. Several techniques are described in the
is aspirated with a 24-gauge angiocatheter-tipped syringe literature and are largely differentiated by single and
or micropipette. The fluid is examined under bright field multilayer techniques. A recent meta-analysis reported
microscopy in the surgical theater to confirm presence of a mean patency rate of 89.4% for VVs and pregnancy
sperm. The fluid collected is then placed in sperm transport rate of 73.0%. They also determined that no statistically
media in Eppendorf tubules. significant differences existed in the patency of single vs
multilayer techniques.56 However, the highest reported
patency rate in the literature of 99.5% involves a multi-
The fluid is then assessed on a microscope for presence
layer technique involving six microdots to separate plan-
of motile sperm that can be used for IVF-ICSI. PESA
ning from suture placement of corresponding 10-0 nylon
can be performed with local anesthetic in the office set-
sutures to intricately reapproximate the mucosal layer,
ting. Sperm is successfully retrieved in 61-96% of cases
followed by six deep muscularis sutures using 9-0 nylon,
using PESA.50,51 Microsurgical epididymal sperm aspira-
six superficial 9-0  nylon sutures, and a final adventitial
tion (MESA) is the gold standard method for epididymal
layer (Figure 11.6).57
sperm retrieval (Figure 11.4).
MESA is performed under a general or regional anes-
thetic. The testicle is delivered and the epididymis is EJACULATORY DYSFUNCTION
examined for dilated tubules. A 15-degree microknife Causes of Ejaculatory Dysfunction Ejaculatory dysfunc-
is then used to incise an “epididymal tubule and the fluid tion may be due to failure of emission, anejaculation (AE),
is aspirated by capillary action into an ordinary 5-µl or retrograde ejaculation (RE).
capacity laboratory pipette and examined under a bench
microscope to identify motile sperm. MESA is successful Retrograde ejaculation
in retrieving sperm in 96%–100% of patients with OA,52,53 Retrograde ejaculation occurs when the bladder neck
typically yielding 15–95 million sperm, which is adequate is incompetent. RE may be implicated in up to 18% of
for cryopreservation in 98%–100% of patients.34 men presenting with azoospermia.21 RE may be due

Table 11.4  Intraoperative vasal fluid is sampled through a hemivasotomy at the time of vasal reconstruction for OA.
Vasal fluid Microscopic findings Surgical procedure indicated
Copious clear fluid No sperm Vasovasostomy
Cloudy fluid Intact sperm Vasovasostomy
Creamy yellow fluid Sperm heads Vasovasostomy
Thick white toothpaste-like fluid No sperm Vasoepididymostomy
Dry vas with no granuloma No sperm Vasoepididymostomy
Dry vas with granuloma at vasectomy site Barbotage fluid reveals sperm Vasovasostomy
Note: The decision to proceed with vasovasostomy compared to vasoepididymostomy is made by examining the appearance of the
vasal fluid and the microscopic findings.
148  Male infertility

(a) (b)

(d)
(c)

(e) (f )

Figure 11.5  Longitudinal intussusception vasoepididymostomy (LIVE) technique is performed in men with obstructive azo-
ospermia. Vasoepididymostomies are performed under a general anesthetic. Once the testicle is delivered and the transition
from dilated to collapsed tubules has been identified, a region of dilated tubules just prior to the transition is selected. The tunica
is incised exposing the dilated tubules (a). A double-armed 10-0 nylon suture is placed longitudinally on one side of the epi-
didymal tubule (b). The needle is only placed halfway through so that the curve of the needle remains in the tubule. The needle
is not driven all the way to prevent decompression of the fluid around the suture, which is smaller diameter than the needle. A
second suture is then placed parallel to the initial suture (b). Four microdots are drawn on the muscularis of the vas deferens. A
microknife is used to incise the tubule longitudinally between the two needles (c). The fluid is sampled for sperm or sperm parts
under a microscope in the surgical theater. If present, the anastomosis is completed; if no sperm or sperm parts are present, then
the surgeon must move proximally. The remaining ends of the double-armed sutures are then placed inside-out through the
microdot and tied (d and e). Then, 9-0 nylon sutures are used to reapproximate the muscularis and adventitia to the tunica of the
epididymis (f).

to congenital abnormalities such as posterior urethral Causes


valves, utricular cysts, or extrophy. RE may also be due Medical conditions known to be associated with retro-
to an anatomic defect following prostatic resection, inci- grade ejaculation include alpha-blocker medications, pre-
sion, or ablation or due to a functional deficit. Functional vious transurethral resection of the prostate, diabetes, and
deficiencies may be due to use of alpha blockers, psycho- spinal cord lesions.
tropic medications, abdominal or pelvic surgeries such as
retroperitoneal lymph node dissection, aortoiliac vascu- Management
lar surgery and abdominal perineal resection, or neuro- A trial of a sympathomimetic such as oral pseudoephed-
logical diseases such as diabetic autonomic neuropathy, rine or a tricyclic antidepressant imipramine may be trialed
multiple sclerosis, myelodysplasia, multisystem atrophy, prior to ejaculation for men with a functionally incompe-
cerebrovascular accident, spinal cord injuries, and lum- tent bladder neck. Other agents described in the literature
bar sympathectomy. include brompheniramine, midodrine, and ephedrine.
Ejaculatory dysfunction  149

(a) (b)

(c) (d)

(e)

Figure 11.6  Vasovasostomy microdot multilayer closure performed for men with obstructive azoospermia where the level of
obstruction is in the vas deferens. This procedure is performed under a general anesthetic and the testicle is delivered through
a vertical scrotal incision on the respective side. The site of obstruction is identified and the vas deferens is transected (a).
Patency of the vas deferens is checked on the abdominal side and fluid from the testicular side is evaluated under a microscope
for sperm or sperm parts. Reconstruction is then performed as depicted above. Six microdots are drawn onto the muscularis
layer of the cut surface (b). Three 10-0 double-armed nylon sutures are then placed in an inside-out fashion and tied (c). Three
9-0 sutures are placed in the muscularis between the 10-0 sutures (d). An additional layer of three 9-0 sutures are placed super-
ficially through the muscularis between the previous 9-0s. The vas deferens is then flipped and the second half is completed in
the same sequence. Finally, a fourth layer of 9-0s are placed to reapproximate the adventitia to complete a patent and water-
tight anastomosis (e).

Pseudoephedrine (Sudafed) 60-mg tablets may be taken multiple sclerosis.58 Pelvic surgery and retroperitoneal
four times per day in the 2–3 days leading up to ejacula- lymph node dissection may be associated with ejacula-
tion.21 If pseudoephedrine is not successful or an anatomi- tory dysfunction or failure of emission where sperm and
cally abnormal bladder neck is present (i.e., transurethral semen are transported to the posterior urethra prior to
resection of the prostate), then the postejaculate urine ejaculation.58
specimen may be collected, centrifuged, and the pellet of
sperm may be resuspended and used for assisted reproduc- Management of anejaculation and failure
tive techniques, IUI, or if of poor quality, IVF/ICSI. of emission
Management of RE is described above in the manage-
Anejaculation ment of low semen volume. Modifiable factors such as
Anejaculation occurs when the patient fails to ejaculate. psychological stressors, hyperstimulation, use of selec-
This may be due to psychological stress, hyperstimula- tive serotonin reuptake inhibitors, and low testoster-
tion of sexual content, chronic medical conditions such one should be identified and attempts made to address
as chronic kidney disease requiring dialysis, selec- these factors. Among patients still unable to ejaculate
tive serotonin reuptake inhibitor use, low testosterone, through intercourse, outercourse, or masturbation, then
decreased penile sensation, and neurological disorders vibrostimulation (VS) or electroejaculation (EEJ) should
such as spinal cord injury (SCI), and some men with be considered.
150  Male infertility

Vibrostimulation meta-analysis determined that elevated DNA fragmenta-


Is less invasive than EEJ and should be considered first. tion was associated with lower clinical pregnancy rates in
Here, a vibrator device is applied to the frenular surface of both IVF and ICSI techniques. However, the positive and
the glans until ejaculation ensues. This technique may be negative predictive abilities are limited at 36% and 40%,
used for men with psychogenic anejaculation or second- respectively, for ICSI and are worse for IVF.67 Another
ary to SCIs above T12. For VS to work successfully, the meta-analysis reports improved live birth rates for IVF in
patient’s ejaculatory reflex arc must be intact, including men with low sperm DNA fragmentation.68 Reports have
the afferent sensory nerves, S2-4 ejaculatory center, and also determined increased miscarriage rates among men
the efferent motor nerves. This technique may be per- with elevated sperm DNA fragmentation undergoing ICSI
formed at home or in the office. The treating physician but not IVF.69
should always be aware of autonomic dysreflexia among Several assays presently exist: sperm chromatin disper-
patients with a SCI above T6. Sperm may be used for intra- sion (SCD) or halo test,70 sperm chromatin structure assay
vaginal insemination, IUI, IVF, or IVF-ICSI.59,60 (SCSA),71 comet assay,72 and terminal deoxynucleotidyl
transferase dUTP nick-end labeling assay (TUNEL).73
Electroejaculation SCD, SCSA, and TUNEL measure the proportion of sperm
Involves using a rectal probe that serves as an electrode to with DNA strand breaks, while comet assay measures
deliver electrical stimulation to the prostate and seminal the degree of DNA fragmentation per sperm. The comet
vesicles. The patient is positioned to right lateral decubitus. assay has the ability to detect single- and double strand-
The patient is catheterized prior to the procedure using DNA breaks as well as protamine and histone associated
mineral oil as lubrication and the urine is drained. Twenty breaks in chromatin, while TUNEL can detect single- and
to 30 mL of sperm transport media (e.g., human tubal fluid double-strand DNA breaks, and both SCD and SCSA can
[HTF] buffered with hydroxyethyl-piperazineethane-sul- only detect single-strand DNA breaks.
fonic acid buffer and plasmanate) is then instilled into the Management
bladder. Anoscopy is performed to ensure that the rectal
mucosa is healthy. The Seager Electroejaculator© probe Presently, DNA fragmentation assays are not considered
is then inserted into the rectum and either a peaked sine routine evaluation; however, as more data from clini-
wave or 1-second block paradigm is used with five stimu- cal studies emerge, the role of DNA fragmentation may
lations, repeated up to six times. The probe temperature also evolve. However, many practitioners will assess
must be kept below 38 degrees Celsius and below 30-volts DNA fragmentation in patients with recurrent preg-
maximum. An assistant holds a sterile specimen cup in nancy loss via natural conception or ARTs. Strategies to
front of the meatus to collect the sample. Anoscopy is improve DNA fragmentation are presently limited in
again performed at the end of the procedure. EEJ may be number. Varicocelectomy, use of antioxidants, and use
performed in a surgical theater for sensate patients and of testicular sperm have been strategies reported to date.
may be performed in either the office or surgical theater Varicocelectomies have demonstrated improvements in
for patients not sensate below the waste. However, the DNA fragmentation74; however, a meta-analysis noted lim-
physician must again monitor for autonomic dysreflexia ited improvement of DNA fragmentation.75 Antioxidant
(AD) in patients with SCI above T6 and have the ability formulations vary widely in content. However, Greco
to treat respected changes. Many physicians recommend and colleagues used daily vitamin C and vitamin E in
pretreating >T6 SCI patients with nifedipine; a discus- men with elevated DNA fragmentation and resulted in
sion with the anesthetist preoperatively is recommended. 76% of men reducing their TUNEL scores to the normal
If AD does ensue, the stimulation should be withdrawn range for their lab, and also resulted in significantly more
immediately.21 clinical pregnancies using ICSI compared to those not
using antioxidant formulations.76 A meta-analysis assess-
DNA FRAGMENTATION ing the impact of antioxidants on fertility determined a
Sperm DNA integrity is required for subsequent embryo positive benefit on live birth rates, and the two studies
development. DNA integrity is thought to be maintained assessing DNA fragmentation demonstrated improved
and protected by chromatin repackaging in the nucleus rates.77 Studies have demonstrated that among men with
by disulfide cross-links among protamines. However, the high DNA fragmentation, use of testicular sperm often
DNA can acquire strand breaks and damage. This can has lower DNA fragmentation; two studies have reported
occur intrinsically via dysfunctional DNA repair, prot- improved clinical pregnancies, live births, and fewer mis-
amine deficiencies,61 and incomplete apoptosis.62 Extrinsic carriages in couples with high ejaculated DNA fragmen-
factors are also thought to contribute to DNA fragmen- tation and subsequent use of testicular sperm with lower
tation, such as prolonged abstinence, heat exposures, DNA fragmentation.78,79
radiation, gonadotoxins, and varicoceles.63–65 Increased
DNA fragmentation has been associated with spontane- VARICOCELE
ous recurrent miscarriage, impaired fertilization, early Varicoceles are identified in 12% of men with normal
embryo development, implantation, and pregnancy.66 A semen parameters, 40% of men with primary infertility,
References 151

and 80% of men with secondary infertility. However,


among all men with varicoceles, approximately 20% will Genital femoral nerve
have difficulties with fertility.80 Varicoceles are most Vasal veins (ligated)
Vas deferens
often associated with oligoasthenoteratospermia (OAT);
however, repair has been shown to improve each of these
semen parameters. Eighty-five to 90 percent of varico-
celes arise on the left side, thought to be secondary to the
90-degree entry of the left spermatic vein into the renal Lymphatic
Testicular artery
vein causing higher hydrostatic pressure, compared to the
oblique approach on the right side.81,82 Additional theories
Vessel loop
for development of varicoceles include absent or dysfunc-
tional venous valves and compression of the spermatic vein
between the superior mesenteric artery and renal vein.83,84
The negative effect of varicoceles on testicular function
is thought to occur via multiple mechanisms: increased
Figure 11.7  Subinguinal varicocelectomy. A 2.5-cm oblique
scrotal temperature,85 hypoxia,86 increased venous pres-
incision in the lines of Langerhans is made inferior to the exter-
sure,87 oxidative stress,88 reflux of toxic renal and adrenal
nal inguinal ring and the spermatic cord is delivered. The vas
metabolites,89 and hormone imbalances.90
deferens is identified and excluded. The external and internal
spermatic fascia is opened and the internal spermatic artery is
Management
identified with Doppler ultrasound and marked with a vessel
Varicoceles are diagnosed by physical exam. In stand- loop. The veins are then identified, isolated, and ligated with
ing position, grade 1 varicoceles can be palpated with 4-0 silk ties around the artery and titanium clips peripherally.
Valsalva, grade 2 can be palpated even in the absence of Deferential and cremasteric arteries are also identified in most
Valsalva, and grade 3 can be visualized through the thin cases (not shown here) and preserved. Lymphatics are also iden-
scrotal skin even in the absence of Valsalva. A varicocele tified by their scalloped and clear appearance and are preserved.
that is palpable should become not palpable upon chang-
ing positions from standing to supine. In the event of an
isolated right-sided varicocele, abdominal imaging, typi- ACKNOWLEDGMENT
cally in the form of an ultrasound, is warranted to assess The authors would like to thank Vanessa Dudley for the
for an intra-abdominal mass causing venous compression. figure illustrations.
Furthermore, if physical exam is unclear, a scrotal ultra-
sound should be considered. Here, spermatic cord vein REFERENCES
diameters should be measured both supine and standing, 1. Stephen EH, Chandra A. Declining estimates of
as well as assessment for reversal of blood flow. Vein diam- infertility in the United States: 1982–2002. Fertil
eters larger than 3 mm and reversal of flow are considered Steril. 2006;86(3):516–523.
clinical varicoceles; veins smaller than 3 mm with reversal 2. Thonneau P, Marchand S, Tallec A et al. Incidence
of flow are termed subclinical varicoceles. and main causes of infertility in a resident popula-
Varicoceles may be repaired through various tech- tion (1,850,000) of three French regions (1988–1989).
niques: percutaneous embolization, laparoscopic, ingui- Hum Reprod. 1991;6(6):811–816.
nal, and microscopic subinguinal (Figure 11.7). The latter 3. Sunderam S, Kissin DM, Crawford SB et al. Assisted
has been the gold standard in the literature with the low- Reproductive Technology Surveillance—United
est recurrence rate of 0.8% to 2.1% and the lowest inci- States, 2014. MMWR Surveill Summ. 2017;66(6):1–24.
dence of postoperative hydrocele formation among the 4. Wallace WH, Anderson RA, Irvine DS. Fertility
surgical techniques.91,92 Postoperative hydroceles occur preservation for young patients with cancer: Who
when lymphatics are indiscriminately ligated during is at risk and what can be offered? Lancet Oncol.
varicocele repair (VR); however, these can be visually 2005;6(4):209–218.
identified and avoided in the microscopic subingui- 5. Samplaski MK, Nangia AK. Adverse effects of com-
nal technique. VR has been demonstrated to improve mon medications on male fertility. Nat Rev Urol.
sperm motility, concentration, and morphology, increase 2015;12(7):401–413.
spontaneous pregnancy rates,91 and reduce miscarriage 6. Cooper TG, Noonan E, von Eckardstein S et al.
rates.93 In a meta-analysis, VR has also been shown to World Health Organization reference values for
increase live birth rates among oligospermic men; preg- human semen characteristics. Hum Reprod Update.
nancy rates were higher in men with azoospermia as was 2010;16(3):231–245.
surgical sperm retrieval rates among azoospermic men.94 7. Splingart C, Frapsauce C, Veau S, Barthelemy C,
The effects of a VR are typically not observed prior to 3–6 Royere D, Guerif F. Semen variation in a population
months as one round of spermatogenesis typically takes of fertile donors: Evaluation in a French centre over a
at least 64 to 72 days.95 34-year period. Int J Androl. 2012;35(3):467–474.
152  Male infertility

8. Kruger TF, Menkveld R, Stander FS et al. Sperm 23. Slama R, Eustache F, Ducot B et al. Time to preg-
morphologic features as a prognostic factor in in nancy and semen parameters: A cross-sectional study
vitro fertilization. Fertil Steril. 1986;46(6):1118–1123. among fertile couples from four European cities.
9. Menkveld R, Stander FS, Kotze TJ, Kruger TF, van Hum Reprod. 2002;17(2):503–515.
Zyl JA. The evaluation of morphological character- 24. Bonde JP, Ernst E, Jensen TK et al. Relation between
istics of human spermatozoa according to stricter semen quality and fertility: A population-based study
criteria. Hum Reprod. 1990;5(5):586–592. of 430 first-pregnancy planners. Lancet. 1998;352​
10. Aboutorabi R, Zamani S, Zarrin Y, Mostafavi FS. A (9135):​1172–1177.
survey on main semen parameters in natural preg- 25. Merviel P, Heraud MH, Grenier N, Lourdel E,
nancy and intrauterine insemination: Are there any Sanguinet P, Copin H. Predictive factors for preg-
significant differences? Am J Mens Health. 2016. nancy after intrauterine insemination (IUI): An
11. Kovac JR, Smith RP, Cajipe M, Lamb DJ, Lipshultz LI. analysis of 1038 cycles and a review of the literature.
Men with a complete absence of normal sperm mor- Fertil Steril. 2010;93(1):79–88.
phology exhibit high rates of success without assisted 26. Michelmann HW. Minimal criteria of sperm qual-
reproduction. Asian J Androl. 2017;19(1):39–42. ity for insemination and IVF therapy. Int J Androl.
12. Van Waart J, Kruger TF, Lombard CJ, Ombelet 1995;18 Suppl 2:81–87.
W. Predictive value of normal sperm morphology 27. Barazani Y, Katz BF, Nagler HM, Stember DS. Life­
in intrauterine insemination (IUI): A structured style, environment, and male reproductive health.
literature review. Hum Reprod Update. 2001;7(5):​ Urol Clin North Am. 2014;41(1):55–66.
495–500. 28. Sermondade N, Faure C, Fezeu L et al. BMI in rela-
13. Shabtaie SA, Gerkowicz SA, Kohn TP, Ramasamy tion to sperm count: An updated systematic review
R. Role of abnormal sperm morphology in pre- and collaborative meta-analysis. Hum Reprod Update.
dicting pregnancy outcomes. Curr Urol Rep. 2013;​19(3):221–231.
2016;17(9):67. 29. Sigman M, Jarow JP. Endocrine evaluation of infer-
14. Hotaling JM, Smith JF, Rosen M, Muller CH, Walsh tile men. Urology. 1997;50(5):659–664.
TJ. The relationship between isolated teratozoosper- 30. Jarow JP. Endocrine causes of male infertility. Urol
mia and clinical pregnancy after in vitro fertilization Clin North Am. 2003;30(1):83–90.
with or without intracytoplasmic sperm injection: 31. Foresta C, Garolla A, Bartoloni L, Bettella A, Ferlin A.
A systematic review and meta-analysis. Fertil Steril. Genetic abnormalities among severely oligospermic
2011;95(3):1141–1145. men who are candidates for intracytoplasmic sperm
15. Jouannet P, Ducot B, Feneux D, Spira A. Male fac- injection. J Clin Endocrinol Metab. 2005;90(1):152–156.
tors and the likelihood of pregnancy in infertile cou- 32. Esteves SC. Clinical management of infertile men
ples. I. Study of sperm characteristics. Int J Androl. with nonobstructive azoospermia. Asian J Androl.
1988;11(5):379–394. 2015;17(3):459–470.
16. Larsen L, Scheike T, Jensen TK et al. Computer- 33. Silveira LF, MacColl GS, Bouloux PM. Hypogon­
assisted semen analysis parameters as predictors for adotropic hypogonadism. Semin Reprod Med. 2002;​
fertility of men from the general population. The 20(4):327–338.
Danish First Pregnancy Planner Study Team. Hum 34. Bernie AM, Ramasamy R, Stember DS, Stahl PJ.
Reprod. 2000;15(7):1562–1567. Microsurgical epididymal sperm aspiration: Indi­
17. Zinaman MJ, Brown CC, Selevan SG, Clegg ED. cations, techniques and outcomes. Asian J Androl.
Semen quality and human fertility: A prospective 2013;15(1):40–43.
study with healthy couples. J Androl. 2000;21(1):​ 35. Schlegel PN. Testicular sperm extraction: Micro­
145–153. dissection improves sperm yield with minimal tissue
18. Buck Louis GM, Sundaram R, Schisterman EF et al. excision. Hum Reprod. 1999;14(1):131–135.
Semen quality and time to pregnancy: The Longi­ 36. Shin DH, Turek PJ. Sperm retrieval techniques. Nat
tudinal Investigation of Fertility and the Envi­ron­ Rev Urol. 2013;10(12):723–730.
ment Study. Fertil Steril. 2014;101(2):453–462. 37. Deruyver Y, Vanderschueren D, Van der Aa F.
19. Smith RP, Coward RM, Lipshultz LI. The office visit. Outcome of microdissection TESE compared with
Urol Clin North Am. 2014;41(1):19–37. conventional TESE in non-obstructive azoospermia:
20. Francavilla S, Pelliccione F, Cordeschi G et al. A systematic review. Andrology. 2014;2(1):20–24.
Utrastructural analysis of asthenozoospermic ejacu- 38. Bernie AM, Shah K, Halpern JA et al. Outcomes of
lates in the era of assisted procreation. Fertil Steril. microdissection testicular sperm extraction in men
2006;85(4):940–946. with nonobstructive azoospermia due to maturation
21. Revenig L, Leung A, Hsiao W. Ejaculatory physiology arrest. Fertil Steril. 2015;104(3):569–573 e561.
and pathophysiology: Assessment and treatment in 39. Colpi GM, Colpi EM, Piediferro G et al. Microsurgical
male infertility. Transl Androl Urol. 2014;3(1):41–49. TESE versus conventional TESE for ICSI in non-
22. Lipshultz L, JJ K. Current Urologic Therapy. obstructive azoospermia: A randomized controlled
Philadelphia: Saunders; 1980. study. Reprod Biomed Online. 2009;18(3):315–319.
References 153

40. Caroppo E, Colpi EM, Gazzano G et al. Testicular his- 54. Chan PT, Li PS, Goldstein M. Microsurgical vaso-
tology may predict the successful sperm retrieval in epididymostomy: A prospective randomized study
patients with non-obstructive azoospermia undergo- of 3 intussusception techniques in rats. J Urol.
ing conventional TESE: A diagnostic accuracy study. 2003;169(5):1924–1929.
J Assist Reprod Genet. 2017;34(1):149–154. 55. Chan PT, Libman J. Feasibility of microsurgical
41. Schlegel PN, Liotta D, Hariprashad J, Veeck LL. reconstruction of the male reproductive tract after
Fresh testicular sperm from men with nonobstruc- percutaneous epididymal sperm aspiration (PESA).
tive azoospermia works best for ICSI. Urology. Can J Urol. 2003;10(6):2070–2073.
2004;64(6):1069–1071. 56. Herrel LA, Goodman M, Goldstein M, Hsiao W.
42. Hopps CV, Mielnik A, Goldstein M, Palermo GD, Outcomes of microsurgical vasovasostomy for
Rosenwaks Z, Schlegel PN. Detection of sperm vasectomy reversal: A meta-analysis and systematic
in men with Y chromosome microdeletions of review. Urology. 2015;85(4):819–825.
the AZFa, AZFb and AZFc regions. Hum Reprod. 57. Goldstein M, Li PS, Matthews GJ. Microsurgical
2003;18(8):1660–1665. vasovasostomy: The microdot technique of precision
43. Hussein A, Ozgok Y, Ross L, Rao P, Niederberger C. suture placement. J Urol. 1998;159(1):188–190.
Optimization of spermatogenesis-regulating hor- 58. Chen J. The pathophysiology of delayed ejaculation.
mones in patients with non-obstructive azoosper- Transl Androl Urol. 2016;5(4):549–562.
mia and its impact on sperm retrieval: A multicentre 59. Sonksen J, Sommer P, Biering-Sorensen F et al.
study. BJU Int. 2013;111(3 Pt B):E110–114. Pregnancy after assisted ejaculation procedures in
44. Cavallini G, Biagiotti G, Bolzon E. Multivariate anal- men with spinal cord injury. Arch Phys Med Rehabil.
ysis to predict letrozole efficacy in improving sperm 1997;78(10):1059–1061.
count of non-obstructive azoospermic and crypto- 60. Kathiresan AS, Ibrahim E, Aballa TC, Attia GR,
zoospermic patients: A pilot study. Asian J Androl. Lynne CM, Brackett NL. Pregnancy outcomes by
2013;15(6):806–811. intravaginal and intrauterine insemination in 82
45. Smith JF, Walsh TJ, Turek PJ. Ejaculatory duct couples with male factor infertility due to spinal cord
obstruction. Urol Clin North Am. 2008;35(2):221– injuries. Fertil Steril. 2011;96(2):328–331.
227, viii. 61. Aitken RJ, Bronson R, Smith TB, De Iuliis GN. The source
46. Modgil V, Rai S, Ralph DJ, Muneer A. An update on and significance of DNA damage in human spermato-
the diagnosis and management of ejaculatory duct zoa; A commentary on diagnostic strategies and straw
obstruction. Nat Rev Urol. 2016;13(1):13–20. man fallacies. Mol Hum Reprod. 2013;19(8):475–485.
47. Xu B, Niu X, Wang Z et al. Novel methods for the 62. Aitken RJ, De Iuliis GN. On the possible origins of
diagnosis and treatment of ejaculatory duct obstruc- DNA damage in human spermatozoa. Mol Hum
tion. BJU Int. 2011;108(2):263–266. Reprod. 2010;16(1):3–13.
48. Chan PT. The evolution and refinement of vaso- 63. Practice Committee of the American Society for
epididymostomy techniques. Asian J Androl. 2013;​ Reproductive M. Diagnostic evaluation of the
15(1):49–55. infertile male: A committee opinion. Fertil Steril.
49. Lee R, Goldstein M, Ullery BW et al. Value of serum 2015;103(3):e18–25.
antisperm antibodies in diagnosing obstructive azo- 64. Greco E, Scarselli F, Iacobelli M et al. Efficient treat-
ospermia. J Urol. 2009;181(1):264–269. ment of infertility due to sperm DNA damage by
50. Meniru GI, Gorgy A, Batha S, Clarke RJ, Podsiadly ICSI with testicular spermatozoa. Hum Reprod.
BT, Craft IL. Studies of percutaneous epididymal 2005;20(1):226–230.
sperm aspiration (PESA) and intracytoplasmic sperm 65. Cho CL, Esteves SC, Agarwal A. Novel insights into
injection. Hum Reprod Update. 1998;4(1):57–71. the pathophysiology of varicocele and its association
51. Lin YM, Hsu CC, Kuo TC, Lin JS, Wang ST, Huang with reactive oxygen species and sperm DNA frag-
KE. Percutaneous epididymal sperm aspiration mentation. Asian J Androl. 2016;18(2):186–193.
versus microsurgical epididymal sperm aspira- 66. Lewis SE, John Aitken R, Conner SJ et al. The impact
tion for irreparable obstructive azoospermia— of sperm DNA damage in assisted conception and
Experience with 100 cases. J Formos Med Assoc. beyond: Recent advances in diagnosis and treatment.
2000;99(6):459–465. Reprod Biomed Online. 2013;27(4):325–337.
52. Schlegel PN, Palermo GD, Alikani M et al. 67. Simon L, Zini A, Dyachenko A, Ciampi A, Carrell
Micropuncture retrieval of epididymal sperm with in DT. A systematic review and meta-analysis to deter-
vitro fertilization: Importance of in vitro microma- mine the effect of sperm DNA damage on in vitro
nipulation techniques. Urology. 1995;46(2):238–241. fertilization and intracytoplasmic sperm injection
53. Nudell DM, Conaghan J, Pedersen RA, Givens outcome. Asian J Androl. 2017;19(1):80–90.
CR, Schriock ED, Turek PJ. The mini-micro-­ 68. Osman A, Alsomait H, Seshadri S, El-Toukhy T, Khalaf
epididymal sperm aspiration for sperm retrieval: Y. The effect of sperm DNA fragmentation on live birth
A study of urological outcomes. Hum Reprod. rate after IVF or ICSI: A systematic review and meta-
1998;13(5):1260–1265. analysis. Reprod Biomed Online. 2015;30(2):120–127.
154  Male infertility

69. Zhao J, Zhang Q, Wang Y, Li Y. Whether sperm 83. Shafik A, Bedeir GA. Venous tension patterns in cord
deoxyribonucleic acid fragmentation has an veins. I. In normal and varicocele individuals. J Urol.
effect on pregnancy and miscarriage after in vitro 1980;123(3):383–385.
fertilization/intracytoplasmic sperm injection: A
­ 84. Ahlberg NE, Bartley O, Chidekel N. Right and left
systematic review and meta-analysis. Fertil Steril. gonadal veins. An anatomical and statistical study.
2014;102(4):998–1005 e1008. Acta Radiol Diagn (Stockh). 1966;4(6):593–601.
70. Fernandez JL, Muriel L, Goyanes V et al. Simple 85. Garolla A, Torino M, Miola P et al. Twenty-four-
determination of human sperm DNA fragmentation hour monitoring of scrotal temperature in obese
with an improved sperm chromatin dispersion test. men and men with a varicocele as a mirror of
Fertil Steril. 2005;84(4):833–842. spermatogenic function. Hum Reprod. 2015;30(5):​
71. Evenson DP. Sperm chromatin structure assay 1006–1013.
(SCSA(R)). Methods Mol Biol. 2013;927:147–164. 86. Hu W, Zhou PH, Zhang XB, Xu CG, Wang W. Roles
72. Olive PL, Banath JP. The comet assay: A method of adrenomedullin and hypoxia-inducible fac-
to measure DNA damage in individual cells. Nat tor 1 alpha in patients with varicocele. Andrologia.
Protoc. 2006;1(1):23–29. 2015;47(8):951–957.
73. Sharma R, Masaki J, Agarwal A. Sperm DNA frag- 87. Eisenberg ML, Lipshultz LI. Varicocele-induced
mentation analysis using the TUNEL assay. Methods infertility: Newer insights into its pathophysiology.
Mol Biol. 2013;927:121–136. Indian J Urol. 2011;27(1):58–64.
74. Zini A, Dohle G. Are varicoceles associated with 88. Agarwal A, Makker K, Sharma R. Clinical relevance
increased deoxyribonucleic acid fragmentation? of oxidative stress in male factor infertility: An
Fertil Steril. 2011;96(6):1283–1287. update. Am J Reprod Immunol. 2008;59(1):2–11.
75. Wang YJ, Zhang RQ, Lin YJ, Zhang RG, Zhang WL. 89. Fujisawa M, Yoshida S, Kojima K, Kamidono S.
Relationship between varicocele and sperm DNA Biochemical changes in testicular varicocele. Arch
damage and the effect of varicocele repair: A meta- Androl. 1989;22(2):149–159.
analysis. Reprod Biomed Online. 2012;25(3):307–314. 90. Rajfer J, Turner TT, Rivera F, Howards SS, Sikka SC.
76. Greco E, Romano S, Iacobelli M et al. ICSI in cases of Inhibition of testicular testosterone biosynthesis fol-
sperm DNA damage: Beneficial effect of oral antioxi- lowing experimental varicocele in rats. Biol Reprod.
dant treatment. Hum Reprod. 2005;20(9):2590–2594. 1987;36(4):933–937.
77. Showell MG, Mackenzie-Proctor R, Brown J, 91. Cayan S, Shavakhabov S, Kadioglu A. Treatment
Yazdani A, Stankiewicz MT, Hart RJ. Antioxidants of palpable varicocele in infertile men: A meta-
for male subfertility. Cochrane Database Syst Rev. analysis to define the best technique. J Androl.
2014(12):CD007411. 2009;30(1):33–40.
78. Esteves SC, Sanchez-Martin F, Sanchez-Martin P, 92. Diegidio P, Jhaveri JK, Ghannam S, Pinkhasov R,
Schneider DT, Gosalvez J. Comparison of repro- Shabsigh R, Fisch H. Review of current varicoce-
ductive outcome in oligozoospermic men with high lectomy techniques and their outcomes. BJU Int.
sperm DNA fragmentation undergoing intracyto- 2011;108(7):1157–1172.
plasmic sperm injection with ejaculated and testicu- 93. Mansour Ghanaie M, Asgari SA, Dadrass N,
lar sperm. Fertil Steril. 2015;104(6):1398–1405. Allahkhah A, Iran-Pour E, Safarinejad MR. Effects
79. Mehta A, Bolyakov A, Schlegel PN, Paduch DA. Higher of varicocele repair on spontaneous first trimes-
pregnancy rates using testicular sperm in men with ter miscarriage: A randomized clinical trial. Urol J.
severe oligospermia. Fertil Steril. 2015;104(6):1382–1387. 2012;9(2):505–513.
80. Chiba K, Fujisawa M. Clinical outcomes of varico- 94. Kirby EW, Wiener LE, Rajanahally S, Crowell K,
cele repair in infertile men: A review. World J Mens Coward RM. Undergoing varicocele repair before
Health. 2016;34(2):101–109. assisted reproduction improves pregnancy rate and
81. Chiba K, Ramasamy R, Lamb DJ, Lipshultz LI. The live birth rate in azoospermic and oligospermic men
varicocele: Diagnostic dilemmas, therapeutic chal- with a varicocele: A systematic review and meta-
lenges and future perspectives. Asian J Androl. analysis. Fertil Steril. 2016;106(6):1338–1343.
2016;18(2):276–281. 95. Practice Committee of the American Society for
82. Skoog SJ, Roberts KP, Goldstein M, Pryor JL. The ado- Reproductive M, Society for Male R, Urology. Report
lescent varicocele: What’s new with an old problem on varicocele and infertility: A committee opinion.
in young patients? Pediatrics. 1997;100(1):112–121. Fertil Steril. 2014;102(6):1556–1560.
Effects of chemical pollutants on
spermatogenesis and implications
12
in male infertility
CHRIS KC WONG

ENVIRONMENTAL POLLUTION AND REPRODUCTIVE A recent review postulated that about 40% of human deaths
HEALTH (62 million per year) is to a certain degree attributed to
Introduction exposure to chemical pollutants via different exposure
routes.8 To safeguard public health, instrumental chemi-
In the past century, the remarkable advancement in indus-
cal analysis has been adopted globally and routinely to
trialization and technology have driven changes to the
identify and quantify all these contaminants in order to
environment to a scope that is unprecedented in human
evaluate the possible risk of human exposure to EDCs
history. An overwhelming number of synthetic chemicals
and their metabolites.9 At present, the potentially harmful
of highly heterogeneous structures have been produced for
effects on human health and ecological well-being caused
industrial, medical, and domestic purposes. These chemi-
by endocrine disruptions are one of the top international
cal substances were initially thought to have negligible
concerns. A 2013 report by United Nations Environment
or no biological activity and are extensively used in our
Programme/World Health Organization (UNEP/WHO)
daily lives. The chemical residues are ubiquitous in our
showed that the prevalence of chronic disease is increas-
environment and are found to contaminate air, water, and
ing. An association between EDC exposure and a number
foods. Public awareness on the potential health impacts
of noncommunicable diseases in humans is advocated.
of these man-made chemicals, (i.e., pesticides) was stirred
Plausible causal links between EDC exposures to many
by the book Silent Spring published in 1962 and written
human chronic diseases are widely supported by both lab-
by American biologist Rachel Carson. In recognition of
oratory animal and epidemiological studies. The incidence
toxicities of chemicals in our ecosystem, some man-made
of reproductive problems is also a prevalent issue globally.
chemicals were excluded for their manufacture and usage.
A positive correlation between exposure of chemical pol-
For example, the insecticide dichlorodiphenyltrichloro-
lutants and the increased risk of male subfecundity has
ethane (DDT) was banned for agricultural use in 1972.
been reported.
Other chemicals, like polychlorinated biphenyls (PCBs),
the compounds widely used as dielectric and coolant flu-
ids in transformers and capacitors, were phased out in Human reproductive health is declining
1979. Although public concern about chemical pollution Data from the World Bank and WHO have revealed a
in our environment was elevated, the potential impacts declining trend (from 4.8 to 2.6) of world total fertility
of these chemicals on ecological and animal health were rates  from 1970–2010, affecting over 80 million people
largely undisclosed up until 1994, when the first World worldwide.10 Studies indicated that 30% of infertility cases
Wildlife Federation Wingspread Conference took place are related to male factor problems11 such as immunologi­
and global concerns were raised about the health effects cal  disorders, structure abnormality, and sperm produc-
of chemical pollution.1,2 Some of the more harmful chemi- tion. Recent data for 2011–2013 from WHO show low
cal pollutants were classified as endocrine disrupting fertility rates in developed countries in North America
chemicals (EDCs) since they could presumably interfere (1.6–2.0), Europe (1.4–2.0), and Asia (1.7–3.1) compared
with the synthesis, metabolism, and action of endogenous to  developing countries in Africa (4.1–5.7). To unravel
hormones.3,4 Since then, a considerable number of stud- mechanisms attributing to human infertility, a consider-
ies have been conducted to reveal the ecological distribu- able amount of resources have been allocated to identify
tion, animal body loading, and health effects of EDCs. genetic targets on fertility.12 However, most reported infer-
For instance, a study from the U.S. Centers for Disease tility cases remain unexplained. This may be due to our
Control (CDC) reported that Americans of all ages accu- limited grasp of the empirical mechanisms underlying
mulated over 116 extraneous chemicals into their bodies.5 fertility12 or that the cases were complicated by nongenetic
Over 358 industrial chemicals and pesticides were detected factors including diseases and/or exposure to environ-
in cord blood of American infants.6 The data have shown mental contaminants. In fact, clues regarding the decline
the ubiquitous contamination of chemical contaminants in human reproductive health can be revealed from inves-
in our environment and food-chains, leading to the pres- tigating numerous historical cases of pollution issues
ence of these exogenous chemicals in the human body.7 reported in both wildlife and humans.

155
156  Effects of chemical pollutants on spermatogenesis and implications in male infertility

A great deal of evidence has suggested that the exposure motility, and DNA integrity) in Northern Europeans,31–37
to EDCs is related to animal reproductive problems world- Americans,38 and Indians39 and serum testosterone
wide. Many incidences of pollutant exposure and conse- levels.31,34 In other epidemiological analysis, negative
quent adverse biological effects on reproductive function ­correlations of serum levels of DDT and its metabolite
have been described in wildlife. For example, in the 1990s, (dichlorodiphenyldichloroethylene p [p’-DDE]),33,36,40–43
adult male alligators in Lake Apopka in Central Florida perfluorinated compounds (PFCs),44–47 or urinary bisphe-
exposed to agricultural wastes (dicofol and DDT, its metab- nol A (BPA)48–51 concentrations with semen quality and
olites) produced low testosterone levels and presented reproductive hormones were observed. The exposure
micropenis with disorganized testes.13 A coordinated field and accumulation of organophosphate flame retardants
and laboratory study reported exposure to the herbicide were also linked to decreased semen quality in men.52,53
atrazine that caused gonadal dysgenesis and testicular Although these studies have shown that exposure to
oogenesis in leopard frogs.14,15 A report from CHEM Trust chemical pollutants exerted some negative effects on male
in 2008 revealed the negative impacts of chemical pollu- reproductive health, the results across studies are still not
tions on the reproductive health of male vertebrate wild- conclusive. Nevertheless, the observed effects were related
life, covering many different species of animals (i.e., fish, to the exposure to certain classes of chemical pollutants
amphibians, reptiles, birds, and mammals).16 Moreover, the and may be interlinked in the causation.54–56 To elucidate
report underlined the mixture effects of different classes of the possible causations, most prior research delineated
chemical contaminants on perturbing endocrine systems, that the actions of EDCs were mediated by estrogenic and/
leading to the deformities of sex-linked structures and fer- or (anti)-androgenic pathways. Ubiquitous environmental
tility of animals. In addition to the observations on genital contaminants and organic (i.e., DDT, hydroxylated PCBs,
disruption, effects of chronic, low-dose exposure to meth- BPA, p-nonylphenol, and dioxins) and inorganic (cad-
ylmercury on disrupting courtship behaviors and sexual mium and mercury) contaminants were found to exhibit
preference of white ibises were reported.17 This exposure either or both estrogenic and androgenic activity.57–59
caused a dose-related increase of male-male pairing behav- Some newly identified emerging contaminants, like PFCs
ior and was linked to reproductive deficits. Given the nega- (perfluorooctanoic acid [PFOA], perfluorooctane sulfo-
tive effects of EDC exposure on the genital development nate [PFOS]) and flame retardants were also reported to
and fertility of wildlife,18 the possible harmful effects of possess estrogenic activities.60,61 However, recent studies
environmental pollutants/chemicals on human fecundity have indicated that EDCs can act on different biological
do raise public concerns. components via diverse mechanisms. Other nuclear hor-
In fact, the decreasing trends in human fertility rates mone receptors are found to be the targets of EDCs.
and the manifestation of male reproductive health prob-
lems, cryptorchidism (undescended testis) and poor semen
GENERAL MECHANISTIC ACTIONS OF EDCs
quality, support the notion.19,20 In highly polluted areas or
chemical plants, human exposure to dioxins, polychlori- Receptor mediated pathways
nated biphenyls, polychlorinated dibenzofurans, or mixed Some EDCs or their metabolites perturb estrogenic,
petrochemicals were found to be associated with signifi- androgenic, and thyroid signaling as their chemical struc-
cant reductions in the ratio of male births to total number tures are similar to endogenous hormones.62 A common
of births.21–24 Epidemiological studies have reported an molecular mechanism is their direct interactions with
increased risk of genital malformations and cryptorchi- nuclear receptors (NRs) to modulate downstream signal-
dism in the children of workers exposed to pesticides.25,26 ing pathways and gene expressions. Numerous studies
In the Study for Future Families in the United States, a have demonstrated the stimulatory or inhibitory effects
positive correlation between prenatal exposure to several of some EDCs on estrogen receptors (ERs), androgen
phthalates and shortening of the anogenital index and receptors (ARs), retinoid X receptors (RXRs), peroxi-
incomplete testicular descent was observed.27,28 Although some proliferator-activated receptors (PPARs), constitu-
we are still far from understanding the mechanisms of the tive androstane receptors (CARs), pregnane X receptors
casual relationship, some data suggest that human hypo- (PXRs), glucocorticoid receptors (GRs), and thyroid hor-
spadias and cryptorchidism may be related to endocrine mone receptors (TRs).63 Moreover, different EDCs may
disruption by chemical contaminants.29 Based on pre- share common receptor targets and interfere with vari-
cautionary principles, avoidance of prenatal exposure to ous NRs expressed in several organs with different affini-
chemical pollutants is recommended.30 In addition to the ties and intensities. Therefore, EDC-elicited effects may
possible endocrine-disrupting effects on genital develop- result from perturbations at multiple tissue levels through
ment, many studies evaluated the impacts of the exposure cross-talking among different hormonal signaling path-
on levels of reproductive hormones and semen quality ways. Although investigations on the multiple interac-
(concentration, morphology, mobility, and chromatin tions would benefit our understanding of the fundamental
integrity of sperm). Epidemiological cross-sectional stud- mechanism of EDCs, there are still criticisms about inter-
ies reported negative correlations between serum PCBs or nal doses of EDCs and their potency in mimicking or
the proxy (2,2’4,4’,5,5’-hexachlorobiphenyl, CB-153) for inhibiting the biological actions of endogenous hormones.
total PCB exposure with semen quality (concentration, It is true that some EDCs (dioxins, diethylstilbestrol [DES]
Reproductive toxicity of EDCs from laboratory animal and cell studies  157

and 17α-ethinylestradiol [EE2]) show high or compara- BPA in maternal mice showed a shift of coat color distri-
tive affinity to the endogenous ligands in receptor binding bution in their offspring via reducing CpG methylation in
(i.e., aryl hydrocarbon receptor [AhR] or ERs), but most the Agouti gene.80 These examples illustrate that the leak-
EDCs have low (by several orders of magnitude) bind- ing of EDCs via placenta barriers to developmental fetus
ing affinity to the receptors.64 The concentrations of most is possible. Fetal life is vulnerable as the structural and
EDCs in human body fluid (i.e., blood) are 100–1000-folds functional events are rapidly developed. Therefore, the
lower than the experimental conditions that were used to perturbing effects of placenta-derived EDCs may lead to
demonstrate receptor binding or activation. Although the considerable changes in the developmental paths in the
additive/synergistic effects of EDC mixtures cannot be fetus, leading to a disturbance on the inherited genetic
neglected, it seems unlikely that EDCs can compete with program, the commitment of cells to specific lineages, and
the endogenous ligands for receptor binding. Therefore, the structural/functional differentiation in organogenesis,
with regard to the impact of EDCs (i.e., BPA, phthal- leading to chronic and/or transgenerational effects on the
ates, PFCs, and flame retardants) with weak or unidenti- exposed animals or increase disease susceptibility or dys-
fied hormonal activity, their mechanistic action leading function in their offspring.88
to animal infertility cannot be fully explained by direct
endocrine activities. Hence, the key question that needs to REPRODUCTIVE TOXICITY OF EDCs FROM
be addressed is whether the experimental observation of LABORATORY ANIMAL AND CELL STUDIES
receptor activation/inhibition by EDCs could happen in One of the current hypotheses proposes that EDCs cause dis-
real life. If this is not the case, could EDC-elicited action turbances on the regulatory pathways at the hypothalamic-
be due to some other spectrum of effects? Other than the pituitary-gonadal (HPG) axis to perturb the process of
receptor-mediated actions, EDC-elicited oxidative stress steroidogenesis in affecting the feedback circuit and circu-
has been recognized as the most common mechanism in lating levels of gonadotropin and sex hormones. EDCs may
affecting cellular structures and functions.65 Interestingly, affect the growth and maturation of gonads and the forma-
an elevated level of reactive oxygen species (ROS) has tion of functional gametes. Indeed, the inability to produce
been found in the semen plasma of infertile men.66,67 sufficient numbers of healthy sperm or nonobstructive
This inflammatory/oxidative effect may ultimately affect azoospermia is one of the major reasons for male subfecun-
gonadal functions for the production of healthy gametes dity and infertility.11 Numerous studies have revealed that
and lead to reduced fertility and fecundity in animals. the male reproductive system is the major target of chemi-
cal toxicants. Almost 60 years ago, Parizek and coworkers
Epigenetic changes on animal early development identified that the testis was sensitive to cadmium toxic-
Fetal growth and development are influenced by both ity, which caused testicular injury and male sterility.89 In
genomic and environmental factors.68,69 Presumably the recent years, experimental and epidemiological studies
exposure to EDCs of high potency in early life can pro- demonstrated the effects of EDC exposure on the manifes-
foundly influence the development and long-term health tation of testicular dysgenesis syndrome or the reduction of
of infants via alteration of signaling, gene expression, and male fertility through formulations of the biological rela-
epigenetic modifications.70–72 We have learned these lessons tionship of EDC exposure (i.e., dioxin, PCBs, pesticides,
from human exposure to synthetic estrogen, DES, and the BPA, PFCs, and flame retardants) to the reduction of tes-
sedative drug thalidomide, which induced changes in the tosterone and sperm production.30 Therefore, understand-
development of reproductive tracts in offspring, increased ing the actions and effects of EDCs on the feedback circuit,
incidence of vaginal cancer, and caused severe develop- steroidogenesis, and spermatogenesis can provide clues to
mental malformations.73,74 Converging evidence supports test the hypothesis and to elucidate the roles of EDCs on
the notion that the modulatory effects of different EDCs perturbing reproductive functions.
on epigenetic programming, cell signaling, and tissue
functions would lead to chronic and/or transgenerational Hypothalamic-pituitary gonadal axis
effects on the exposed animals or increase disease suscep- The hypothalamic kisspeptin (KiSS1)/G-protein-coupled
tibility in their offsprin.75,76 Numerous reports have dem- receptor (GPR) 54 system is essential for puberty matu-
onstrated the effects of EDCs on DNA methylation in early ration and gonadal function.90 The activation of KiSS1/
development of mice,77–80 abnormal embryonic karyotype GPR54 stimulates the release of GnRH, which controls the
and chromosome synapsis defects in humans,81,82 spine synthesis of gonadotrophin luteinizing hormone (LH) and
synapses formation in nonhuman primates,83 fetal mouse follicle-stimulating hormone (FSH) by the pituitary. LH
mammary gland development and stromal-epithelial and FSH act on testicular Leydig and Sertoli cells to regu-
signaling,84,85 and Hox gene expression and genitouri- late the processes of steroidogenesis and spermatogenesis,
nary development.86,87 Using in utero exposure studies in respectively.91,92 In rat models, neonatal BPA exposure (sub-
rodent models, transient exposure of gestating female rats cutaneous injection) were found to reduce hypothalamic
to a fungicide (vinclozolin) or a pesticide (methoxychlor) kisspeptin fiber density and KiSS-1 mRNA expression at the
decreased male infertility of their offspring.77 The effects prepubertal stages.93,94 A similar BPA exposure to neonatal
were found to be manifested via an alternation of DNA rats affected GnRH signaling at the hypothalamic-pituitary
methylation patterns in germ line. Dietary exposure to axis.95 Oral gavage male rats with BPA at early puberty
158  Effects of chemical pollutants on spermatogenesis and implications in male infertility

showed significantly greater numbers of ERα expression the developing child’s genome than did mothers.109 Using
in neurons in the arcuate nucleus and medial preoptic area a Caenorhabditis elegans (C. elegans) model, an impair-
of the hypothalamus,96 indicating a perturbation of neural ment effect of BPA on the double-strand break repair
circuit of estrogen signaling. Perinatal exposure to BPA machinery in the germ line was demonstrated.111 In rhe-
and di(2-ethylhexyl) phthalate (DEHP) caused a reduction sus monkeys, disturbing effects of BPA on the prophase
in the male-to-female sex ratio and sizes of testes in male events at the first meiotic division during early oogenesis
pups.97 The expression levels of some genes (i.e., antimul- has been reported.110 Even though the corresponding data
lerian hormone, androgen receptor, cyclin A, and steroido- in males of mammals are lacking, it is sensible to assume
genic acute regulatory protein [StAR]) in relation to the that a similar effect may happen during the meiotic divi-
growth and maturation of testes were significantly reduced. sion of male germ cells. In the latter stage of spermato-
In a pregnant mouse model, oral gavage of BPA to mothers’ genesis, the shaping and nuclear condensation of sperm
perturbed mRNA expressions of KiSS-1, GnRH, and FSH at head are tightly regulated processes involving extensive
the hypothalamic-pituitary axis and reduced the expression chromatin remodeling. Dysregulation of the BTB junc-
of steroidogenic enzymes, leading to a decrease of testoster- tion proteins in seminiferous epithelial homeostasis may
one levels in male pups.98 Retrospectively, a recent hypoth- cause an improper change in histone retention and epi-
esis has underlined that gonadal steroidogenesis is one of genetic modification of the developing sperm.112 Previous
the possible targets for EDCs.99,100 The modulatory effects of studies using the chemotherapeutic drug cyclophospha-
EDCs on steroidogenesis were reported in both in vivo and mide113 or vinclozolin77 have demonstrated their effects
in vitro studies.101,102 Using a testicular Leydig R2C cell-line on DNA methylation in male germ cells. Other studies
model, a treatment with BPA increased the expression of have verified the epigenetic action of BPA or phthalate
aromatase.103 Moreover, a recent study revealed that EDCs exposure on DNA methylation,80,114–120 histone modifica-
might target on ATP-binding cassette transporters in the tion,121 and microRNA expression122 in the germ/somatic
blood-testis barrier (BTB) to alter the process of testoster- cells of offspring or the subsequent passages in the cell-
one secretion.104 Therefore, EDCs could interfere with the line models. Collectively EDCs interfere with seminifer-
process of steroidogenesis and modulate the release of the ous epithelial function by disrupting the role of junctional
endogenous steroid hormones and also the hormonal feed- proteins, which in turn impede spermatogenesis and alter
back circuity. Collectively, altered serum levels of the ste- genetic and epigenetic changes in the developing sperm.
roid hormones may interfere with the feedback regulatory Remarkably, anomalous methylation profiles in human
mechanisms at the HPG axis, thereby perturbing normal spermatozoa were reported in men with low sperm counts
reproductive function. and infertility.123,124 Alteration in the normal epigenetic
state of sperms from infertile patients undergoing in vitro
Spermatogenesis fertilization (IVF) has also been proposed as the under-
The process is undertaken in the seminiferous epithelium lying cause of increased anomalies in IVF offspring.125,126
where an interaction of Sertoli cell–spermatogonium/ Further studies are recommended to delineate the effects
spermatid mediate cell cycle progression of germ cells, of EDCS on seminiferous epithelial function and the
spermiogenesis, and spermiation.105 Therefore, disrup- acquired epigenetic modification during spermatogenesis.
tion of the epithelial homeostasis at the Sertoli-Sertoli and Taken together, emerging evidence supports the notion
Sertoli-germ cell interface in the seminiferous epithelium that exposure to EDCs may cause adverse effects on ani-
would result in the loss of germ cell adhesion, sperma- mal reproductive functions via different mechanisms.
tid orientation, and dysregulation of spermatogenesis.106 However, it is still challenging to establish the causal rela-
Emerging evidence has revealed that EDC, like cadmium tionship between the exposure to the observed functional
or BPA elicited oxidative stress may disrupt the integrity disability. In fact, humans are exposed to a complex mix-
of the BTB in seminiferous tubules, leading to an impair- ture of environmental contaminants with heterogeneous
ment of sperm production.107,108 This observation has chemical structures. The effects of their interactions with
revealed the specific but nonhormonal-effect of EDCs on macromolecules at molecular levels may result from their
testicular function, in particular the empirical action of agonized or antagonized actions. Moreover, the outcomes
EDCs on the regulatory roles of “polarity proteins” and of the mixture effect are complicated by different routes,
BTB-associated proteins in the seminiferous epithelium. durations, and doses of the exposure at different devel-
Current evidence suggests that BTB-associated junctional opmental windows of animals. In the following section,
proteins and protein kinases are the potential targets of the historical background and reproductive toxicity of the
some EDCs, since those affect seminiferous epithelial emerging persistent organic contaminant, PFCs (synthetic
function and sperm production. In addition to sperm fluorinated hydrocarbons), is the focus of the discussion.
number, the integrity of sperm chromatin is known to
be a clinical marker of male fertility. The high rate of cell PFCs, HUMAN EXPOSURE RISK, AND REPRODUCTIVE
division of spermatogonium would provide a vulnerable HEALTH
time window for EDC’s actions. This scenario is depicted PFCs are a family of synthetic fluorinated hydrocarbons
in a recent whole genome sequencing study in which (C 4-C14) originally produced by 3M as fluorosurfactants in
fathers passed nearly four times more new mutations to 1949. PFCs have been used in fabric protectors and stain
PFCs, human exposure risk, and reproductive health  159

repellents for textiles and leather and oil-resistant coatings (T1/2 = 15 days) than in females (T1/2 = 1 day). Although
for food wrappers, waxes, and polishes.127 Due to their the underlying mechanism has yet to be elucidated,
numerous applications in commercial products, exposure testosterone was a key factor in the sex-specific differ-
of the general population to PFCs is widespread. Since ence in PFOA elimination.145 Adult rats treated with
the DuPont Washington Works fluorochemical manu- 0.05%–0.5% PFOA exhibited reduced activities of
facturing plant in Parkersburg, West Virginia, began to testicular-metabolic enzymes.146,147 Meanwhile, chronic
release PFCs into the environment, a C8 Health Project exposure of adult rats to PFDA (0.2, 0.5 mg·kg−1·day−1)
has been launched to measure serum concentrations of resulted in disruption of testicular steroidogenesis.148
PFCs in exposed residents. In a recent report from the C8 Using microsomal preparations of human and rat tes-
Health Project, high levels of PFCs (i.e., PFOA [32.9  ng/ tes, PFCs were found to target the steroidogenic enzyme
mL], PFOS [19.2 ng/mL], and perfluorohexane sulfonate 3β-hydroxysteroid dehydrogenase, resulting in a reduc-
[PFHS] [3.3  ng/mL]) were detected in blood samples tion in testosterone production.149 To elucidate the mech-
from 69,030 participants enrolled between 2003–2006.128 anisms of PFC-induced male reproductive disorder, a
Additional studies have reported the widespread con- proteomic analysis of testes from adult mice exposed to
tamination of human populations with PFCs (1–20 ng/ PFOA (0.21–20  mg·kg−1·day−1) for 4 weeks was utilized
mL).129,130 More importantly, placental transfer of PFCs to screen for molecular targets of PFOA.150 In addition
was demonstrated in humans by analyzing paired mater- to a reduction in testosterone synthesis and lower sperm
nal and umbilical cord blood samples from patients in quality, this study demonstrated that PFOA treatment led
various countries, including Japan, Norway, Germany, to reduced expression of the Leydig cell marker (insulin-
Korea, and Canada.131–135 The evidence of intrauterine like factor 3) and of steroidogenic enzymes, including the
exposure was further strengthened by comparing PFC cytochrome P450 cholesterol side-chain cleavage enzyme
(i.e., PFOA, PFOS) levels in blood samples from pregnant and 17β-hydroxysteroid dehydrogenase. The modulating
and nonpregnant women.136 These observations imply a effects of PFCs on testicular steroidogenesis were then
widespread and high risk of exposure to PFCs for fetuses supported by studies using human adrenocortical carci-
during early development. Moreover, breastfeeding is rec- noma (H295R) cells. In this cell line, exposure to PFOA,
ognized as a route of PFC exposure in infants.137 Because PFOS, or perfluorononanoic acid (PFNA), at concentra-
of the stability of carbon-fluoride bonds, PFCs are highly tions of 6 nM–600 μM, resulted in a reduction in the
resistant to biodegradation/metabolism. Indeed, lengthy mRNA expression levels of CYP11A.151 A similar study
serum elimination half-lives were reported in humans using H295R cells illustrated the inhibitory effects of six
(5 years)138,139 and mice (20 days).140 As a result, bioaccu- PFCs on the expression levels of steroidogenic enzymes
mulation of these chemicals can lead to greater adverse and on testosterone synthesis.152 These results are con-
health effects in humans. sistent with observations made in zebrafish embryos
Epidemiological studies demonstrated that the pres- exposed to PFOS (500 μg/L) in interfering with functions
ence of PFCs was implicated in male reproductive dys- of NRs and steroidogenesis.153
function, including reduced sperm count and semen In addition to its effects on steroidogenesis and sex hor-
quality, in humans. High serum PFC levels correlated mone production, studies have investigated the effects of
with low semen quality in a population of 105 Danish PFC exposure on sperm production. Qiu and coworkers
men44 and in 588 men from Artic and European popu- reported that exposure of CD-1 mice to PFOS (0.25–50
lations.47 A study of 247 Danish men also demonstrated mg·kg−1·day−1) was accompanied by disorganization of
a negative association between serum levels of PFCs and the BTB and reduced sperm count.154 These data were
testosterone production.45 Additionally, PFOA exposure supported by a follow-up experiment using cultured pri-
was positively correlated with a reduction in sperm DNA mary Sertoli cells, which revealed that PFOS (5–60 μM)
integrity in 199 subjects from Greenland.46 In the studies disrupted BTB integrity in vitro by affecting the localiza-
of 1240 pregnant women from the Danish National Birth tion of junctional proteins. PFOS (50 μM) treatment also
Cohort141 and 910 women enrolled in the Norwegian resulted in an oxidative stress-mediated reduction in the
Mother and Child Cohort,142 high maternal levels of PFCs expression of cytoskeletal proteins and a defect in cyto-
(i.e., PFOA and PFOS) were associated with a longer time- skeletal organization in a coculture of neonatal gonocytes
to-pregnancy. Furthermore, a follow-up study by a preg- and Sertoli cells.155 Correspondingly, in utero exposure
nancy cohort in Denmark demonstrated a link between of female rats to PFOS (5 or 20 mg·kg−1·day−1 maternal
in utero exposure to PFCs and reduced semen quality in a dose) from gestational day 11–19 reduced the number
group of 169 adult men.143 of fetal Leydig cells and impaired cell functions in com-
The toxicity of PFCs to male reproductive health has parison to the untreated control group, as evidenced by
primarily been evaluated using animal and cell culture a decrease in steroidogenic activity and in testosterone
models. Early studies using 14C-labeled perfluorodeca- levels.156 Oral exposure to PFOS (0.5–6 mg·kg−1·day−1) for
noic acid (PFDA) and PFOA demonstrated covalent 4 weeks caused a disruption in the reproductive axis, in
binding of these compounds to cellular proteins in the particular a loss of spermatozoids, in adult male rats.157
plasma, liver, and testes of rats.144 Interestingly, renal PFOS-elicited degeneration of spermatogonium was also
excretion of PFOA was significantly slower in male rats reported in the testes of Xenopus laevis.158 Furthermore,
160  Effects of chemical pollutants on spermatogenesis and implications in male infertility

PFOA exposure (1, 3, 5, 10, 20, or 40 mg·kg-1·day-1) in proteins and activity of focal adhesion kinase at Sertoli
timed-pregnant CD-1 mice (GD 1-17) enhanced sexual cell BTB. Significant reductions of actin crossing linking
maturation in male offspring.159 Exposure of adult male proteins (filamin A, palladin), tight junctional proteins
CD-1 mice to PFOS (1–10 mg·kg−1·day−1) caused pro- (zonula occludens-1, occludin), adherens junction pro-
gressive deterioration of testicular signaling manifested teins (β-catenin, N-cadherin), and the gap junction (GJ)
by significant reductions in testosterone synthesis and protein connexin-43 (Cx43) were observed.190 The over-
epididymal sperm counts.160 Collectively, these findings expression of Cx43 rescued PFOs-disrupted TJ barrier.191
support the human epidemiological studies to indicate Intriguingly, CX43 is known to be a downstream target
the negative association between high serum levels of of the lipid mediator PG. Therefore, further investigation
PFCs and testicular steroidogenesis, testosterone pro- is necessary to determine whether PFC exposure disrupts
duction, and semen quality. AA metabolism, and the PG signaling pathway in particu-
lar, resulting in male sexual dysfunction.
Perfluoroalkyl chemicals and their mechanistic
activities CONCLUSION
PFCs are classified as peroxisome proliferators. This class EDC-related health issues have been widely reported in
of molecules activate ligand-dependent transcription scientific journals, appearing extensively in the media,
factors of the PPAR family to alter lipid metabolism and and have sparked considerable discussion in society.
cell differentiation.161–164 PPARs belong to the steroid hor- Many reports have suggested that EDCs could modulate
mone receptor superfamily and are able to interact with the masculinization process during human fetal develop-
steroid receptors to modify the activities of other steroid ment and result in the manifestation of reproductive dis-
hormones.165 The identification of PPARs in human testes order similar to testicular dysgenesis syndrome. Given the
and developing rat testes suggests PPAR-signaling may adverse effects of EDC exposure on reproductive health,
play a role in testicular cell differentiation during early one can surmise that the global increase in the prevalence
development.166 Animal and cell culture studies revealed of subfertility in humans is likely due to an increased
that an activation of PPARs in the testes was associated exposure to EDCs. While the increasing incidence of
with gonadal steroidogenesis,167 spermatogenesis,168 and reproductive problems is a global issue, the mechanism
Sertoli cell metabolism.169 Conversely, inactivation of by which EDCs predispose humans to reproductive dys-
PPAR-elicited peroxisome β-oxidation in mouse testes function is still unclear. Identification of the effects and
was found to negatively affect spermatogenesis and cause mechanistic actions of EDCs on reproductive functions in
male infertility.170 Although various studies had illus- humans would help to understand their influence on ani-
trated the involvement of the PPAR-pathways in PFC- mal health. Changes in government policies, manufactur-
elicited intoxication, studies of wild-type and PPAR-α ing best practices, and even in the lifestyles of people can
null mice treated with PFOS (3–10 mg·kg−1·day−1) dem- help to eliminate the excessive use of man-made chemicals
onstrated PPAR-independent effects on reproductive and thus lessen the immense financial burden of EDC-
health.171,172 elicited diseases on the global health care system.
In previous studies, exposure to PFOS resulted in
increased expression of the arachidonic acid (AA)- REFERENCES
metabolizing cytochrome P450 (CYP) isoforms, CYP2, and 1. Colborn T, vom Saal FS, Soto AM. Developmental
CYP4.173,174 These enzymes convert AA into hydroxyeicosa- effects of endocrine-disrupting chemicals in
tetraenoic acids (HETEs) or epoxyeicosatrienoic acids wildlife and humans. Environ Health Perspect.
(EETs). It was proposed that the PFOS-mediated perturba- 1993;101(5):378–384.
tion of fatty acid metabolism and the resulting physiologi- 2. Soto AM, Rubin BS, Sonnenschein C. Interpreting
cal outcomes were due to the modulation of lipid signaling endocrine disruption from an integrative biology
via dysregulation of the AA cascade.175 Interestingly, a perspective. Mol Cell Endocrinol. 2009;304(1–2):3–7.
study reported that certain endocrine-disrupting chemi- 3. Phillips KP, Foster WG, Leiss W et al. Assessing and
cals (i.e., BPA and phthalates) inhibited the prostaglandin managing risks arising from exposure to endocrine-
(PG) signaling pathway in a mouse Sertoli cell line and active chemicals. J Toxicol Environ Health B Crit Rev.
in ex vivo rat testes.176 PGs are a group of lipid signaling 2008;11(3–4):351–372.
mediators generated by the oxidation of AA. AA and the 4. Phillips KP, Foster WG. Key developments in endo-
lipoxygenase-catalyzed products of the AA cascade play crine disrupter research and human health. J Toxicol
critical roles in steroidogenesis, early male sexual devel- Environ Health B Crit Rev. 2008;11(3–4):322–344.
opment, and masculinization.177–187 Interestingly, most 5. Centers for Disease Control (CDC). Second national
lipid-mediators (i.e., PGs and HETEs) produced by this report on human exposure to environmental chemi-
cascade are also agonists of PPARs.188,189 Using primary cals, Atlanta, GA: CDC, Nov. 11, 2003.
mouse Sertoli cell cultures, PFOS treatment (10–20 6. Environmental Working Group (EWG). Pollution in
μM) disrupted actin polymerization and bundling via people: Cord blood contaminants in minority new-
affecting the levels and/or localization of cell adhesion borns. Jul. 2009.
References 161

7. Rappaport SM, Barupal DK, Wishart D, Vineis P, 23. Mocarelli P, Gerthoux PM, Ferrari E et al. Paternal
Scalbert A. The blood exposome and its role in dis- concentrations of dioxin and sex ratio of offspring.
covering causes of disease. Environ Health Perspect. Lancet. 2000;355(9218):1858–1863.
2014;122(8):769–774. 24. Moshammer H, Neuberger M. Sex ratio in the
8. Pimentel D, Cooperstein S, Randell H et al. Ecology children of the Austrian chloracne cohort. Lancet.
of increasing diseases: Population growth and envi- 2000;356(9237):1271–1272.
ronmental degradation. Hum Ecol. 2007;35:653–668. 25. Garcia-Rodriguez J, Garcia-Martin M, Nogueras-
9. Hotchkiss AK, Rider CV, Blystone CR et al. Fifteen Ocana M et al. Exposure to pesticides and cryptorchi-
years after “Wingspread”—Environmental endo- dism: Geographical evidence of a possible association.
crine disrupters and human and wildlife health: Environ Health Perspect. 1996;104(10):​1090–1095.
Where we are today and where we need to go. Toxicol 26. Weidner IS, Moller H, Jensen TK, Skakkebaek
Sci. 2008;105(2):235–259. NE. Cryptorchidism and hypospadias in sons of
10. World Bank. https://data.worldbank.org/indicator. gardeners and farmers. Environ Health Perspect.
Accessed June 22, 2018. 1998;106(12):793–796.
11. Sharlip ID, Jarow JP, Belker AM et al. Best prac- 27. Swan SH, Main KM, Liu F et al. Decrease in ano-
tice policies for male infertility. Fertil Steril. genital distance among male infants with prena-
2002;77(5):873–882. tal phthalate exposure. Environ Health Perspect.
12. Matzuk MM, Lamb DJ. The biology of infertility: 2005;113(8):1056–1061.
Research advances and clinical challenges. Nat Med. 28. Jurewicz J, Hanke W. Exposure to phthalates:
2008;14(11):1197–1213. Reproductive outcome and children health. A review
13. Guillette LJ Jr, Gross TS, Masson GR, Matter JM, of epidemiological studies. Int J Occup Med Environ
Percival HF, Woodward AR. Developmental abnor- Health. 2011;24(2):115–141.
malities of the gonad and abnormal sex hormone 29. Hsieh MH, Breyer BN, Eisenberg ML, Baskin LS.
concentrations in juvenile alligators from contami- Associations among hypospadias, cryptorchidism,
nated and control lakes in Florida. Environ Health anogenital distance, and endocrine disruption. Curr
Perspect. 1994;102(8):680–688. Urol Rep. 2008;9(2):137–142.
14. Hayes T, Haston K, Tsui M, Hoang A, Haeffele C, 30. Toppari J, Virtanen HE, Main KM, Skakkebaek NE.
Vonk A. Herbicides: Feminization of male frogs in Cryptorchidism and hypospadias as a sign of tes-
the wild. Nature. 2002;419(6910):895–896. ticular dysgenesis syndrome (TDS): Environmental
15. Hayes T, Haston K, Tsui M, Hoang A, Haeffele C, connection. Birth Defects Res A Clin Mol Teratol.
Vonk A. Atrazine-induced hermaphroditism at 2010;88(10):910–919.
0.1 ppb in American leopard frogs (Rana pipiens): 31. Richthoff J, Rylander L, Jonsson BA et al. Serum
Laboratory and field evidence. Environ Health levels of 2,2’,4,4’,5,5’-hexachlorobiphenyl (CB-153)
Perspect. 2003;111(4):568–575. in relation to markers of reproductive function in
16. CHEMTrust. Effects of pollutants on the reproduc- young males from the general Swedish population.
tive health of male vertebrate wildlife—Males under Environ Health Perspect. 2003;111(4):409–413.
threat. Nov 11, 2008. 32. Toft G, Rignell-Hydbom A, Tyrkiel E et al. Semen
17. Frederick P, Jayasena N. Altered pairing behaviour quality and exposure to persistent organochlorine
and reproductive success in white ibises exposed to pollutants. Epidemiology. 2006;17(4):450–458.
environmentally relevant concentrations of methyl- 33. Spano M, Toft G, Hagmar L et al. Exposure to PCB
mercury. Proc Biol Sci. 2011;278(1713):1851–1857. and p, p’-DDE in European and Inuit populations:
18. Lyons G. Viewpoint: Policy requirements for pro- Impact on human sperm chromatin integrity. Hum
tecting wildlife from endocrine disruptors. Environ Reprod. 2005;20(12):3488–3499.
Health Perspect. 2006;114 Suppl 1:142–146. 34. Bonde JP, Toft G, Rylander L et al. Fertility and
19. Andersson AM, Jorgensen N, Main KM et al. markers of male reproductive function in Inuit and
Adverse trends in male reproductive health: We may European populations spanning large contrasts in
have reached a crucial ‘tipping point.’ Int J Androl. blood levels of persistent organochlorines. Environ
2008;31(2):74–80. Health Perspect. 2008;116(3):269–277.
20. Skakkebaek NE, Jorgensen N, Main KM et al. 35. Stronati A, Manicardi GC, Cecati M et al.
Is human fecundity declining? Int J Androl. Relationships between sperm DNA fragmentation,
2006;29(1):2–11. sperm apoptotic markers and serum levels of CB-153
21. del RG, I, Marshall T, Tsai P, Shao YS, Guo YL. and p,p’-DDE in European and Inuit populations.
Number of boys born to men exposed to polychlori- Reproduction. 2006;132(6):949–958.
nated byphenyls. Lancet. 2002;360(9327):143–144. 36. Rignell-Hydbom A, Rylander L, Giwercman A
22. Mackenzie CA, Lockridge A, Keith M. Declining sex et  al. Exposure to PCBs and p,p’-DDE and human
ratio in a first nation community. Environ Health sperm chromatin integrity. Environ Health Perspect.
Perspect. 2005;113(10):1295–1298. 2005;113(2):175–179.
162  Effects of chemical pollutants on spermatogenesis and implications in male infertility

37. Dallinga JW, Moonen EJ, Dumoulin JC, Evers JL, 51. Lassen TH, Frederiksen H, Jensen TK et al. Urinary
Geraedts JP, Kleinjans JC. Decreased human semen bisphenol A levels in young men: Association with
quality and organochlorine compounds in blood. reproductive hormones and semen quality. Environ
Hum Reprod. 2002;17(8):1973–1979. Health Perspect. 2014;122(5):478–484.
38. Hauser R, Chen Z, Pothier L, Ryan L, Altshul L. 52. Meeker JD, Stapleton HM. House dust concentra-
The relationship between human semen parameters tions of organophosphate flame retardants in relation
and environmental exposure to polychlorinated to hormone levels and semen quality parameters.
biphenyls and p,p’-DDE. Environ Health Perspect. Environ Health Perspect. 2010;118(3):318–323.
2003;111(12):1505–1511. 53. Meeker JD, Cooper EM, Stapleton HM, Hauser
39. Rozati R, Reddy PP, Reddanna P, Mujtaba R. Role of R. Exploratory analysis of urinary metabolites of
environmental estrogens in the deterioration of male phosphorus-containing flame retardants in rela-
factor fertility. Fertil Steril. 2002;78(6):1187–1194. tion to markers of male reproductive health. Endocr
40. Haugen TB, Tefre T, Malm G et al. Differences in Disruptors (Austin). 2013;1(1):e26306.
serum levels of CB-153 and p,p’-DDE, and reproduc- 54. Sharpe RM, Skakkebaek NE. Testicular dysgenesis syn-
tive parameters between men living south and north drome: Mechanistic insights and potential new down-
in Norway. Reprod Toxicol. 2011;32(3):261–267. stream effects. Fertil Steril. 2008;89(2 Suppl):e33–e38.
41. Hauser R, Altshul L, Chen Z et al. Environmental 55. Skakkebaek NE, Rajpert-De ME, Jorgensen N et al.
organochlorines and semen quality: Results of a pilot Testicular cancer trends as ‘whistle blowers’ of tes-
study. Environ Health Perspect. 2002;110(3):229–233. ticular developmental problems in populations. Int J
42. de Jager C, Farias P, Barraza-Villarreal A et al. Androl. 2007;30(4):198–204.
Reduced seminal parameters associated with envi- 56. Skakkebaek NE, Rajpert-De ME, Main KM.
ronmental DDT exposure and p,p’-DDE concentra- Testicular dysgenesis syndrome: An increasingly
tions in men in Chiapas, Mexico: A cross-sectional common developmental disorder with environmen-
study. J Androl. 2006;27(1):16–27. tal aspects. Hum Reprod. 2001;16(5):972–978.
43. Aneck-Hahn NH, Schulenburg GW, Bornman MS, 57. Martin MB, Reiter R, Pham T et al. Estrogen-like
Farias P, de Jager C. Impaired semen quality asso- activity of metals in MCF-7 breast cancer cells.
ciated with environmental DDT exposure in young Endocrinology. 2003;144(6):2425–2436.
men living in a malaria area in the Limpopo Province, 58. Johnson MD, Kenney N, Stoica A et al. Cadmium
South Africa. J Androl. 2007;28(3):423–434. mimics the in vivo effects of estrogen in the uterus
44. Joensen UN, Bossi R, Leffers H, Jensen AA, and mammary gland. Nat Med. 2003;9(8):1081–1084.
Skakkebaek NE, Jorgensen N. Do perfluoroalkyl 59. McLachlan JA. Environmental signaling: What
compounds impair human semen quality? Environ embryos and evolution teach us about endocrine dis-
Health Perspect. 2009;117(6):923–927. rupting chemicals. Endocr Rev. 2001;22(3):319–341.
45. Joensen UN, Veyrand B, Antignac JP et al. PFOS 60. Maras M, Vanparys C, Muylle F et al. Estrogen-like
(perfluorooctanesulfonate) in serum is nega- properties of fluorotelomer alcohols as revealed by
tively associated with testosterone levels, but not mcf-7 breast cancer cell proliferation. Environ Health
with semen quality, in healthy men. Hum Reprod. Perspect. 2006;114(1):100–105.
2013;28(3):599–608. 61. Meerts IA, Letcher RJ, Hoving S et al. In vitro
46. Specht IO, Hougaard KS, Spano M et al. Sperm DNA estrogenicity of polybrominated diphenyl ethers,
integrity in relation to exposure to environmen- hydroxylated PDBEs, and polybrominated bisphe-
tal perfluoroalkyl substances—A study of spouses nol A compounds. Environ Health Perspect. 2001;​
of pregnant women in three geographical regions. 109(4):399–407.
Reprod Toxicol. 2012;33(4):577–583. 62. Crews D, McLachlan JA. Epigenetics, evolu-
47. Toft G, Jonsson BA, Lindh CH et al. Exposure to per- tion, endocrine disruption, health, and disease.
fluorinated compounds and human semen quality Endocrinology. 2006;147(6 Suppl):S4–S10.
in Arctic and European populations. Hum Reprod. 63. Casals-Casas C, Desvergne B. Endocrine disruptors:
2012;27(8):2532–2540. From endocrine to metabolic disruption. Annu Rev
48. Meeker JD, Ehrlich S, Toth TL et al. Semen qual- Physiol. 2011;73:135–162.
ity and sperm DNA damage in relation to urinary 64. Vandenberg LN, Maffini MV, Sonnenschein C,
bisphenol A among men from an infertility clinic. Rubin BS, Soto AM. Bisphenol-A and the great
Reprod Toxicol. 2010;30(4):532–539. divide: A review of controversies in the field of endo-
49. Li DK, Zhou Z, Miao M et al. Urine bisphenol-A crine disruption. Endocr Rev. 2009;30(1):75–95.
(BPA) level in relation to semen quality. Fertil Steril. 65. Braconi D, Bernardini G, Santucci A. Linking protein
2011;95(2):625–630. oxidation to environmental pollutants: Redox pro-
50. Knez J, Kranvogl R, Breznik BP, Voncina E, Vlaisavljevic teomic approaches. J Proteomics. 2011;​74(11):2324–2337.
V. Are urinary bisphenol A levels in men related to 66. Sheweita SA, Tilmisany AM, Al Sawaf H. Mechanisms
semen quality and embryo development after medically of male infertility: Role of antioxidants. Curr Drug
assisted reproduction? Fertil Steril. 2014;101(1):215–221. Metab. 2005;6(5):495–501.
References 163

67. Aitken RJ, Sawyer D. The human spermatozoon— 83. Leranth C, Hajszan T, Szigeti-Buck K, Bober J,
Not waving but drowning. Adv Exp Med Biol. MacLusky NJ. Bisphenol A prevents the synaptogenic
2003;518:85–98. response to estradiol in hippocampus and prefrontal
68. Bateson P, Barker D, Clutton-Brock T et al. cortex of ovariectomized nonhuman primates. Proc
Developmental plasticity and human health. Nature. Natl Acad Sci U S A. 2008;105(37):14187–14191.
2004;430(6998):419–421. 84. Soto AM, Sonnenschein C. Environmental causes
69. Gluckman PD, Hanson MA, Mitchell MD. of cancer: Endocrine disruptors as carcinogens. Nat
Developmental origins of health and disease: Reduc­ Rev Endocrinol. 2010;6(7):363.
ing the burden of chronic disease in the next genera- 85. Vandenberg LN, Maffini MV, Wadia PR, Sonnenschein
tion. Genome Med. 2010;2(2):14. C, Rubin BS, Soto AM. Exposure to environmentally
70. Gluckman PD, Hanson MA, Pinal C. The develop- relevant doses of the xenoestrogen bisphenol-A alters
mental origins of adult disease. Matern Child Nutr. development of the fetal mouse mammary gland.
2005;1(3):130–141. Endocrinology. 2007;148(1):​116–127.
71. Gluckman PD, Hanson MA, Spencer HG, Bateson 86. Block K, Kardana A, Igarashi P, Taylor HS. In utero
P. Environmental influences during development diethylstilbestrol (DES) exposure alters Hox gene
and their later consequences for health and disease: expression in the developing mullerian system.
Implications for the interpretation of empirical FASEB J. 2000;14(9):1101–1108.
studies. Proc Biol Sci. 2005;272(1564):671–677. 87. Yiee JH, Baskin LS. Environmental factors in genito-
72. Gluckman PD, Hanson MA, Beedle AS. Early life urinary development. J Urol. 2010;184(1):34–41.
events and their consequences for later disease: A 88. Jirtle RL, Skinner MK. Environmental epigenomics and
life history and evolutionary perspective. Am J Hum disease susceptibility. Nat Rev Genet. 2007;8(4):253–262.
Biol. 2007;19(1):1–19. 89. Parizek J, Zahor Z. Effect of cadmium salts on tes-
73. Finnell RH, Waes JG, Eudy JD, Rosenquist TH. ticular tissue. Nature. 1956;177(4518):1036.
Molecular basis of environmentally induced birth 90. Castellano JM, Roa J, Luque RM et al. KiSS-1/​
defects. Annu Rev Pharmacol Toxicol. 2002;42:​181–208. kisspeptins and the metabolic control of reproduction:
74. Rubin MM. Antenatal exposure to DES: Lessons Physiologic roles and putative physiopathological
learned...future concerns. Obstet Gynecol Surv. 2007;​ implications. Peptides. 2009;30(1):139–145.
62(8):548–555. 91. Pineda R, Aguilar E, Pinilla L, Tena-Sempere M.
75. Diamanti-Kandarakis E, Palioura E, Kandarakis Physiological roles of the kisspeptin/GPR54 system
SA, Koutsilieris M. The impact of endocrine dis- in the neuroendocrine control of reproduction. Prog
ruptors on endocrine targets. Horm Metab Res. Brain Res. 2010;181:55–77.
2010;42(8):543–552. 92. Roseweir AK, Millar RP. The role of kisspeptin in
76. Ho SM, Johnson A, Tarapore P, Janakiram V, Zhang the control of gonadotrophin secretion. Hum Reprod
X, Leung YK. Environmental epigenetics and its Update. 2009;15(2):203–212.
implication on disease risk and health outcomes. 93. Navarro VM, Sanchez-Garrido MA, Castellano JM
ILAR J. 2012;53(3–4):289–305. et al. Persistent impairment of hypothalamic KiSS-1
77. Anway MD, Cupp AS, Uzumcu M, Skinner MK. system after exposures to estrogenic compounds
Epigenetic transgenerational actions of endo- at critical periods of brain sex differentiation.
crine disruptors and male fertility. Science. 2005;​ Endocrinology. 2009;150(5):2359–2367.
308(5727):1466–1469. 94. Patisaul HB, Todd KL, Mickens JA, Adewale HB.
78. Anway MD, Skinner MK. Epigenetic programming Impact of neonatal exposure to the ERalpha agonist
of the germ line: Effects of endocrine disruptors PPT, bisphenol-A or phytoestrogens on hypotha-
on the development of transgenerational disease. lamic kisspeptin fiber density in male and female
Reprod Biomed Online. 2008;16(1):23–25. rats. Neurotoxicology. 2009;30(3):350–357.
79. Bernal AJ, Jirtle RL. Epigenomic disruption: The 95. Fernandez M, Bianchi M, Lux-Lantos V, Libertun C.
effects of early developmental exposures. Birth Neonatal exposure to bisphenol a alters reproductive
Defects Res A Clin Mol Teratol. 2010;88(10):938–944. parameters and gonadotropin releasing hormone
80. Dolinoy DC, Huang D, Jirtle RL. Maternal nutrient signaling in female rats. Environ Health Perspect.
supplementation counteracts bisphenol A-induced 2009;117(5):757–762.
DNA hypomethylation in early development. Proc 96. Ceccarelli I, Della SD, Fiorenzani P, Farabollini F,
Natl Acad Sci U S A. 2007;104(32):13056–13061. Aloisi AM. Estrogenic chemicals at puberty change
81. Susiarjo M, Hassold TJ, Freeman E, Hunt PA. ERalpha in the hypothalamus of male and female
Bisphenol A exposure in utero disrupts early oogen- rats. Neurotoxicol Teratol. 2007;29(1):108–115.
esis in the mouse. PLoS Genet. 2007;3(1):e5. 97. Wei X, Wan HT, Zhao YG, Wong MH, Giesy JP,
82. Sugiura-Ogasawara M, Ozaki Y, Sonta S, Makino Wong CK. Effects of perinatal exposure to bisphe-
T, Suzumori K. Exposure to bisphenol A is asso- nol A and di(2-ethylhexyl)-phthalate on gonadal
ciated with recurrent miscarriage. Hum Reprod. development of male mice. Environ Sci Pollut Res Int.
2005;20(8):2325–2329. 2012;19(7):2515–2527.
164  Effects of chemical pollutants on spermatogenesis and implications in male infertility

98. Wei X, Lee CK, Yeung WS et al. Effect of perina- 112. Lelievre SA. Contributions of extracellular matrix
tal and postnatal bisphenol A exposure to the signaling and tissue architecture to nuclear mecha-
regulatory circuits at the hypothalamus-pituitary- nisms and spatial organization of gene expression
gonadal axis of CD-1 mice. Reprod Toxicol. 2011;​ control. Biochim Biophys Acta. 2009;1790(9):925–935.
31(4):409–417. 113. Barton TS, Robaire B, Hales BF. Epigenetic program-
99. Sanderson JT. The steroid hormone biosynthesis ming in the preimplantation rat embryo is disrupted
pathway as a target for endocrine-disrupting chemi- by chronic paternal cyclophosphamide exposure.
cals. Toxicol Sci. 2006;94(1):3–21. Proc Natl Acad Sci U S A. 2005;102(22):7865–7870.
100. Scott HM, Mason JI, Sharpe RM. Steroidogenesis 114. Ho SM, Tang WY, Belmonte dF, Prins GS.
in the fetal testis and its susceptibility to dis- Developmental exposure to estradiol and bisphenol
ruption by exogenous compounds. Endocr Rev. A increases susceptibility to prostate carcinogenesis
2009;30(7):883–925. and epigenetically regulates phosphodiesterase type
101. Peretz J, Gupta RK, Singh J, Hernandez-Ochoa I, 4 variant 4. Cancer Res. 2006;66(11):5624–5632.
Flaws JA. Bisphenol A impairs follicle growth, inhib- 115. Tang WY, Morey LM, Cheung YY, Birch L, Prins
its steroidogenesis, and downregulates rate-limiting GS, Ho SM. Neonatal exposure to estradiol/
enzymes in the estradiol biosynthesis pathway. bisphenol A alters promoter methylation and
Toxicol Sci. 2011;119(1):209–217. expression of Nsbp1  and Hpcal1 genes and tran-
102. Zhang X, Chang H, Wiseman S et al. Bisphenol A scriptional programs of Dnmt3a/b and Mbd2/4 in
disrupts steroidogenesis in human H295R cells. the rat prostate gland throughout life. Endocrinology.
Toxicol Sci. 2011;121(2):320–327. 2012;153(1):42–55.
103. Kim JY, Han EH, Kim HG, Oh KN, Lee KY, Jeong HG. 116. Bromer JG, Zhou Y, Taylor MB, Doherty L, Taylor
Bisphenol A-induced aromatase activation is mediated HS. Bisphenol-A exposure in utero leads to epi-
by cyclooxygenase-2 up-regulation in rat testicular genetic alterations in the developmental program-
Leydig cells. Toxicol Lett. 2010;93(2):200–208. ming of uterine estrogen response. FASEB J. 2010;​
104. Dankers AC, Roelofs MJ, Piersma AH et al. 24(7):2273–2280.
Endocrine disruptors differentially target ATP- 117. Yaoi T, Itoh K, Nakamura K, Ogi H, Fujiwara Y,
binding cassette transporters in the blood-testis bar- Fushiki S. Genome-wide analysis of epigenomic
rier and affect Leydig cell testosterone secretion in alterations in fetal mouse forebrain after exposure
vitro. Toxicol Sci. 2013;136(2):382–391. to low doses of bisphenol A. Biochem Biophys Res
105. Cheng CY, Mruk DD. The blood-testis barrier and its Commun. 2008;376(3):563–567.
implications for male contraception. Pharmacol Rev. 118. Weng YI, Hsu PY, Liyanarachchi S et al. Epigenetic
2012;64(1):16–64. influences of low-dose bisphenol A in primary human
106. Wan HT, Mruk DD, Wong CK, Cheng CY. The api- breast epithelial cells. Toxicol Appl Pharmacol. 2010;​
cal ES-BTB-BM functional axis is an emerging tar- 248(2):111–121.
get for toxicant-induced infertility. Trends Mol Med. 119. Wu S, Zhu J, Li Y et al. Dynamic effect of di-
2013;19(7):396–405. 2-(ethylhexyl) phthalate on testicular toxicity:
107. Li MW, Mruk DD, Lee WM, Cheng CY. Disruption Epigenetic changes and their impact on gene expres-
of the blood-testis barrier integrity by bisphenol A sion. Int J Toxicol. 2010;29(2):193–200.
in vitro: Is this a suitable model for studying blood- 120. Kang SC, Lee BM. DNA methylation of estrogen
testis barrier dynamics? Int J Biochem Cell Biol. receptor alpha gene by phthalates. J Toxicol Environ
2009;41(11):2302–2314. Health A. 2005;68(23–24):1995–2003.
108. Siu ER, Wong EW, Mruk DD, Sze KL, Porto CS, 121. Doherty LF, Bromer JG, Zhou Y, Aldad TS, Taylor
Cheng CY. An occludin-focal adhesion kinase HS. In utero exposure to diethylstilbestrol (DES) or
protein complex at the blood-testis barrier: A bisphenol-A (BPA) increases EZH2 expression in the
study using the cadmium model. Endocrinology. mammary gland: An epigenetic mechanism linking
2009;150(7):3336–3344. endocrine disruptors to breast cancer. Horm Cancer.
109. Kong A, Frigge ML, Masson G et al. Rate of de novo 2010;1(3):146–155.
mutations and the importance of father’s age to dis- 122. Avissar-Whiting M, Veiga KR, Uhl KM et al. Bisphenol
ease risk. Nature. 2012;488(7412):471–475. A exposure leads to specific microRNA alterations
110. Hunt PA, Lawson C, Gieske M et al. Bisphenol A in placental cells. Reprod Toxicol. 2010;29(4):401–406.
alters early oogenesis and follicle formation in the 123. Hammoud SS, Purwar J, Pflueger C, Cairns BR,
fetal ovary of the rhesus monkey. Proc Natl Acad Sci Carrell DT. Alterations in sperm DNA methylation
U S A. 2012;109(43):17525–17530. patterns at imprinted loci in two classes of infertility.
111. Allard P, Colaiacovo MP. Bisphenol A impairs the Fertil Steril. 2010;94(5):1728–1733.
double-strand break repair machinery in the germ- 124. Marques CJ, Costa P, Vaz B et al. Abnormal methylation
line and causes chromosome abnormalities. Proc of imprinted genes in human sperm is associated with
Natl Acad Sci U S A. 2010;107(47):20405–20410. oligozoospermia. Mol Hum Reprod. 2008;14(2):67–74.
References 165

125. Manipalviratn S, DeCherney A, Segars J. Imprinting 138. Olsen GW, Burris JM, Ehresman DJ et al. Half-life of
disorders and assisted reproductive technology. serum elimination of perfluorooctanesulfonate, per-
Fertil Steril. 2009;91(2):305–315. fluorohexanesulfonate, and perfluorooctanoate in
126. Steel AJ, Sutcliffe A. Long-term health implications retired fluorochemical production workers. Environ
for children conceived by IVF/ICSI. Hum Fertil Health Perspect. 2007;115(9):1298–1305.
(Camb). 2009;12(1):21–27. 139. Seals R, Bartell SM, Steenland K. Accumulation and
127. Paul AG, Jones KC, Sweetman AJ. A first global pro- clearance of perfluorooctanoic acid (PFOA) in cur-
duction, emission, and environmental inventory rent and former residents of an exposed community.
for perfluorooctane sulfonate. Environ Sci Technol. Environ Health Perspect. 2011;119(1):119–124.
2009;43(2):386–392. 140. Lou I, Wambaugh JF, Lau C et al. Modeling single
128. Frisbee SJ, Brooks AP, Jr., Maher A et al. The C8 and repeated dose pharmacokinetics of PFOA in
health project: Design, methods, and participants. mice. Toxicol Sci. 2009;107(2):331–341.
Environ Health Perspect. 2009;117(12):1873–1882. 141. Fei C, McLaughlin JK, Lipworth L, Olsen J. Maternal
129. Calafat AM, Wong LY, Kuklenyik Z, Reidy JA, levels of perfluorinated chemicals and subfecundity.
Needham LL. Polyf luoroalkyl chemicals in Hum Reprod. 2009;24(5):1200–1205.
the U.S. population: Data from the National 142. Whitworth KW, Haug LS, Baird DD et al.
Health and Nutrition Examination Survey Perfluorinated compounds and subfecundity
(NHANES) 2003–2004 and comparisons with in pregnant women. Epidemiology. 2012;23(2):​
NHANES 1999–2000. Environ Health Perspect. 257–263.
2007;115(11):1596–1602. 143. Vested A, Ramlau-Hansen CH, Olsen SF et al.
130. Gewurtz SB, Backus SM, De Silva AO et al. Associations of in utero exposure to perfluorinated
Perfluoroalkyl acids in the Canadian environment: alkyl acids with human semen quality and reproduc-
Multi-media assessment of current status and trends. tive hormones in adult men. Environ Health Perspect.
Environ Int. 2013;59:183–200. 2013;121(4):453–455.
131. Gutzkow KB, Haug LS, Thomsen C, Sabaredzovic 144. Vanden Heuvel JP, Kuslikis BI, Peterson RE.
A, Becher G, Brunborg G. Placental transfer of per- Covalent binding of perfluorinated fatty acids to pro-
fluorinated compounds is selective—A Norwegian teins in the plasma, liver and testes of rats. Chem Biol
Mother and Child sub-cohort study. Int J Hyg Interact. 1992;82(3):317–328.
Environ Health. 2012;215(2):216–219. 145. Vanden Heuvel JP, Davis JW, Sommers R, Peterson
132. Inoue K, Okada F, Ito R et al. Perfluorooctane sulfo- RE. Renal excretion of perfluorooctanoic acid in
nate (PFOS) and related perfluorinated compounds male rats: Inhibitory effect of testosterone. J Biochem
in human maternal and cord blood samples: Toxicol. 1992;7(1):31–36.
Assessment of PFOS exposure in a susceptible popu- 146. Mehrotra K, Morgenstern R, Lundqvist G, Becedas
lation during pregnancy. Environ Health Perspect. L, Bengtsson AM, Georgellis A. Effects of peroxi-
2004;112(11):1204–1207. some proliferators and/or hypothyroidism on
133. Lee YJ, Kim MK, Bae J, Yang JH. Concentrations xenobiotic-metabolizing enzymes in rat testis. Chem
of perfluoroalkyl compounds in maternal and Biol Interact. 1997;104(2–3):131–145.
umbilical cord sera and birth outcomes in Korea. 147. Mehrotra K, Morgenstern R, Ahlberg MB, Georgellis
Chemosphere. 2013;90(5):1603–1609. A. Hypophysectomy and/or peroxisome prolifera-
134. Midasch O, Drexler H, Hart N, Beckmann MW, tors strongly influence the levels of phase II xenobi-
Angerer J. Transplacental exposure of neonates to otic metabolizing enzymes in rat testis. Chem Biol
perfluorooctanesulfonate and perfluorooctanoate: Interact. 1999;122(2):73–87.
A pilot study. Int Arch Occup Environ Health. 2007;​ 148. Shi Z, Ding L, Zhang H, Feng Y, Xu M, Dai J.
80(7):643–648. Chronic exposure to perfluorododecanoic acid
135. Monroy R, Morrison K, Teo K et al. Serum levels disrupts testicular steroidogenesis and the expres-
of perfluoroalkyl compounds in human maternal sion of related genes in male rats. Toxicol Lett.
and umbilical cord blood samples. Environ Res. 2009;188(3):192–200.
2008;108(1):56–62. 149. Zhao B, Hu GX, Chu Y et al. Inhibition of human
136. Javins B, Hobbs G, Ducatman AM, Pilkerton C, and rat 3beta-hydroxysteroid dehydrogenase and
Tacker D, Knox SS. Circulating maternal per- 17beta-hydroxysteroid dehydrogenase 3 activities by
fluoroalkyl substances during pregnancy in the perfluoroalkylated substances. Chem Biol Interact.
C8 Health Study. Environ Sci Technol. 2013;47(3):​ 2010;188(1):38–43.
1606–1613. 150. Zhang H, Lu Y, Luo B, Yan S, Guo X, Dai J. Proteomic
137. Brantsaeter AL, Whitworth KW, Ydersbond TA et al. analysis of mouse testis reveals perfluorooctanoic
Determinants of plasma concentrations of perfluo- acid-induced reproductive dysfunction via direct
roalkyl substances in pregnant Norwegian women. disturbance of testicular steroidogenic machinery.
Environ Int. 2013;54:74–84. J Proteome Res. 2014;13(7):3370–3385.
166  Effects of chemical pollutants on spermatogenesis and implications in male infertility

151. Kraugerud M, Zimmer KE, Ropstad E, Verhaegen S. 165. Llopis J, Westin S, Ricote M et al. Ligand-
Perfluorinated compounds differentially affect ste- dependent interactions of coactivators steroid
roidogenesis and viability in the human adrenocor- receptor coactivator-1 and peroxisome proliferator-
tical carcinoma (H295R) in vitro cell assay. Toxicol activated receptor binding protein with nuclear hor-
Lett. 2011;205(1):62–68. mone receptors can be imaged in live cells and are
152. Rosenmai AK, Nielsen FK, Pedersen M et al. required for transcription. Proc Natl Acad Sci U S A.
Fluorochemicals used in food packaging inhibit male 2000;97(8):4363–4368.
sex hormone synthesis. Toxicol Appl Pharmacol. 166. Schultz R, Yan W, Toppari J, Volkl A, Gustafsson
2013;266(1):132–142. JA, Pelto-Huikko M. Expression of peroxisome
153. Du G, Hu J, Huang H et al. Perfluorooctane sulfo- proliferator-activated receptor alpha messenger ribo-
nate (PFOS) affects hormone receptor activity, ste- nucleic acid and protein in human and rat testis.
roidogenesis, and expression of endocrine-related Endocrinology. 1999;140(7):2968–2975.
genes in vitro and in vivo. Environ Toxicol Chem. 167. Kowalewski MP, Dyson MT, Manna PR, Stocco DM.
2013;32(2):353–360. Involvement of peroxisome proliferator-activated
154. Qiu L, Zhang X, Zhang X et al. Sertoli cell is a poten- receptor gamma in gonadal steroidogenesis and
tial target for perfluorooctane sulfonate-induced steroidogenic acute regulatory protein expression.
reproductive dysfunction in male mice. Toxicol Sci. Reprod Fertil Dev. 2009;21(7):909–922.
2013;135(1):229–240. 168. Thomas K, Sung DY, Chen X et al. Developmental
155. Zhang J, Liang J, Zhu H, Li C, Wu Q. PFOS and PCB patterns of PPAR and RXR gene expression dur-
153 have direct adverse effects on neonatal testis ing spermatogenesis. Front Biosci (Elite Ed). 2011;​
modeled using a coculture of primary gonocyte and 3:1209–1220.
Sertoli cells. Environ Toxicol. 2013;28(6):322–331. 169. Regueira M, Riera MF, Galardo MN, Pellizzari EH,
156. Zhao B, Li L, Liu J et al. Exposure to perfluorooctane Cigorraga SB, Meroni SB. Activation of PPAR alpha
sulfonate in utero reduces testosterone production in and PPAR beta/delta regulates Sertoli cell metabo-
rat fetal Leydig cells. PLoS One. 2014;9(1):e78888. lism. Mol Cell Endocrinol. 2014;382(1):271–281.
157. Lopez-Doval S, Salgado R, Pereiro N, Moyano R, 170. Huyghe S, Schmalbruch H, De Gendt K et al.
Lafuente A. Perfluorooctane sulfonate effects on the Peroxisomal multifunctional protein 2 is essential
reproductive axis in adult male rats. Environ Res. for lipid homeostasis in Sertoli cells and male fertil-
2014;134C:158–168. ity in mice. Endocrinology. 2006;147(5):2228–2236.
158. Lou QQ, Zhang YF, Zhou Z et al. Effects of perfluo- 171. Abbott BD, Wolf CJ, Das KP et al. Developmental
rooctanesulfonate and perfluorobutanesulfonate on toxicity of perfluorooctane sulfonate (PFOS) is not
the growth and sexual development of Xenopus lae- dependent on expression of peroxisome proliferator
vis. Ecotoxicology. 2013;22(7):1133–1144. activated receptor-alpha (PPAR alpha) in the mouse.
159. Lau C, Thibodeaux JR, Hanson RG et al. Effects of Reprod Toxicol. 2009;27(3–4):258–265.
perfluorooctanoic acid exposure during pregnancy 172. Rosen MB, Schmid JR, Corton JC et al. Gene
in the mouse. Toxicol Sci. 2006;90(2):510–518. Expression profiling in wild-type and PPARalpha-
160. Wan HT, Zhao YG, Wong MH et al. Testicular signal- null mice exposed to perfluorooctane sulfonate
ing is the potential target of perfluorooctanesulfonate- reveals PPARalpha-independent effects. PPAR Res.
mediated subfertility in male mice. Biol Reprod. 2010;2010.
2011;84(5):1016–1023. 173. Kim HS, Jun KS, Sik HE, Seok KT, Hee KS, Young HS.
161. Desvergne B, Michalik L, Wahli W. Be fit or be sick: Induction of apoptosis and CYP4A1 expression in
Peroxisome proliferator-activated receptors are down Sprague-Dawley rats exposed to low doses of perfluo-
the road. Mol Endocrinol. 2004;18(6):1321–1332. rooctane sulfonate. J Toxicol Sci. 2011;36(2):201–210.
162. Maloney EK, Waxman DJ. trans-Activation of 174. Rosen MB, Schmid JE, Das KP, Wood CR, Zehr
PPARalpha and PPARgamma by structurally diverse RD, Lau C. Gene expression profiling in the liver
environmental chemicals. Toxicol Appl Pharmacol. and lung of perfluorooctane sulfonate-exposed
1999;161(2):209–218. mouse fetuses: Comparison to changes induced by
163. Shipley JM, Hurst CH, Tanaka SS et al. trans-activation exposure to perfluorooctanoic acid. Reprod Toxicol.
of PPARalpha and induction of PPARalpha target 2009;27(3–4):278–288.
genes by perfluorooctane-based chemicals. Toxicol 175. Lee YY, Wong CK, Oger C, Durand T, Galano JM,
Sci. 2004;80(1):151–160. Lee JC. Prenatal exposure to the contaminant perflu-
164. Vanden Heuvel JP, Thompson JT, Frame SR, Gillies orooctane sulfonate elevates lipid peroxidation dur-
PJ. Differential activation of nuclear receptors by ing mouse fetal development but not in the pregnant
perfluorinated fatty acid analogs and natural fatty dam. Free Radic Res. 2015;49(8):1015–1025.
acids: A comparison of human, mouse, and rat per- 176. Kristensen DM, Skalkam ML, Audouze K et al.
oxisome proliferator-activated receptor-alpha, -beta, Many putative endocrine disruptors inhibit pros-
and -gamma, liver X receptor-beta, and retinoid X taglandin synthesis. Environ Health Perspect. 2011
receptor-alpha. Toxicol Sci. 2006;92(2):476–489. Apr;119(4):534–541.
References 167

177. Abayasekara DR, Band AM, Cooke BA. Evidence for 185. Lopez-Ruiz MP, Choi MS, Rose MP, West AP, Cooke
the involvement of phospholipase A2 in the regula- BA. Direct effect of arachidonic acid on protein
tion of luteinizing hormone-stimulated steroidogen- kinase C and LH-stimulated steroidogenesis in rat
esis in rat testis Leydig cells. Mol Cell Endocrinol. Leydig cells; Evidence for tonic inhibitory control of
1990;70(2):147–153. steroidogenesis by protein kinase C. Endocrinology.
178. Adams IR, McLaren A. Sexually dimorphic devel- 1992;130(3):1122–1130.
opment of mouse primordial germ cells: Switching 186. Maloberti P, Cornejo MF, Castillo AF et al.
from oogenesis to spermatogenesis. Development. Enzymes involved in arachidonic acid release in
2002;129(5):1155–1164. adrenal and Leydig cells. Mol Cell Endocrinol.
179. Amateau SK, McCarthy MM. Induction of PGE2 by 2007;265–266:113–120.
estradiol mediates developmental masculinization of 187. Solano AR, Dada L, Podesta EJ. Lipoxygenase
sex behavior. Nat Neurosci. 2004;7(6):643–650. products as common intermediates in cyclic AMP-
180. Didolkar AK, Sundaram K. Arachidonic acid is dependent and -independent adrenal steroidogenesis
involved in the regulation of hCG induced steroido- in rats. J Mol Endocrinol. 1988;1(2):147–154.
genesis in rat Leydig cells. Life Sci. 1987;41(4):471–477. 188. Fang X, Dillon JS, Hu S et al. 20-carboxy-arachidonic
181. Dix CJ, Habberfield AD, Sullivan MH, Cooke BA. acid is a dual activator of peroxisome proliferator-
Inhibition of steroid production in Leydig cells by non- activated receptors alpha and gamma. Prostaglandins
steroidal anti-inflammatory and related compounds: Other Lipid Mediat. 2007;82(1–4):175–184.
Evidence for the involvement of lipoxygenase products 189. Huang JT, Welch JS, Ricote M et al. Interleukin-
in steroidogenesis. Biochem J. 1984;219(2):529–537. 4-dependent production of PPAR-gamma ligands
182. Gupta C, Goldman AS. The arachidonic acid cascade in macrophages by 12/15-lipoxygenase. Nature.
is involved in the masculinizing action of testoster- 1999;400(6742):378–382.
one on embryonic external genitalia in mice. Proc 190. Wan HT, Mruk DD, Wong CK, Cheng CY.
Natl Acad Sci U S A. 1986;83(12):4346–4349. Perfluorooctanesulfonate (PFOS) perturbs male rat
183. Gupta C. The role of prostaglandins in masculine Sertoli cell blood-testis barrier function by affecting
differentiation: Modulation of prostaglandin levels F-actin organization via p-FAK-Tyr(407): An in vitro
in the differentiating genital tract of the fetal mouse. study. Endocrinology. 2014;155(1):249–262.
Endocrinology. 1989;124(1):129–133. 191. Li N, Mruk DD, Chen H, Wong CK, Lee WM, Cheng
184. Kojima I, Kojima K, Rasmussen H. Possible role CY. Rescue of perfluorooctanesulfonate (PFOS)-
of phospholipase A2 action and arachidonic acid mediated Sertoli cell injury by overexpression of
metabolism in angiotensin II-mediated aldosterone gap junction protein connexin 43. Sci Rep. 2016; Jul
secretion. Endocrinology. 1985;117(3):1057–1066. 20;6:29667.
Advances in our understanding
of human spermatogenesis
13
QING WEN, ELIZABETH I. TANG, TITO JESUS, BRUNO SILVESTRINI,
and C. YAN CHENG

INTRODUCTION men after puberty at ~35–45 days of age in mice and rats
In the mammalian testis, spermatogenesis takes place in versus ~12 years of age in humans to produce 30–50 mil-
the testis. Spermatogenesis is divided into three distinctive lion versus 200 million functional sperm daily in rodents
cellular events. First, during the premeiotic steps, sper- versus humans in order to fertilize the egg. In humans,
matogonial stem cells, known as Asingle (As) in rodents,1,2 rats, and mice, spermatogenesis takes 64–68, 48–58, and
which are capable of undergoing self-renewal, enter mitosis 34–35 days, respectively, to complete.9,12,14,15 During the
and transform to Apaired (Apr) and then Aaligned (Aal) undif- past two decades, advances have been made in under-
ferentiated spermatogonia (Figure 13.1). Apr and Aal in standing the molecular mechanisms that regulate these
rodents that are not synchronized with the seminiferous events. In this chapter, we focus our discussion on stud-
epithelial cycle are further expanded to spermatogonial ies in humans except for specific topics where few reports
clones of differentiated spermatogonial A1, A2, A3, and A4, focused on human studies are found in the literature, we
which are now synchronized with the epithelial cycle.3,4 turn our discussion to rodents. The goal is to provide a
Finally, these type A differentiated spermatogonia divide brief overview of a few specific areas of research that might
and transform to intermediate spermatogonia and type B interest investigators so that more emphasis can be placed
spermatogonia, which are the interconnected cohorts of on studies that bridge the gap between studies in rodents
spermatogonia that are then transformed to spermato- and humans.
cytes (Figure 13.1). In humans, Adark spermatogonia in
men, considered to be human testicular stem cells and the GERM CELL DEVELOPMENT
regenerative reserve5 capable of undergoing self-renewal,
From undifferentiated spermatogonia to haploid
transform to Apale undifferentiated spermatogonia, which
spermatids
are the male germline progenitor and the functional
reserve.5 An undifferentiated Apale spermatogonium in In humans, a small group of cells with a unique morphol-
humans undergoes one mitotic division and transforms ogy designated human primordial germ cells (hPGCs)
to two differentiated type B spermatogonia, which in turn (also known as prespermatogonia, which are the gamete
undergo another mitotic division to form four spermato- founder cells in the male) is derived from extraembryonic
cytes.1 It is noted that all the spermatogonia are residing tissues surrounding the yolk sac in week 4 embryos.16,17
outside the blood-testis barrier (BTB) in the basal com- Male gonadal development begins at ~6 weeks postcon-
partment or at the stem cell niche (for spermatogonial ception when Sertoli cells make their first appearance,18
stem cells).2 Type B spermatogonia transform to prelep- and these changes are initiated and governed by the testis-
totene spermatocytes and which the germ cells that are determining genes Sry and Sox9.19,20 hPGCs are known
transported across the BTB to become leptotene, zygotene, to express key pluripotency markers such as POU5F1,
and pachytene spermatocytes in the adluminal compart- NANGO, and SSEA1 and to proliferate until the first tri-
ment. Second, the most prominent series of cellular events mester (weeks 10–12).21 It is noted that hPGCs migrate to
is meiosis, during which one spermatocyte undergoes the gonadal ridge and differentiate into gonocytes, and
meiosis I to form two secondary spermatocytes, and each together with the Sertoli cells they begin to form the semi-
of these undergoes meiosis II to form two haploid sperma- niferous or testis cords, the precursors of the seminifer-
tids. Third, once haploid spermatids arise, they undergo ous tubules. In short, during the second trimester (weeks
rapid morphological changes via spermiogenesis so that 13–28), most of hPGCs progressively become mitotically
elongated spermatids via 16 and 19 steps in mice and inactive (or mitotic arrest) and differentiate into prosper-
rats6–8 versus six steps in humans9 eventually transform to matogonia, and some of these prospermatogonia trans-
spermatozoa, which will be released into the tubule lumen locate to the basal region of the tubules and differentiate
during spermiation to complete spermatogenesis, entering into spermatogonia, forming type A spermatogonia dur-
the epididymis1,10–13 for further development. In theory, a ing the third trimester (weeks 28–40), and some type B
single As in mice or rats can become 1024 spermatocytes spermatogonia are found by birth.21 During embryonic
and thus 4096 haploid spermatids versus 16 haploid sper- development, peritubular myoid cells originating from
matids from one Apale spermatogonium (Figure 13.1). the mesonephros also migrate to the site under the influ-
These series of events take place nonstop in rodents and ence of Sry, which is necessary to assemble functional

168
(a) Rodent Testis
in vivo BTB

+ =

PGC Gonocyte As Apr Aal A1 A2 A3 A4 In B Spc sS rS ES Oocyte


(1) (2) (4-8-16) (16) (32) (64) (128) (256) (512) (1024) (2048) (4096) (4096)

Migration Quiescence Mitosis Differentiation Mitosis Meiosis I Meiosis II Spermiogenesis Fertilization

piRNAs

tsRNAs
miRNAs
(Epididymis maturation)

(b) Human Testis (in vivo)


in vivo BTB

+ =

PGC Gonocyte Adark Apale B Spc sS rS ES Oocyte


(1) (1) (2) (4) (8) (16) (32)
Spermatogonial stem cell
Migration Quiescence Differentiation Mitosis Meiosis I Meiosis II Spermiogenesis Fertilization Undifferentiated
spermatogonium
piRNAs level? miRNAs level? tsRNAs level?

Figure 13.1  A schematic illustration of spermatogenesis in rodents (a) and humans (b). The numeral bracketed below each germ cell type (see below) denotes the number of daughter cells
derived from an earlier progenitor cell via either mitosis or meiosis. It is noted that meiosis takes place behind the blood-testis barrier (BTB) in both rodents and humans. Theoretically, 4096 elon-
gated spermatids (ES) (i.e., spermatozoa) are formed from a single Asingle spermatogonium in rodents (versus 32 ES and sperm are formed from a single Adark or Apale spermatogonium in humans).
However, more than 75% of germ cells, most notably spermatocytes and spermatids, undergo apoptosis and/or degeneration to avoid overwhelming the population of Sertoli cells in the testis
since, unlike germ cells, Sertoli cells in adult testes by day ~17-19 in the rat testis (or after puberty in humans after age ~12-year-old) are differentiated cells [44,146]. Thus, the number of spermatozoa
derived from a single spermatogonium can be considerably less than the theoretical number. The BTB divides the seminiferous epithelium into the basal and apical (adluminal) compartment in
rodent and human testes. Spermatogonial mitotic proliferation and differentiation take place in the basal compartment. Type B spermatogonia differentiate into preleptotene spermatocytes (at
stage VIII and III of the epithelial cycle in rat and human testes, respectively) which are the germ cells connected in clones via intercellular bridges to be transported across the BTB at stage VIII or
stage III of the epithelial in rat and human testes, respectively) so that meiosis I/II and spermiogenesis take place behind the BTB in the apical compartment. Post-meiotic spermatid development
through spermiogenesis also takes place behind the BTB until the release of fully developed spermatids (i.e., spermatozoa) at spermiation to empty sperm into the tubule lumen. Subsequent
sperm maturation takes place in the epididymis so that fertilization can take place in the fallopian tube in the female to give rise of new offspring. Studies have also shown the involvement of
small no-coding regulatory RNAs (ncRNAs) such as piRNA (piwi-interacting RNA), miRNA (microRNA) and tsRNA (tRNA-derived small RNA) that regulate different stages of the spermatogenesis in
rodents [144], however, their involvement in regulatory human spermatogenesis remains largely unknown. Abbreviations: In rodents: A, type A spermatogonium; In, intermediate spermatogo-
nium; B, type B spermatogonium; Spc, spermatocyte; sS, secondary spermatocyte; rS, round spermatid (step 1-7); ES, elongating/elongated spermatid; As, Asingle spermatogonium, Apr, Apaired sper-
Germ cell development  169

matogonium, Aal, Aaligned spermatogonium in rodents are undifferentiated spermatogonia; A1-A4 are differentiated spermatogonia. In humans: Adark and Apale are undifferentiated spermatogonia.
170  Advances in our understanding of human spermatogenesis

testis cords.22 However, factors and/or genes that provide Studies in rodents have shown that the decision for undif-
the necessary signaling functions to regulate these events ferentiated spermatogonia to take the route of self-renewal
remain largely unknown. For instance, recent studies in via mitosis, differentiate to type B, or become apoptotic is
the mouse have shown that this is regulated, at least in regulated by Sertoli cell- and peritubular myoid-­produced
part, by the Sertoli cell Wt1 gene,23 yet the involvement of biological factors, cytokines, paracrine factors, and/or
Wt1 in human testis assembly remains unknown. At the hormones as well as Leydig cell-produced testosterone and
same time, outside these testis cords, Leydig cells differ- possibly microRNAs (miRNAs).38–41 For instance, GDNF
entiate12 along with rapid proliferation of germ cells and is involved in spermatogonial self-renewal, whereas stem
Leydig cells 24,25 to develop into the testis. cell factor (SCF), colony stimulating factor 1 (CSF-1),
The somatic cells of the newly formed testis begin to bone morphogenetic protein 4 (BMP4), retinoic acid (RA),
secrete anti-Mullerian hormone26 and testosterone,27 and the Notch1/Jagged2 signaling system are involved
which are necessary to confer male differentiation of the in differentiation, and miRNAs are involved in deter-
secondary sex organs. Also, primordial germ cells and mining spermatogonia cell fate.40 The number of germ
Sertoli cells within the testis cords and Leydig cells found cells in the testis is also determined by the population of
outside the cords rapidly proliferate via mitotic divisions Sertoli cells10,42,43 that become differentiated in humans by
without embarking on meiosis as seen in the fetal ovary.18 puberty at ~10–13 years of age.44 Thus, many of the devel-
For instance, the number of germ cells per human testis oping germ cells, including differentiated spermatogonia,
at week 6 postconception is ~3000 which rises to ~30,000 spermatocytes, and spermatids, during spermatogenesis
by week 9, with a germ:Sertoli cell ratio of ~1:11 versus a elect, being programmed or mandated (or instructed)
germ:somatic cell (including Sertoli, Leydig, peritubular by Sertoli cells, to go through the apoptotic pathway to
myoid, and mesenchymal cell) ratio of ~1:44 at 6- versus keep the proper ratio of Sertoli:germ cells at ~1:30–1:50 in
9-week postconception, respectively.24 Studies in rodents rodents.45 As such, as much as ~75% of germ cells undergo
have shown that a number of paracrine factors are crucial apoptosis46 to avoid overwhelming the fixed population of
to confer migration, differentiation, and proliferation of differentiated Sertoli cells in the testis following puberty in
primordial germ cells and Sertoli cells during testis devel- humans at ~12–13 years of age versus ~17 days postpartum
opment. These include members of the TGFß family (e.g., (dpp) in rodents. Since Sertoli cells are the only somatic
TGF-ßs, activins, and inhibins),28,29 nerve growth factor cells in the seminiferous epithelium to support germ cell
(NGF) family (e.g., NGF, brain-derived neurotrophic fac- development, germ cells, in particular the highly differen-
tor [BDNF], neurotrophin [NT] 3, NT4 [also known as tiated postmeiotic spermatids, rely almost exclusively on
NT5], and NT6, and pituitary adenylate cyclase-activating Sertoli cells for the provision of nutrients, cytokines, and
polypeptide [PACAP]).30–32 However, the roles of these paracrine factors and also structural support, in particu-
factors in human testis development also remain to be lar their transport across the seminiferous epithelium to
investigated. coordinate with the seminiferous epithelial cycle. Thus,
In this context, it is of interest to note that epigen- fully developed spermatids (i.e., spermatozoa) line up at
etic reprogramming, in particular genomic-wide DNA the adluminal edge of the apical compartment near the
demethylation in hPGCs, is somewhat similar to mouse tubule lumen in early stage VIII tubules in rodents versus
primordial germ cells.33 For instance, hPGCs undergo stage II in humans to prepare for their release at spermia-
extensive demethylation at week 5 at the time of their colo- tion in the corresponding late stage VIII versus II. Since
nization of embryonic gonads in humans, which continue it is known that developing spermatids are metabolically
thereafter.33 The mechanism for epigenetic reprogram- quiescent cells lacking the ultrastructures of lamellipodia
ming in human and mouse germ cells appear to be well and filopodia to initiate cell movement, they rely solely on
conserved between species.34,35 the Sertoli cell to support their transport across the semi-
In human testes, spermatogonia undergo self-renewal niferous epithelium during spermiogenesis.7,47
via mitosis, and they are classified into three subtypes based It is noted that all spermatogonia reside in the basal
on their heterochromatin content known as Adark, Apale, and compartment of the seminiferous epithelium. Type B
B spermatogonia.36,37 At present, Adark spermatogonia are spermatogonia transform into preleptotene spermato-
considered to be the true testicular stem cells (also known cytes, marking the end of the mitotic proliferation phase
as quiescent stem cells) or the regenerative reserve, which of spermatogenesis. Furthermore, preleptotene sper-
are characterized by their low mitotic activity and possibly matocytes are the only primary spermatocytes to be
involved in preserving the genome integrity in the germ transported across the BTB, which physically divides the
line in humans.1,5 However, Apale spermatogonia are con- seminiferous epithelium into the basal and the adluminal
sidered to be the male germline progenitor (or the active (apical) compartment.7 According to stages of prophase 1,
stem cells), the functional reserve, which are capable of primary spermatocytes in humans and rodents are classi-
undergoing mitosis through cyclic proliferation to main- fied into preleptotene, leptotene, zyogtene, and pachytene
tain its population. But some Apale cells undergo mitosis so spermatocytes, which eventually transform into diplotene
that each Apale spermatogonial cell divides and transforms spermatocytes to begin meiosis I.12 In men and rodents,
to two type B spermatogonia to meet the need of produc- DNA recombination that is being used to exchange genetic
ing 200 million sperm daily from an adult human male.1,5 material between homologous chromosomes is mediated
Development of round spermatids to functional spermatozoa through spermiogenesis  171

by the synaptonemal complex, which takes place at the which is known to play an important role in maintain-
pachytene stage of spermatocytes with strict checkpoints ing the BTB integrity in rodents,65 may not be essential to
from metaphase 1 through anaphase 1 during meiosis. the BTB homeostasis in humans since BTB integrity was
In men with nonobstructive azoospermia (NOA), a type not compromised in healthy men following suppression
of idiophatic infertility, about 10% of these infertile men of spermatogenesis treated with testosterone or a combi-
display a higher recombination failure rate versus normal nation of testosterone and levonorgestrel (LNG), and the
fertile men, and they are also at risk of producing chromo- claudin-3 level was not down-regulated.66 A recent study
somally compromised offspring.48,49 used testis biopsies from healthy men treated with a testos-
The rationale for technological advance and a better terone plus LNG,67 similar to the earlier study,66 but also
understanding of obtaining haploid spermatids from sper- with a GnRH antagonist (acyline) or 5α-reductase inhibi-
matogonia is to assist men with NOA to father their own tor (dutasteride) that provided a ~25%–40% added sup-
children. Recent studies in rodents using 3-dimensional pression of spermatogenesis versus testosterone and LNG
(3-D) in vitro organ cultures with fragments of neonatal alone,68,69 for BTB integrity assessment. It was found that
mouse testes embedded in soft agarose gel in a gas-liquid the BTB in these men with spermatogenesis suppression
interphase method have generated some excitement in the was compromised (i.e., the BTB was “leaky”), with exten-
field. For instance, it was shown that using this in vitro cul- sive downregulation and mislocalzation of claudin-11,
ture system, functional haploid spermatids were obtained connexin 43, and vinculin at the BTB, but the distribu-
from undifferentiated mouse spermatogonia. However, tion of ZO-1 and ß-catenin was unaffected.67 Also, in men
the efficacy of this approach was exceedingly low, at about with primary seminiferous tubule failure, such as meiotic
<2%,50,51 thereby making this approach unsuitable for arrest that led to, Sertoli-cell-only syndrome, the spatial
widespread adoption for human studies. Interestingly, a localization of claudin-11 and connexin 43 at the BTB
recent report has provided important advances wherein was grossly disorganized, failing to localize at the BTB to
functional spermatids were efficiently produced from confer barrier integrity, thereby causing spermatogenesis
undifferentiated spermatogonia when cultured in vitro.52 dysfunction.70 Collectively, these findings illustrate that
This approach must now be tested in studies in humans. the human BTB is likely regulated analogous to rodents,
In fact, a recent report using human spermatogonial at least in part, but there are some substantial differences.
stem cells (SSCs) obtained from cryptorchid patients and Thus, studies using rodent testes as study models should
cultured in vitro were found to develop into functional be cautiously interpreted before applying the pertinent
haploid spermatids,53 illustrating the feasibility of this information to humans. Therefore, the use of an in vitro
approach. As such, it is likely that functional spermatids culture system involving human Sertoli cells may be an
could be developed from undifferentiated spermatogonia alternative approach to study human BTB function and
among infertile men, perhaps within the next decade, so its role in spermatogenesis before human biopsy samples
that men with nonobstructive azoospermia could father are used for additional analysis.71 The BTB is important
their own children. to spermatogenesis since its disruption, at least in rodents,
is known to cause infertility. For instance, environmen-
Blood-testis barrier (BTB) tal toxicants such as cadmium and glycerol that cause
In rodents and humans, the BTB is constituted by coex- irreversible BTB damage in rodents are known to cause
isting actin-based tight junction (TJ), basal ectoplasmic sterility.72–74 However, it remains to be established if some
specialization (ES, a testis-­specific actin-rich adherens infertile men with idiopathic infertility are associated
junction [AJ]), and gap junction, as well as intermediate with irreversible BTB disruption.
filament-based desmosome between adjacent Sertoli cells
near the basement membrane.14,54-56 However, in rodents,
DEVELOPMENT OF ROUND SPERMATIDS
the BTB is also considerably contributed by the peritubular
TO FUNCTIONAL SPERMATOZOA THROUGH
myoid cells in the tunica propria since administration of
SPERMIOGENESIS
electron-dense substances (e.g., colloidal carbon, thorium,
lanthanum) failed to penetrate the peritubular myoid cell Spermiogenesis
layer in ~85% of the tubules examined in rodent testes.57,58 In rodents and humans, primary spermatocytes at the
However, in primate testes, the peritubular myoid cells beginning of meiosis I are diploid but have 4n amount of
were shown to be less effective in blocking the penetration DNA, and secondary spermatocytes derived from meio-
of electron-dense markers across the tubules,59 implicat- sis I contain the haploid number of chromosomes but 2n
ing that the human BTB may share similar characteristics. DNA content. There is a brief interphase between meio-
More important, BTB integral membrane proteins such sis I and II without DNA synthesis. Almost immediately,
as occludin, which is the best-studied TJ protein in the secondary spermatocytes begin meiosis II, moving from
rodent testis, is not found in humans.60,61 Also, claudin-3, prophase through metaphase, anaphase, and telophase
an important BTB integral membrane protein in the to form round spermatids that are haploid and contain-
mouse testis62 and shown to be delicately involved in the ing 1n DNA content. In humans, studies have shown that
transport of preleptotene spermatocytes across the BTB,63 maturation arrest at this stage is rare.75,76 Once formed,
is absent in the rat testis.64 Interestingly, testosterone, round spermatids undergo a series of transformation and
172  Advances in our understanding of human spermatogenesis

development, forming elongating and elongated sperma- (e.g., Spem 1 [sperm maturation 1]). In this context, it is
tids via spermiogenesis. Spermatids are also divided into of interest to note that early stages of spermiogenesis are
different stages known as steps, with 16, 19, and 6 steps in monitored with various checkpoints, perhaps up to step
mice, rats, and humans, respectively.9,77 Due to the unique 8 spermatids in rodents. For instance, round sperma-
association of developing spermatids with Sertoli cells in tids with defects are usually removed from the epithe-
cross sections of the seminiferous epithelium through- lium, such as through the formation of multinucleated
out spermatogenesis, Leblond and Clermont78 were able spermatids—a common feature of abnormal spermio-
to divide the epithelium into different stages. This stag- genesis87 that leads to infertility. It appears that these
ing was aided in part by using the periodic acid-Schiff multinucleated spermatid cells are formed, at least in
(PAS) reaction to visualize changes in the Golgi region of part, as a result of defects in intercellular bridges, such
spermatids (to identify the steps of spermatids) with the as observed after colchicine treatment,88 which is known
development of the acrosome and known as the epithelial to induce microtubule disassembly.89 Also, deletion of
cycle.6–8,12,78,79 In mouse, rat, and human testes, there are genes important to early spermiogenesis, such as genes
12 (I–XII), 14 (I–XIV), and 6 (I–VI) stages of the cycle, and pertinent to acrosome biogenesis (e.g., Gopc), leads to
the duration of a complete seminiferous epithelial cycle is infertility.90 However, a similar quality control mecha-
8.6, 12.8, and 16 days, respectively.6,12,14 Since an undiffer- nism is absent in late spermiogenesis because deletion of
entiated spermatogonium (Asingle in rodents versus Adark in late spermatid-associated genes (e.g., Catsper1-4, Spem
humans) takes ~35, 53, and 64 days to develop into step 1) in mice fail to affect testis weights, and sperm counts
16, 19, and 12 spermatids in the testis of the mouse, rat, are normal and development of defective late spermatids
and human, respectively, it thus takes a spermatogonium continue to proceed to spermiation even though abnor-
to go through the cycle ~4.5 times to develop into func- mal sperms are found in these mice.86 At least spermatids
tional sperm. The development of round spermatids into from these mice could be used for ICSI to obtain success-
functional sperm is known as spermiogenesis, referring to ful pregnancy;however, the biology of spermiogenesis in
the transformation of a spherical spermatid to a sperm- humans remains largely unexplored.
like elongated spermatid known as spermatozoon. During
this process, no DNA synthesis or cell division takes place Spermiation
except for a series of morphological changes in the cyto-
At the end of spermiogenesis, fully developed spermatids,
sol and the nucleus, such as condensation of the genetic
namely spermatozoa, appear and they line up at the edge
material and their transport to the spermatid head overly-
of the tubule lumen to prepare for their release at sper-
ing with the acrosome, and coupled with the elongation
miation, which takes place at stage VIII of the epithelial
of the tail.77,80,81 As such, the genetic material of sperma-
cycle in rodent testes versus stage II in human testes.6,7,9,47
tids is considered to be transcriptionally inactive except
Interestingly, the biology of spermiation in humans, in
that spermatids, similar to other germ cells, contain con-
particular the underlying regulatory mechanism(s), is vir-
siderable amounts of small miRNAs and siRNA82,83 that
tually unknown and much of the findings in the literature
are capable of regulating Sertoli cell function, possibly
are based on studies using the rodent testis.87,91,92 As such,
through gap junctions or intercellular bridges. This pos-
the following discussion is based on studies in rodents
sibility, however, has not been investigated. During sper-
unless otherwise specified. Spermiation is a highly com-
miogenesis, defects in acrosome assembly in human testes
plicated cellular process, consisting of at least three sepa-
leads to globozoospermia, a rare (incidence <0.1% among
rate series of cellular events.
human males) but severe disorder of male infertility84 in
which spermatozoa lack acrosomes, which is difficult to 1. Active spermatid transport and endocytic vesicle-­mediated
manage therapeutically except with the use of intracyto- protein trafficking event. First, spermiation begins with
plasmic sperm injection (ICSI) to ensure successful preg- the active transport of spermatids across the adluminal
nancy.85 Table 13.1 lists some defects in fertility associated compartment toward the luminal edge of the tubule
with mutations of specific genes that regulate spermato- lumen starting at stage VII in rodents (versus stage I
genesis in men. in humans), and by about mid-stage VIII (versus early
Studies using genetic models in mice have identi- stage II in humans), elongated spermatids have lined
fied a number of genes associated with different cellular up at the luminal edge of the tubule lumen. During
events during spermiogenesis.86 These include acrosome this series of events, apical ES, the only anchoring
biogenesis (e.g., Gopc [Golgi-associated PDZ and coiled- device that anchors spermatids onto the Sertoli cell as
coil motif containing protein] and Gba2 [ß glucosidase it makes its initial appearance at stage VIII of the epi-
2]), flagella assembly (e.g., Tekt2 [Tektin 2], Tek4 [Tektin thelial cycle by replacing desmosome and gap junction,
4], Vdac3 [voltage-dependent anion channel 3]), energy undergo some remarkable changes. These changes are
metabolism for motility (e.g., Catsper1-4 [cation chan- first detected at the concave (ventral) side of the sper-
nel, sperm associated 1, 2, 3, and 4], Gapdhs [glyceral- matid head, where apical ES transforms into an ultra-
dehyde-3-phospahte dehydrogenase sperm-specific]), structure known as the apical tubulobulbar complex
nuclear condensation (e.g., Tnp1, [transition protein 1], (apical TBC) as named by some investigators.93–95 In
Tnp2, Prm1 [protamine 1] Prm2), and cytoplasm removal essence, the TBC represents endocytic vesicle-mediated
Development of round spermatids to functional spermatozoa through spermiogenesis  173

Table 13.1  Genetic anomalies or mutation associated with male infertility and associated phenotypes in men.
Genetic anomalies Phenotype
Chromosomic alterations, such as Klinefelter’s syndrome, Azoospermia, oligozoospermia, or
Robertsonian translocations, reciprocal translocations147–149 normozoospermia
Y chromosome microdeletions, such as deletion of AZFa, AZFb, Azoospermia, oligozoospermia, SCOS,
or AZFc 150,151 spermatogenic arrest, NOA or normozoospermia
Genetic Mutation
Nuclear receptor 5A1 (NR5A1), also known as steroidogenic Azoospermia or oligozoospermia
factor 1 (SF-1)152
Kallmann syndrome 1 sequence (KAL-1)153,154 Kallmann syndrome-hypogonadotropic
hypogonadism
Androgen receptor (AR)155 Azoospermia or oligozoospermia
CFTR (cystic fibrosis transmembrane conductance regulator)156 Obstructive azoospermia
INSL3- RXFP2 (insulin-like factor 3-its receptor, relaxin/insulin-like Cryptorchidism
family peptide receptor 2)157,158
CATSPER1 (Cation channel sperm-associated protein 1)159 Oligo-astheno-theratozoospermia
AURKC (aurora kinase C)160 Macrozoospermia
SPATA16 (spermatogenesis-associated 16)161 Globozoospermia
DPY19L2 [Dpy-19 like 2 (C. elegans)] 162,163 Globozoospermia
DNAI1 (dynein axonemal intermediate chain 1), DNAH5 (dynein Asthenozoospermia
axonemal heavy chain 5) and DNAH11 (dynein axonemal heavy
chain 11)164
SNRPA1* (Small nuclear ribonucleoprotein polypeptide A’)165 NOA
RHOX* (reproductive homeobox)166 Poor semen quality
DAZL* (deleted in azoospermia-like)167 Spermatogenic failure
MTHFR* (methylenetetrahydrofolate reductase) 168,169 Spermatogenic defects
KATNB1* (katanin regulatory subunit B1)129 Oligoasthenotheratozoospermia
RABL2A* (RAB, member of RAS oncogene family-like 2A)170 Oligoasthenozoospermia
SHBG* (sex hormone–binding globulin)171 Spermatogenic defects and poor semen quality
TAF7L* (TATA-box binding protein associated factor 7 like)172,173 Spermatogenic failure
USP26* (ubiquitin specific peptidase 26)174–177 Azoospermia
FSHR* (follicle stimulating hormone receptor) 178,179 Spermatogenic defects
ESR1* (estrogen receptor 1) and ESR2* (estrogen receptor 2)180,181 Oligozoospermia
ETV5* (ETS variant 5)182 NOA and SCOS
*Candidate genes correlated with male infertility; AZF: azoospermia factor; NOA: nonobstructive azoospermia; SCOS: Sertoli
cell–only syndrome.

intracellular trafficking organelles, similar to giant spermatids in the mouse and human testis) and grad-
endosomes, to elicit endosome-mediated recycling and ual degeneration of the apical ES. This event is tightly
also protein degradation87,96 (Figure 13.2). In fact, TBC associated with the condensation of the spermatid
is constituted by proteins found in endocytic vesicles, remaining cell cytosol and eventually shed as the resid-
including clathrin, cortactin, and other endosome ual body to be engulfed by the Sertoli cells and actively
markers.97–100 TBC is also found at the basal ES, is transported to the base of the tubule. Phagosomes are
associated with the BTB, and is known as basal TBC101 then fused with lysosomes to form phagolysosomes,
clearly visible under electron microscopy.102 In short, which are to be degraded and eliminated in the Sertoli
proteins at the apical ES are rapidly endocytosed and cell.12,103 As such, the Sertoli cell is also known as a
likely recycled to assemble the newly formed apical ES scavenger in the epithelium.
when step 8 spermatids appear at stage VIII of the epi- 3. Sperm release. At late stage VIII in rodents (versus
thelial cycle in the rat testis. stage II in humans), spermatozoa transformed from
2. Remodeling/degeneration of the apical ES and residual elongated spermatids are released into the tubule
body disposal. Second, elongated spermatids undergo lumen due to a complete degeneration of the apical ES.
extensive remodeling near or at the luminal edge, Studies have shown that apical ES degeneration that
including the formation of spermatozoa from step 19 facilitates the release of sperm in the rat testis is medi-
spermatids in the rat testis (versus step 16 and step 6 ated, at least in part, through the action of MMP2,104
174  Advances in our understanding of human spermatogenesis

which likely induces proteolysis of the laminin frag- elongating of the tail, and others, 86,112,113 the actin- and
ments at the apical ES, such as laminin-γ3 chain.105–107 microtubule (MT)-based cytoskeletons are also nota-
This, in turn, generates biologically active fragments bly involved. 8,87,92,114 (Figure 13.2). This notion was first
from laminin-γ3 and laminin-β3 chains.106 One of noted in studies using toxicant models in which rodents
these peptides, known as F5-peptide, was shown to be were treated with toxicants, such as 2,5-hexanedione
capable of inducing BTB restructuring106,108 so that the and carbendazim. It was shown that the Sertoli cell
events of sperm release and BTB remodeling that take cytoskeletons, in particular MT-based cytoskeleton,
place simultaneously across the epithelium at stage were grossly disrupted.114–116 This in turn affects sper-
VIII of the cycle can be coordinated.109,110 Studies have matid and luminal fluid transport, causing retention of
shown that the events pertinent to spermiation are spermatids in the seminiferous epithelium and/or fluid
regulated by p-FAK-Tyr407 and p-FAK-Tyr397 through accumulation in the tubule lumen. For instance, sper-
their effects on the actin-based cytoskeleton at the api- matids at or near the luminal edge are emptied into the
cal ES.108,111 tubule lumen, whereas spermatids inside the epithelium
fail to be transported to the luminal edge.117,118 This fail-
Regulation of spermatogenesis ure of spermatid transport, such as premature release of
Studies have shown that during spermatogenesis, in spermatids in nonstage VIII tubules and/or retention of
particular spermiogenesis and spermiation, besides the late spermatids in the epithelium, such as step 19 sper-
timely expression of specific genes necessary to regu- matids found in stages IX-XI tubules, is also found in
late spermatid transformation, packaging of the genetic rat testes following in vivo knockdown of genes known
material into the spermatid head, DNA condensation, to be involved in conferring cytoskeletal function, such

Spermiation

Late stage VII - early stage VIII Late stage VIII

Residual body (outside)


Spermatid cytoplasm Phagosome (inside)

Tubulobulbar complex (TBC) Fluid-phase endocytosis

Microtubule-based cytoskeleton Phagolysosome

Actin-based cytoskeleton Dissolution of phagosome

Figure 13.2  A schematic illustration of the developing spermatids and residual bodies that are transported across the seminifer-
ous epithelium during the epithelial cycle of spermatogenesis, and are supported by the actin- and microtubule (MT)-based cyto-
skeletons in adult rat testes. The left panel illustrates the cross section of a seminiferous tubule at stage VII-early VIII of the epithelial
cycle vs. a tubule at late stage VIII on the right panel when the release of sperm takes place. The transport of elongated spermatids
across the epithelium in a stage VII-VIII tubule is supported by the track-like structures of MTs and actin filaments so that sperm can
be lined up near the tubule lumen for their release at spermiation in late stage VIII of the cycle (right panel). Also, at stage VIII, phago-
somes derived from residual bodies engulfed by the Sertoli cell are being transported to the base of the tubule for their degradation
via the lysosomal degradation pathway.
Development of round spermatids to functional spermatozoa through spermiogenesis  175

as actin nucleation protein formin 1119 and actin bun- (e.g., connexin-43, connexin-33), and focal adhesion com-
dling protein plastin 3.120 plex (also known as focal contact) (e.g., ß1-­integrin).125,126
A disruption of the actin-based cytoskeleton thus com-
Role of actin- and MT-based cytoskeletons promises adhesion protein complexes at the apical ES,121
Rats treated with adjudin, 1-(2,4-dichlorobenzyl)-1H-in- thereby causing germ cell loss from the epithelium, leading
dazole-3-carbohydrazide were shown to induce extensive to infertility.
germ cell exfoliation, but in some tubules, spermatids
remained embedded deep inside the seminiferous epithe- Cytoskeletal reorganization
lium even though apical ES was compromised and degener- As noted above, cytoskeletons that provide the tracklike
ated.121 A recent report using the adjudin model has shown structures are crucial to the transport of spermatids, resid-
that the tracklike structures conferred by both actin- or ual bodies, and phagasomes across the epithelium during
MT- (visualized by α-tubulin or ß-tubulin staining, which the epithelial cycle. However, the actin- and MT-based
are the building blocks of MTs) based cytoskeletons are cytoskeletons also require dynamic changes to confer
truncated and grossly downregulated within 6 hours fol- plasticity to Sertoli cells so that the Sertoli cell can mod-
lowing adjudin treatment.122 In short, no distinctive track- ify its cell shape to accommodate changes in spermatid
like structures of actin- or MT-based cytoskeletons are morphology during spermiogenesis. As such, cleavage
found in the testis of adjudin-treated rats (50 mg/kg b.w. by proteins specific to the actin- and MT-cytoskeletons are
oral gavage) from 6 through 24 hours,122 and these changes necessary to confer cytoskeletal dynamics. Recent studies
are associated with extensive spermatid exfoliation, lead- have shown that mutation or inactivation/deletion of a MT
ing to transient infertility in these rats. More detailed cleavage protein, katanin, causes defects in spermiogen-
analysis of these rat testes revealed that the apical ES at esis, leading to infertility.127,128 In fact, genetic mutation of
the spermatid-Sertoli cell interface in spermatids located katanin gene in humans lead to oligoasthenotetratozoo-
near the tubule lumen and in spermatids embedded deep spermia in humans, an infertile condition manifested by
inside the epithelium were equally disrupted. However, low spermatozoa number, abnormal sperm morphology,
spermatids embedded inside the epithelium failed to be and poor motility.129 On the other hand, Sertoli-cell-
transported across the epithelium to be emptied into the specific deletion of N-WASP, an upstream activator of the
tubule lumen. The plausible explanation is due to the lack Arp2/3 complex, perturbs the ability of actin cytoskeleton
of “tracks” conferred by cytoskeletons, analogous to the to reorganize from its bundled to unbundled/branched
absence of subway tracks for transporting cargo via sub- configuration also shown to cause infertility in male
way cars. It was also shown that these unwanted changes in mice.130 A detailed analysis using these Sertoli cell-­specific
the actin- and MT-based cytoskeletons are due to changes N-WASP deleted mice has shown that the loss of the
in the spatiotemporal expression of Arp3 and Eps8122 such branched actin polymerization capability in Sertoli cells
that actin microfilaments at the apical ES fail to organize leads to spermiogenesis arrest, since meiosis can be seen in
into bundles to support apical ES function. Studies have some tubules, but none of the round spermatids undergo
shown that the dynamic conversion of actin microfila- spermiogenesis to advance into elongating spermatids.131
ments between their branched/unbundled and bundled Collectively, these findings illustrate the importance of
configuration at the apical ES are conferred by Arp3, a cytoskeletal reorganization to support spermiogenesis.
branched actin polymerization protein that causes nucle-
ation of an actin microfilament at its barbed ends, creat- Hormonal regulation
ing a branched actin network. On the other hand, Eps8, Studies have shown that intratesticular testosterone (T)
an actin barbed end capping and bundling protein that level is maintained at ~100-fold higher than the systemic
causes microfilaments to organize into bundles.123 Thus, circulation in both rodents and humans to support sper-
the tracklike structures conferred by F-actin were grossly matogenesis.132–134 Thus, T implants are used to perturb
disrupted following adjudin treatment due to disruptive the hypothalamic-pituitary-testicular axis in rodents by
spatial expression of these actin regulatory proteins. For increasing the serum T level, which in turn shuts down
MT conferred tracklike structures, it was shown that the the release of GnRH from the hypothalamus. This thus
spatiotemporal expression of EB1 (end-binding protein 1, a reduces LH secretion from the pituitary gland, lowering
plus (+) -end microtubule tracking protein [+TIP]) was also the intratesticular T level due to reduced T production
disrupted following adjudin treatment, so that the tracklike by Leydig cells. These changes thus contribute to exfolia-
structures conferred by MTs were grossly disrupted follow- tion of germ cells from the epithelium.135,136 An elevated
ing adjudin treatment.122 In this context, it is important to intratesticular T level is also crucial to m
­ aintain spermio-
note that cytoskeletons, in particular F-actin, also provide genesis.137 Studies have also illustrated the significance of
the attachment site for adhesion protein complexes, such estrogen in male reproductive function since earlier stud-
as α6ß1-integrin, JAM-C, JAM-A, nectin-2, nectin-3, and ies demonstrated the role of estrogen in fluid reabsorption
others at the apical ES.54,124 In this context, it is of interest to of luminal fluid in efferent ducts in the rodent testes.138,139
note that apical ES is a hybrid adherens junction (AJ) since Interestingly, by deleting the Cyp19 gene encoding P450arom
it is constituted by proteins that are usually found at the TJ was found to perturb estrogen synthesis capability in
(e.g., JAM-A, JAM-C), AJ (e.g., N-cadherin), gap junction these mice without perturbing the levels of gonadotropins
176  Advances in our understanding of human spermatogenesis

or androgens in the systemic circulation and in the tes- mutations of many genes that are crucial to maintain sper-
tis. Also, Cyp19 KO mice were found subfertile, having a matogenesis in humans (Table 13.1). As noted in various
reduced number of round and elongated spermatids by sections discussed above, many questions remain to be
the time they reached ~1-year of age.140 Thus, it is likely answered. However, the next decade is likely to be very
that the relative ratio of androgen:estrogen in the micro- promising in terms of providing new tools and techniques
environment of the testis is crucial to support spermio- to manage some male infertility, such as helping men with
genesis and/or spermiation. These findings will need to be unobstructed azoospermia to father their own children.
better examined in humans to explore the role of T and
estradiol-17ß on human spermiogenesis and spermiation. ACKNOWLEDGMENTS
The authors have no conflicts of interest to declare. This
Genetic regulation work was supported by grants from the National Institutes
Studies using genetic models have shown that more of Health, NICHD R01 HD056034 to C.Y.C., and U54
than 400 genes are essential for male fertility, involving HD029990 Project 5 to C.Y.C.
­k nockout/knockin/gene-trapped, transgenic, and chem-
ical-mediated point mutant mice.112 In humans, genetic REFERENCES
factors also play a crucial role in maintaining fertility in 1. Ehmcke J, Schlatt S. A revised model for spermato-
men since a number of reproductive disorders in men are gonial expansion in man: Lessons from non-human
now known to be the result of genetic anomalies and muta- primates. Reproduction. 2006;132;673–680.
tions (or genetic variations) of specific genes113,141; some of 2. de Rooij DG. The spermatogonial stem cell niche.
these findings are listed in Table 13.1. Furthermore, recent Microsc Res Tech. 2009;72:580–585.
studies have shown that abnormal expression of miRNAs 3. de Rooij DG, Russell LD. All you wanted to know
in human testes also leads to NOA. These include upreg- about spermatogonia but were afraid to ask. J Androl.
ulation of miR-302a, miR-491-3p, miR-141, miR-429, and 2000:21:776–798.
miR-7-1-3p, and also downregulation of miR-520d-3p and 4. Dettin L, Ravindranath N, Hofmann MC, Dym M.
miR-383 in men with NOA.142,143 These findings thus illus- Morphological characterization of the spermato-
trate the complexity of regulation pertinent to spermato- gonial subtypes in the neonatal mouse testis. Biol
genesis. Future studies should focus on identifying the Reprod. 2003;69:1565–1571.
common signaling and/or regulatory pathway(s) utilized 5. Ehmcke J, Wistuba J, Schlatt S. Spermatogonial stem
by multiple sets of genes to regulate specific cellular events cells: Questions, models and perspectives. Hum
during spermatogenesis. In this context, it is of interest to Reprod Update. 2006;12:275–282.
note that certain paternally acquired traits are known to 6. Hess RA, de Franca LR. Spermatogenesis and cycle
be memorized or impregnated in germ cells as epigenetic of the seminiferous epithelium. Adv Exp Med Biol.
information through RNAs and/or sperm RNA modifica- 2008;636:1–15.
tions.144 In short, epigenetic inheritance can be acquired 7. Xiao X, Mruk DD, Wong CKC, Cheng CY. Germ cell
through sperm, and studies have shown that this is medi- transport across the seminiferous epithelium during
ated mainly through sperm RNAs and RNA modifica- spermatogenesis. Physiology. 2014;29(4):286–298.
tions.144 These findings also illustrate that much research 8. Tang EI, Mruk DD, Cheng CY. Regulation of micro-
is needed to better understand the genetic material of hap- tubule (MT)-based cytoskeleton in the seminiferous
loid spermatids. epithelium during spermatogenesis. Semin Cell Dev
Biol. 2016;59:35–45.
CONCLUDING REMARKS AND FUTURE PERSPECTIVES 9. Amann RP. The cycle of the seminiferous epithe-
We briefly review advances in the field regarding molecu- lium in humans: A need to revisit? J Androl. 2008;​
lar mechanisms that regulate some aspects of spermato- 29:469–487.
genesis in selected areas that have implications to human 10. Sharpe RM. Regulation of spermatogenesis. In
males. Nonetheless, studies in understanding the biol- Knobil E, Neill JD, eds. The Physiology of Reproduction.
ogy of spermatogenesis in humans are far behind stud- New York: Raven Press; 1994:​1363–1434.
ies in rodents. One of the major obstacles is the lack 11. Neto FTL, Back PV, Najari B, Li PS, Goldstein M.
of human samples for analysis and functional studies. Spermatogenesis in humans and its affecting factors.
However, some recent advances in the field, such as the Semin Cell Dev Biol. 2016;59:10–26.
use of human Sertoli cells that are capable of undergoing 12. de Kretser DM, Kerr JB. The cytology of the testis. In
mitotic proliferation when cultured in medium contain- Knobil E, Neill JB, Ewing LL, Greenwald GS, Markert
ing ~5%–10% fetal calf serum61,145 and the development CL, Pfaff DW, eds. The Physiology of Reproduction.
of simple in vitro testis-like cultures in rodents,50,52 sug- Vol. 1. New York: Raven Press; 1988:837–932.
gest that these human Sertoli cells can be used in func- 13. McLachlan RI, O’Donnell L, Meachem SJ et
tional studies. In fact, human Sertoli cells have been used al. Hormonal regulation of spermatogenesis in
to probe the molecular mechanism(s) by which environ- primates and man: Insights for development
mental toxicants induce male reproductive dysfunction.61 of the male hormonal contraceptive. J Androl.
Furthermore, advances in genetic analysis have identified 2002;23:149–162.
References 177

14. Cheng CY, Mruk DD. The blood-testis barrier and its 32. Vaudry D, Falluel-Morel A, Bourgault S et al.
implication in male contraception. Pharmacol Rev. Pituitary adenylate cyclase-activating polypep-
2012;64:16–64. tide and its receptors: 20 years after the discovery.
15. Amann RP, Schanbacher BD. Physiology of male Pharmacol Rev. 2009;61(3):283–357.
reproduction. J Anim Sci. 1983;57(Suppl 2):380–403. 33. Smith ZD, Chan MM, Mikkelsen TS et al. A unique
16. De Felici M. Origina, migration, and proliferation regulatory phase of DNA methylation in the early
of humna primordial germ cells. In Coticchio G, mammalian embryo. Nature. 2012;484:339–344.
Albertini DF, De Santis L, eds. Oogenesis. London: 34. Kagiwada S, Kurimoto K, Hirota T, Yamaji M, Saitou
Springer-Verlag; 2013:19–37. M. Replication-coupled passive DNA demethylation
17. Saitou M, Miyauchi H. Gametogenesis from pluripo- for the erasure of genome imprints in mice. EMBO J.
tent stem cells. Cell Stem Cell. 2016;18:721–735. 2013;32:340–353.
18. Byskov AG. Differentiation of mammalian embry- 35. Seisenberger S, Andrews S, Krueger F et al. The
onic gonad. Physiol Rev. 1986;66:71–117. dynamics of genome-wide DNA methylation repro-
19. Tilmann C, Capel B. Cellular and molecular path- gramming in mouse primordial germ cells. Mol Cell.
ways regulating mammalian sex determination. 2012;48:849–862.
Recent Prog Horm Res. 2002;57:1–18. 36. Clermont Y. Renewal of spermatgonia in man. Am J
20. Hanley NA, Hagan DM, Clement-Jones M et al. SRY, Anat. 1966;118,509–529.
SOX9, and DAX1 expression patterns during human 37. Goossens E, Tournaye H. Adult stem cells in the
sex determination and gonadal development. Mech human testis. Semin Reprod Med. 2013;31:39–48.
Dev. 2000;91:403–407. 38. Oatley JM, Brinster RL. Regulation of spermatogo-
21. Culty M. Gonocytes, the forgotten cells of the gterm nial stem cell self-renewal in mammals. Annu Rev
cell lineage. Birth Defects Res C Embryo Today. Cell Dev Biol, 2008;24:263–286.
2009;87:1–26. 39. Yang QE, Oatley JM. Spermatogonial stem cell func-
22. Capel B, Albrecht KH, Washburn LL, Elcher EM. tions in physiological and pathological conditions.
Migration of mesonephric cells into the mammalian Curr Top Dev Biol. 2014;107:235–267.
gonad depends on Sry. Mech Dev. 1999;84:127–131. 40. Phillips BT, Gassei K, Orwig KE. Spermatogonial
23. Wen Q, Cheng CY, Liu YX. Development, function stem cell regulation and spermatogenesis. Philos
and fate of fetal Leydig cells. Semin Cell Dev Biol. Trans R Soc Lond B Biol Sci. 2010;365:1663–1678.
2016;59:89–98. 41. van den Driesche S, Sharpe RM, Saunders PT,
24. Bendsen E, Byskov AG, Laursen SB, Larsen HP, Mitchell RT. Regulation of the germ stem cell niche
Andersen CY, Westergaard LG. Number of germ as the foundation for adult spermatogenesis: A role
cells and somatic cells in human fetal testes dur- for miRNAs? Semin Cell Dev Biol. 2014;29:76–83.
ing the first weeks after sex differentiation. Human 42. Orth JM, Gunsalus GL, Lamperti AA. Evidence
Reprod. 2003;18:13–18. from Sertoli cell-depleted rats indicates that sper-
25. Murray TJ, Fowler PA, Abramovich DR, Haltes N, matid number in adults depends on numbers of
Lea RG. Human fetal testis: Second trimester prolif- Sertoli cells produced during perinatal development.
erative and steroidogenic capacities. J Clin Endocrinol Endocrinology. 1988;122:787–794.
Metab. 2000;85:4812–4817. 43. Cooke P, Hess R, Porcelli J, Meisami E. Increased
26. Josso N, Lamarre I, Picard JY et al. Anti-Mullerian sperm production in adult rats following transient
hormone in early human development. Early Hum neonatal hypothyroidism. Endocrinology. 1991;129:​
Dev. 1993;33:91–99. 244–248.
27. Winter JSD, Faiman C, Reyes FI. Sex steroid produc- 44. Sharpe RM, McKinnell C, Kivlin C, Fisher JS.
tion by the human fetus: Its role in morphogenesis and Proliferation and functional maturation of Sertoli
control by gonadotrophins. In Blandau RJ and Bergma cells, and their relevance to disorders of testis func-
D, eds. Morphogenesis and Malformation of the Genital tion in adulthood. Reproduction. 2003;125:769–784.
System. New York: Alan R. Liss; 1977: 41–58. 45. Weber JE, Russell LD, Wong V, Peterson RN. Three
28. Young JC, Wakitani S, Loveland KL. TGF-β super- dimensional reconstruction of a rat stage V Sertoli
family signaling in testis formation and early male cell: II. Morphometry of Sertoli-Sertoli and Sertoli-
germline development. Semin Cell Dev Biol. 2015;45:​ germ cell relationships. Am J Anat. 1983;167:163–179.
94–103. 46. Shaha C, Tripathi R, Mishra DP. Male germ cell
29. Barakat B, Itman CM, Mendis SH, Loveland KL. apoptosis: Regulation and biology. Philos Trans R Soc
Activins and inhibins in mammalian testis develop- Lond B Biol Sci. 2010;365:1501–1515.
ment: New models, new insights. Mol Cell Endocrinol. 47. Cheng CY, Mruk DD. Biochemistry of Sertoli
2012;359:66–77. cell/germ cell junctions, germ cell transport, and
30. Li C, Zhou X. The potential roles of neurotrophins in spermiation in the seminiferous epithelium. In
male reproduction. Reproduction. 2013;145:R89–R95. Griswold, M. D., ed. Sertoli Cell Biology, 2nd Edition.
31. Cool J, Capel B. Mixed signals: Development of the Amsterdam: Elsevier; 2015:333–383. DOI: http://
testis. Semin Reprod Med. 2009;27:5–13. dx.doi.org/10.1016/B978-0-12-417047-6.00012.0.
178  Advances in our understanding of human spermatogenesis

48. Gonsalves J, Sun F, Schlegel PN et al. Defectrive 63. Smith BE, Braun RE. Germ cell migration across
recombination in infertile men. Hum Mol Genet. Sertoli cell tight junctions. Science. 2012;338:​
2004;13:2875–2883. 798–802.
49. Sun F, Mikhaail-Philips M, Oliver-Bonet M, Ko E, 64. Kaitu’u-Lino TJ, Sluka P, Foo CF, Stanton PG.
Rademaker AT, P., Martin FH. Reduced meiotic Claudin-11 expression and localisation is regulated
recombination on the XY bivalent is correlated with by androgens in rat Sertoli cells in vitro. Reproduction.
an increased incidence of sex chromnosome aneu- 2007;133:1169–1179.
ploidy in men with non-obstructive azoospermia. 65. Meng J, Holdcraft RW, Shima JE, Griswold MD,
Mol Hum Reprod. 2008;14:399–404. Braun RE. Androgens regulate the permeability of
50. Sato T, Katagiri K, Gohbara A et al. In vitro produc- the blood-testis barrier. Proc Natl Acad Sci U S A.
tion of functional sperm in cultured neonatal mouse 2005;102:16696–16670.
testes. Nature. 2011;471:504–507. 66. Ilani N, Armanious N, Lue YH et al. Integrity of the
51. Sato T, Katagiri K, Yoknishi T et al. In vitro pro- blood-testis barrier in healthy men after suppression
duction of fertile sperm from murine spermato- of spermatogenesis with testosterone and levonorg-
gonial stem cell lines. Nat Commun. 2011;2:472. estrel. Hum Reprod. 2012;27(12):3403–3411.
DOI:10.1038/ncomms1478. 67. McCabe MJ, Tarulli GA, Laven-Law G et al.
52. Zhou Q, Wang M, Yuan Y et al. Complete meio- Gonadotropin suppression in men leads to a reduc-
sis from embryonic stem cell-derived germ cells in tion in claudin-11 at the Sertoli cell tight junction.
vitro. Cell Stem Cell. 2016;18:330–340. Hum Reprod. 2016;31(4):875–886.
53. Yang S, Ping P, Ma M et al. Generation of hap- 68. Matthiesson KL, Stanton PG, O’Donnell L et al.
loid spermatids with fertilization and develop- Effects of testosterone and levonorgestrel combined
ment capacity from human spermatogonial stem with a 5α-reductase inhibitor or gonadotropin-
cells of cryptorchid patients. Stem Cell Reports. releasing hormone antagonist on spermatogenesis
2014;3(4):663–675. and intratesticular steroid levels in normal men.
54. Cheng CY, Mruk DD. Cell junction dynamics in the J Clin Endocrinol Metab. 2005;90(10):5647–5655.
testis: Sertoli-germ cell interactions and male contra- 69. Matthiesson KL, Amory JK, Berger R, Ugoni A,
ceptive development. Physiol Rev. 2002;82:825–874. McLachlan RI, Bremner WJ. Novel male hormonal
55. Franca LR, Auharek SA, Hess RA, Dufour JM, contraceptive combinations: The hormonal and
Hinton BT. Blood-tissue barriers: Morphofunctional spermatogenic effects of testosterone and levonorg-
and immunological aspects of the blood-testis estrel combined with a 5α-reductase inhibitor or
and blood-epididymal barriers. Adv Exp Med Biol. gonadotropin-releasing hormone antagonist. J Clin
2012;763:237–259. Endocrinol Metab. 2005;90(1):91–97.
56. Pelletier RM. The blood-testis barrier: The junc- 70. Haverfield JT, Meachem SJ, O’Bryan MK,
tional permeability, the proteins and the lipids. Prog McLachlan RI, Stanton PG. Claudin-11 and con-
Histochem Cytochem. 2011;46:49–127. nexin-43 display altered spatial patterns of orga-
57. Fawcett DW, Leak LV, Heidger PM. Electron micro- nization in men with primary seminiferous tubule
scopic observations on the structural components of failure compared with controls. Fertil Steril. 2013;​
the blood-testis barrier. J Reprod Fertil. 1970;Suppl 100(3):658–666.
10:105–122. 71. Li N, Mruk DD, Lee WM, Wong CKC, Cheng CY.
58. Dym M, Fawcett DW. The blood-testis barrier in the Is toxicant-induced Sertoli cell injury in vitro a use-
rat and the physiological compartmentation of the ful model to study molecular mechanisms in sper-
seminiferous epithelium. Biol Reprod. 1970;3:308–326. matogenesis? Semin Cell Dev Biol. 2016;59:141–156.
59. Dym M. The fine structure of the monkey (Macaca) DOI:10.1016/j.semcdb.2016.01.003.
Sertoli cell and its role in maintaining the blood- 72. Siu ER, Mruk DD, Porto CS, Cheng CY. Cadmium-
testis barrier. Anat Rec. 1973;175:639–656. induced testicular injury. Toxicol Appl Pharmacol.
60. Moroi S, Saitou M, Fujimoto K et al. Occludin is 2009;238:240–249.
concentrated at tight junctions of mouse/rat but not 73. Wiebe J, Barr K. Suppression of spermatogen-
human/guinea pig Sertoli cells in testes. Am J Physiol esis without inhibition of steroidogenesis by 1,2,​
Cell Physiol. 1998;274:C1708–C1717. 3-​trihydroxypropane solution. Life Sci. 1984;34:​
61. Xiao X, Mruk DD, Tang EI et al. Environmental 1747–1754.
toxicants perturb human Serotli cell adhesive func- 74. Wiebe J, Kowalik A, Gallardi R, Egeler O, Clubb B.
tion via changes in F-actin organization medi- Glycerol disrupts tight junction-associated actin micro­
cated by actin regulatory proteins. Hum Reprod. filaments, occludin, and microtubules in Sertoli cells.
2014;29:1279–1291. J Androl. 2000;21:625–635.
62. Komljenovic D, Sandhoff R, Teigler A, Heid H, Just 75. Sofikitis N, Mantzavinos T, Loutradis D, Yamamoto
WW, Gorgas K. Disruption of blood-testis barrier Y, Tarlatzis V, Miyagawa I. Ooplasmic injections of
dynamics in either-lipid deficient mice. Cell Tissue secondary spermatocytes for non-obstructive azo-
Res. 2009;337:281–299. ospermia. Lancet. 1998;351:1177–1178.
References 179

76. Tesarik J, Balaban B, Isiklar A et al. In-vitro sper- 92. O’Donnell L, O’Bryan MK. Microtubules and sper-
matogenesis resumption in men with maturation matogenesis. Semin Cell Dev Biol. 2014;30:45–54.
arrest: Relationship with in-vivo blocking stage and 93. Russell LD. Spermatid-Sertoli tubulobulbar com-
serum FSH. Hum Reprod. 2000;15:1350–1354. plexes as devices for elimination of cytoplasm from
77. Clermont Y, Leblond CP. Spermiogenesis of man, the head region in late spermatids of the rat. Anat
monkey, ram and other mammals as shown by Rec. 1979;194:233–246.
the “periodic acid-Schiff” technique. Am J Anat. 94. Russell LD. Further observations on tubulobulbar
1955;96:229–253. complexes formed by late spermatids and Sertoli
78. Leblond C, Clermont Y. Definition of the stages of cells in the rat testis. Anat Rec. 1979;194:213–232.
the cycle of the seminiferous epithelium in the rat. 95. Vogl AW. Distribution and function of organized
Ann NY Acad Sci. 1952;55:548–573. concentrations of actin filaments in mammalian
79. Clermont Y. Kinetics of spermatogenesis in mam- spermatogenic cells and Sertoli cells. Int Rev Cytol.
mals: Seminiferous epithelium cycle and spermato- 1989;119:1–56.
gonial renewal. Physiol Rev. 1972;52:198–235. 96. Vogl AW, Young JS, Du M. New insights into roles of
80. Kierszenbaum AL, Rivkin E, Tres LL. Cytoskeletal tubulobulbar complexes in sperm release and turn-
track selection during cargo transport in sperma- over of blood-testis barrier. Int Rev Cell Mol Biol.
tids is relevant to male fertility. Spermatogenesis. 2013;303:319–355.
2011;1:221–230. 97. Young JS, Guttman JA, Vaid KS, Vogl AW. Cortactin
81. Kierszenbaum AL, Tres LL. The acrosome-acroplaxome-​ (CTTN), N-WASP (WASL), and clathrin (CLTC) are
manchette complex and the shaping of the spermatid present at podosome-like tubulobulbar complexes in
head. Arch Histol Cytol. 2004;67:271–284. the rat testis. Biol Reprod. 2009;80:153–161.
82. Song R, Ro S, Michaels JD, Park C, McCarrey JR, 98. Young JS, Guttman JA, Vaid KS, Vogl AW.
Yan W. Many X-linked microRNAs escape mei- Tubulobulbar complexes are intercellular podosome-­
otic sex chromosome inactivation. Nat Genet. like structures that internalize intact intercellular
2009;41:488–493. junctions during epithelial remodeling events in the
83. Song R, Hennig GW, Wu Q, Jose C, Zheng H, Yan W. rat testis. Biol Reprod. 2009;80:162–174.
Male germ cells express abundant endogenous siR- 99. Young JS, Takai YK, KL, Vogl AW. Internalization
NAs. Proc Natl Acad Sci U S A. 2011;108:​13159–131564. of adhesion junction proteins and their association
84. Dam AH, Feenstra I, Westphal JR, Ramos L, van with recycling endosome marker proteins in rat
Golde RJ, Kremer JA. Globozoospermia revisited. seminiferous epithelium. Reproduction. 2012;143:​
Hum Reprod Update. 2007;13(1):63–75. 347–357.
85. Taylor SL, Yoon SY, Morshedi MS et al. Complete 100. Young JS, Vogl AW. Focal adhesion proteins zyxin
globozoospermia associated with PLCζ deficiency and vinculin are co-distributed at tubulobulbar
treated with calcium ionophore and ICSI results complexes. Spermatogenesis. 2012;2:63–68.
in pregnancy. Reproductive BioMedicine Online. 101. Russell LD. Observations on the inter-­relationships
2010;20(4):559–564. of Sertoli cells at the level of the blood-testis barrier:
86. Yan W. Male infertility caused by spermiogenic Evidence for formation and resorption of Sertoli-
defects: Lessons from gene knockouts. Mol Cell Sertoli tubulobulbar complexes during the sper-
Endocrinol. 2009;306(1–2):24–32. matogenic cycle of the rat. Am J Anat. 1979;155:​
87. O’Donnell L. Mechanisms of spermiogenesis 259–279.
and spermiation and how they are disturbed. 102. Xiao X, Mruk DD, Wong EWP et al. Differential
Spermatogenesis. 2014;4(2):e979623. effects of c-Src and c-Yes on the endocytic v­ esicle-​
88. Huang WP, Ho HC. Role of microtubule-­pdependent mediated trafficking events at the Sertoli cell
membrane trafficking in acrosomal biogenesis. Cell blood-testis barrier: An in vitro study. Am J Physiol
Tissue Res. 2006;323:495–503. Endocrinol Metab. 2014;307:E553–E562.
89. Vandecandelaere A, Martin SR, Engelborghs Y. 103. Clermont Y, Morales C, Hermo L. Endocytic activi-
Response of mirotubules to the addition of colchicine ties of Sertoli cells in the rat. Ann NY Acad Sci. 1987;​
and tubulin-colchicine: Evaluation of models for the 513:1–15.
interaction of drugs with microtubules. Biochem J. 104. Siu MKY, Cheng CY. Interactions of proteases, pro-
1997;323:189–196. tease inhibitors, and the β1 integrin/laminin γ3
90. Ito C, Suzuki-Toyota F, Maekawa M et al. Failure to protein complex in the regulation of ectoplasmic
assemble the peri-nuclear structures in GOPC defi- specialization dynamics in the rat testis. Biol Reprod.
cient spermatids as found in round-headed sperma- 2004;70:945–964.
tozoa. Arch Histol Cytol. 2004;67:349–360. 105. Koch M, Olson PF, Albus A et al. Characterization
91. O’Donnell L, Nicholls PK, O’Bryan MK, McLachlan and expression of the laminin γ3 chain: A novel,
RI, Stanton PG. Spermiation: The process of sperm non-basement membrane-associated, laminin chain.
release. Spermatogenesis. 2011;1:14–35. J Cell Biol. 1999;145:605–618.
180  Advances in our understanding of human spermatogenesis

106. Yan HHN, Mruk DD, Wong EWP, Lee WM, Cheng 121. Cheng CY. Toxicants target cell junctions in the
CY. An autocrine axis in the testis that coordinates testis – insights from the indazole-carboxylic acid
spermiation and blood-testis barrier restructuring model. Spermatogenesis. 2014;4:e981485. DOI:10.416
during spermatogenesis. Proc Natl Acad Sci U S A. 1/21565562.2014.9814895.
2008;105:8950–8955. 122. Tang EI, Lee WM, Yan Cheng C. Coordination of
107. Yan HHN, Cheng CY. Laminin α3 forms a com- actin- and microtubule-based cytoskeletons sup-
plex with β3 and γ3 chains that serves as the ligand port transport of spermatids and residual bodies/­
for α6β1-integrin at the apical ectoplasmic special- phagosomes during spermatogenesis in the testis of
ization in adult rat testes. J Biol Chem. 2006;281:​ the male rat. Endocrinology. 2016;157:1644–1659.
17286–17303. 123. Cheng CY, Mruk DD. Regulation of spermiogen-
108. Su L, Mruk DD, Lie PPY, Silvestrini B, Cheng CY. A esis, spermiation and blood-testis barrier d
­ ynamics:
peptide derived from laminin-γ3 reversibly impairs Novel insights from studies on Eps8 and Arp3.
spermatogenesis in rats. Nat Communs. 2012;3:1185. Biochem J. 2011;435:553–562.
DOI:1110.1038/ncomms2171. 124. Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli-
109. Cheng CY, Mruk DD. A local autocrine axis in the germ cell interactions and their significance in germ
testes that regulates spermatogenesis. Nature Rev cell movement in the seminiferous epithelium dur-
Endocrinol. 2010;6:380–395. ing spermatogenesis. Endocr Rev. 2004;25:747–806.
110. Cheng CY, Mruk DD. An intracellular traffick- 125. Wong EWP, Mruk DD, Cheng CY. Biology and
ing pathway in the seminiferous epithelium regulation of ectoplasmic specialization, an atypi-
regulating spermatogenesis: A biochemical and cal adherens junction type, in the testis. Biochem
molecular ­perspective. Crit Rev Biochem Mol Biol. Biophys Acta. 2008;1778:692–708.
2009;44:245–263. 126. Mruk DD, Cheng CY. Cell-cell interactions at the
111. Lie PPY, Mruk DD, Mok KW, Su L, Lee WM, ectoplasmic specialization in the testis. Trends
Cheng CY. Focal adhesion kinase-Tyr407 and -Tyr397 Endocrinol Metab. 2004;15:439–447.
exhibit antagonistic effects on blood-testis bar- 127. Smith LB, Milne L, Nelson N et al. KATNAL1 regula-
rier dynamics in the rat. Proc Natl Acad Sci U S A tion of Sertoli cell microtubule dynamics is essential
2012;109:12562–12567. for spermiogenesis and male fertility. PLoS Genet.
112. Jamsai D, O’Bryan MK. Mouse models in male fertil- 2012;8:e1002697.
ity research. Asian J Androl. 2011;13:139–151. 128. O’Donnell L, Rhodes D, Smith SJ et al. An essential
113. McLachlan RI, O’Bryan MK. State of the art for role for katanin p80 and microtubule severing in male
genetic testing of infertile men. J Clin Endocrinol gamete production. PLoS Genet. 2012;8(5):e1002698.
Metab. 2010;95(3):1013–1024. 129. O’Donnell L, McLachlan RI, Merriner DJ, O’Bryan MK,
114. Johnson KJ. Testicular histopathology associated Jamsai D. KATNB1 in the human testis and its genetic
with disruption of the Sertoli cell cytoskeleton. variants in fertile and oligoasthenoteratozoospermic
Spermatogenesis. 2014;4:e979106. DOI:10.4161/2156 infertile men. Andrology. 2014;2(6):884–891.
5562.2014.979106. 130. Rotkopf S, Hamberg Y, Aigaki T, Snapper SB, Shilo
115. Boekelheide K, Fleming SL, Allio T et al. BZ, Shejter ED. The WASp-based actin polymeriza-
2,5-Hexanedione-induced testicular injury. Annu tion machinery is required in somatic support cells
Rev Pharmacol Toxciol. 2003;43:125–147. for spermatid maturation and release. Development.
116. Boekelheide K, Neely MD, Sioussat TM. The Sertoli 2011;138:2729–2739.
cell cytoskeleton: A target for toxicant-induced germ 131. Xiao X, Mruk DD, Tang EI et al. N-WASP is required
cell loss. Toxicol Appl Pharmacol. 1989;101:373–389. for structural integrity of the blood-testis barrier.
117. Allard EK, Johnson KJ, Boekelheide K. Colchicine PLoS Genet. 2014;10(6):e1004447.
disrupts the cytoskeleton of rat testis seminifer- 132. Zirkin BR. Spermatogenesis: Its regulation by testos-
ous epithelium in a stage-dependent manner. Biol terone and FSH. Semin Cell Dev Biol. 1998;9:417–421.
Reprod. 1993;48:143–153. 133. Jarow JP, Zirkin BR. The androgen microen-
118. Nakai M, Hess RA. Morphological changes in the rat vironment of the human testis and hormonal
Sertoli cell induced by the microtubule poison car- control of ­ spermatogenesis. Ann NY Acad Sci.
bendazim. Tissue Cell. 1994;26:917–927. 2005;1061:208–220.
119. Li N, Mruk DD, Wong CKC, Han D, Lee WM, Cheng 134. Turner TT, Jones CC, Howards SS, Ewing LL, Zegeye
CY. Formin 1 regulates ectoplamic specialization in B, Gunsalus GL. On the androgen microenviron-
the rat testis through its actin nucleation and bun- ment of maturing spermatozoa. Endocrinology.
dling activity. Endocrinology. 2015;156:2969–2983. 1984;115:1925–1932.
120. Li N, Mruk DD, Wong CK, Lee WM, Han D, Cheng 135. O’Donnell L, McLachlan RI, Wreford NG, de Kretser
CY. Actin-bundling protein plastin 3 is a regula- DM, Robertson DM. Testosterone withdrawal pro-
tor of ectoplasmic specialization dynamics dur- motes stage-specific detachment of round sper-
ing spermatogenesis in the rat testis. FASEB J. matids from the rat seminiferous epithelium. Biol
2015;29:3788–3805. Reprod. 1996;55:895–901.
References 181

136. Xia W, Wong CH, Lee NPY, Lee WM, Cheng CY. 152. Bashamboo A, Ferraz-de-Souza B, Lourenco D et al.
Disruption of Sertoli-germ cell adhesion function Human male infertility associated with mutations in
in the seminiferous epithelium of the rat testis can NR5A1 encoding steroidogenic factor 1. Am J Hum
be limited to adherens junctions without affecting Genet. 2010;87(4):505–512.
the blood-testis barrier integrity: An in vivo study 153. Seminara SB, Oliveira LM, Beranova M, Hayes FJ,
using an androgen suppression model. J Cell Physiol, Crowley WF, Jr. Genetics of hypogonadotropic hypo-
2005;205:141–157. gonadism. J Endocrinol Invest. 2000;23(9):560–565.
137. O’Donnell L, McLachlan R, Wreford N, Robertson D. 154. Oliveira LM, Seminara SB, Beranova M et al. The
Testosterone promotes the conversion of round sper- importance of autosomal genes in Kallmann syn-
matids between stages VII and VIII of the rat sper- drome: Genotype-phenotype correlations and neu-
matogenic cycle. Endocrinology. 1994;135:2608–2614. roendocrine characteristics. J Clin Endocrinol Metab.
138. Hess RA, Bunick D, Lee KH et al. A role for oes- 2001;86(4):1532–1538.
trogens in the male reproductive system. Nature. 155. Quigley CA, De Bellis A, Marschke KB, el-Awady
1997;390:509–512. MK, Wilson EM, French FS. Androgen receptor
139. Hess RA. Estrogen in the adult male reproductive defects: Historical, clinical, and molecular perspec-
tract: A review. Reprod Biol Endocrinol. 2003;1:52:​1–14. tives. Endocr Rev. 1995;16(3):271–321.
140. Robertson KM, O’Donnell L, Jones ME et al. 156. Casals T, Bassas L, Ruiz-Romero J et al. Extensive anal-
Impairment of spermatogenesis in mice lacking a ysis of 40 infertile patients with congenital absence of
functional aromatase (cyp19) gene. Proc Natl Acad the vas deferens: In 50% of cases only one CFTR allele
Sci U S A. 1999;96:7986–7991. could be detected. Hum Genet. 1995;95(2):205–211.
141. Cram D, Lynch M, O’Bryan MK, Salvado C, 157. Ferlin A, Bogatcheva NV, Gianesello L et al. Insulin-
McLachlan RI, De Kretser DM. Genetic screening of like factor 3 gene mutations in testicular dysgenesis
infertile men. Reprod Fertil Dev. 2004;16:573–580. syndrome: Clinical and functional characterization.
142. Lian J, Zhang X, Tian H et al. Altered microRNA Mol Hum Reprod. 2006;12(6):401–406.
expression in patients with non-obstructive azo- 158. Foresta C, Ferlin A. Role of INSL3 and LGR8 in
ospermia. Reprod Biol Endocrinol. 2009;7:13. cryptorchidism and testicular functions. Reprod
143. Wu W, Qin Y, Li Z et al. Genome-wide microRNA Biomed Online. 2004;9(3):294–298.
expression profiling in idiopathic non-­obstructive 159. Avenarius MR, Hildebrand MS, Zhang Y et al. Human
azoospermia: Significant up-regulation of miR- male infertility caused by mutations in the CATSPER1
141, miR-429 and miR-7-1-3p. Hum Reprod. channel protein. Am J Hum Genet. 2009;84(4):505–510.
2013;28(7):1827–1836. 160. Dieterich K, Soto Rifo R, Faure AK et al. Homozygous
144. Chen Q, Yan W, Duan E. Epigenetic inheritance mutation of AURKC yields large-headed polyploid
of acquired traits through sperm RNAs and sperm spermatozoa and causes male infertility. Nat Genet.
RNA modifications. Nat Rev Genet. 2016;17:733–743. 2007;39(5):661–665.
145. Chui K, Trivedi A, Cheng CY et al. Characterization 161. Dam AH, Koscinski I, Kremer JA et al. Homozygous
and functionality of proliferative human Sertoli mutation in SPATA16 is associated with male infer-
cells. Cell Transplant. 2011;20:619–635. tility in human globozoospermia. Am J Hum Genet.
146. Orth JM. Proliferation of Sertoli cells in fetal and 2007;81(4):813–820.
postnatal rats: A quantitative autoradiographic study. 162. Koscinski I, Elinati E, Fossard C et al. DPY19L2 dele-
Anat Rec, 1982;203:485–492. tion as a major cause of globozoospermia. Am J Hum
147. Krausz C, Forti G, McElreavey K. The Y chromo- Genet. 2011;88(3):344–350.
some and male fertility and infertility. Int J Androl. 163. Harbuz R, Zouari R, Pierre V et al. A recurrent dele-
2003;26(2):70–75. tion of DPY19L2 causes infertility in man by block-
148. Ferlin A, Garolla A, Foresta C. Chromosome abnor- ing sperm head elongation and acrosome formation.
malities in sperm of individuals with constitutional Am J Hum Genet. 2011;88(3):351–361.
sex chromosomal abnormalities. Cytogenet Genome 164. Zuccarello D, Ferlin A, Cazzadore C et al. Mutations
Res. 2005;111(3–4):310–316. in dynein genes in patients affected by isolated
149. O’Flynn O’Brien KL, Varghese AC, Agarwal A. The non-syndromic asthenozoospermia. Hum Reprod.
genetic causes of male factor infertility: A review. 2008;23(8):1957–1962.
Fertil Steril. 2010;93(1):1–12. 165. Wu H, Sun L, Wen Y et al. Major spliceosome defects
150. Tiepolo L, Zuffardi O. Localization of factors con- cause male infertility and are associated with nonob-
trolling spermatogenesis in the nonfluorescent por- structive azoospermia in humans. Proc Natl Acad Sci
tion of the human Y chromosome long arm. Hum U S A. 2016; 113(15):4134–4139.
Genet. 1976;34(2):119–124. 166. Richardson ME, Bleiziffer A, Tuttelmann F,
151. Vogt PH, Edelmann A, Kirsch S et al. Human Y Gromoll J, Wilkinson MF. Epigenetic regulation of
chromosome azoospermia factors (AZF) mapped the RHOX homeobox gene cluster and its associa-
to different subregions in Yq11. Hum Mol Genet. tion with human male infertility. Hum Mol Genet.
1996;5(7):933–943. 2014;23(1):12–23.
182  Advances in our understanding of human spermatogenesis

167. Reynolds N, Collier B, Maratou K et al. Dazl binds in 176. Ravel C, El Houate B, Chantot S et al. Haplotypes,
vivo to specific transcripts and can regulate the pre- mutations and male fertility: The story of the testis-
meiotic translation of Mvh in germ cells. Hum Mol specific ubiquitin protease USP26. Mol Hum Reprod.
Genet. 2005;14(24):3899–3909. 2006;12(10):643–646.
168. Park JH, Lee HC, Jeong YM et al. MTHFR C677T 177. Ribarski I, Lehavi O, Yogev L et al. USP26 gene
polymorphism associates with unexplained infertile variations in fertile and infertile men. Hum Reprod.
male factors. J Assist Reprod Genet. 2005;22(9–10):​ 2009;24(2):477–484.
361–368. 178. Tapanainen JS, Aittomaki K, Min J, Vaskivuo T,
169. Singh K, Singh SK, Sah R, Singh I, Raman R. Mutation Huhtaniemi IT. Men homozygous for an inactivat-
C677T in the methylenetetrahydrofolate reductase ing mutation of the follicle-stimulating hormone
gene is associated with male infertility in an Indian (FSH) receptor gene present variable suppression of
population. Int J Androl. 2005;28(2):115–119. spermatogenesis and fertility. Nat Genet. 1997;15(2):​
170. Jamsai D, Lo JC, McLachlan RI, O’Bryan MK. 205–206.
Genetic variants in the RABL2A gene in fertile and 179. Ahda Y, Gromoll J, Wunsch A et al. Follicle-
oligoasthenospermic infertile men. Fertil Steril. 2014;​ stimulating hormone receptor gene haplotype distri-
102(1):223–229. bution in normozoospermic and azoospermic men.
171. Lazaros L, Xita N, Kaponis A, Zikopoulos K, J Androl. 2005;26(4):494–499.
Sofikitis N, Georgiou I. Evidence for association of 180. Guarducci E, Nuti F, Becherini L et al. Estrogen
sex ­hormone-binding globulin and androgen recep- receptor alpha promoter polymorphism: Stronger
tor genes with semen quality. Andrologia. 2008;40(3):​ estrogen action is coupled with lower sperm count.
186–191. Hum Reprod. 2006;21(4):994–1001.
172. Akinloye O, Gromoll J, Callies C, Nieschlag E, 181. Yoshida R, Fukami M, Sasagawa I, Hasegawa T,
Simoni M. Mutation analysis of the X-chromosome Kamatani N, Ogata T. Association of cryptorchi-
linked, testis-specific TAF7L gene in spermatogenic dism with a specific haplotype of the estrogen recep-
failure. Andrologia. 2007;39(5):190–195. tor alpha gene: Implication for the susceptibility
173. Stouffs K, Willems A, Lissens W, Tournaye H, Van to  estrogenic environmental endocrine disruptors.
Steirteghem A, Liebaers I. The role of the testis-­ J Clin Endocrinol Metab. 2005;90(8):4716–4721.
specific gene hTAF7L in the aetiology of male infer- 182. O’Bryan MK, Grealy A, Stahl PJ, Schlegel PN,
tility. Mol Hum Reprod. 2006;12(4):263–267. McLachlan RI, Jamsai D. Genetic variants in the ETV5
174. Stouffs K, Lissens W, Tournaye H, Van Steirteghem gene in fertile and infertile men with nonobstructive
A, Liebaers I. Possible role of USP26 in patients azoospermia associated with Sertoli cell-only syn-
with severely impaired spermatogenesis. Eur J Hum drome. Fertil Steril. 2012;98(4):827–835, e821–e823.
Genet. 2005;13(3):336–340.
175. Paduch DA, Mielnik A, Schlegel PN. Novel muta-
tions in testis-specific ubiquitin protease 26 gene
may cause male infertility and hypogonadism.
Reprod Biomed Online. 2005;10(6):747–754.
A look into the testis as a reservoir for
HIV and ZIKV—A reproductive biologist’s
14
perspective
ELIZABETH I. TANG, CHRISTOPHER L. ROBINSON, CHI NOK CHONG,
SHUIBING CHEN, and C. YAN CHENG

INTRODUCTION this organ has few major diseases. Although the testis is
Immune privilege is a concept which arose from a group immune-privileged, it does not mean that it is entirely
of work that revealed that specific sites in the body, such immune from infections. Studies have shown viruses,
as the eye, brain, testis, and others, can protect grafted such as mumps, human immunodeficiency v­ irus-(HIV) 1,
tissue from rejection.1,2 There are two distinct aspects of human papillomavirus (HPV), and Zika virus (ZIKV)
immune privilege: privileged sites and privileged tissues, can afflict the testes.13–15 Some of these viruses are sexu-
which should be noted.3 A privileged site describes a site ally transmitted, as studies detected the presence of the
in which foreign tissue grafts can survive longer, or indefi- viruses in semen.13,16–18 There is increasing evidence sug-
nitely, than in a nonprivileged site. A privileged tissue gesting that the testis may serve as a sanctuary site for
refers to tissue that resists rejection when grafted into a viruses, which take advantage of the immune-privileged
nonprivileged site. The testis is a unique organ because it nature of the testis, making the testis a safe haven.19,20 This
encompasses both aspects of immune privilege.4 The first chapter explores what is currently known in the literature
evidence that the testis is immune-privileged dates back to about the presence of select viruses in the testis and how
1786, when John Hunter transplanted a cock testis into the they may evade or exploit the protective mechanisms of
abdomen of a hen.5 This was the first recorded indication the testis. Further understanding on how the viruses may
that the testis can induce tolerance when transplanted into harbor themselves in the testis will be helpful in develop-
an allogeneic recipient, as the testis transplant appeared to ing treatments for these viruses such as their eradication.
have no noticeable effects on the hen. During the 1970s,
almost 200 years after Hunter’s experiment, evidence that CONTRIBUTING FACTORS OF TESTIS IMMUNE
the testis can receive foreign cells and suppress rejection PRIVILEGE
arose. In these studies, tissue allografts or parathyroid Systemic immune tolerance, local immunoregulatory
glands were implanted in the interstitial space of rat testis mechanisms, as well as the physical structure of the tes-
and survived.6 These discoveries paved the way for later tis are all contributing factors of testis immune privilege.
cotransplantation studies in which pancreatic islet cells In a typical cross section of a testis, one can see that the
were transplanted with testicular Sertoli cells (SCs), which testis is primarily composed of the seminiferous tubules
have been found to suppress immune responses, helping with interstitial tissue (space) between them (Figure 14.1).
the islet cells to survive.7–12 Sertoli cells have been deemed The interstitial tissue contains blood vessels, immune
the “nurse” cells of the testis and serve many roles, one of cells, such as macrophages, dendritic cells, lymphocytes,
which is conferring immune privilege. and mast cells, which confer the local immunoregulatory
Immune privilege of the testis is necessary for safe- mechanisms of the immune-privileged testis (Figure 14.1).
guarding the propagation and proliferation of genetic These immunoregulatory mechanisms have been reviewed
material, such as during meiosis and postmeiotic sperma- recently4,21,22 and also appear elsewhere in this book, and
tid development through spermiogenesis and the release thus will not be discussed further herein.
of sperm at spermiation (Figure 14.1). The testis is an The structure of the seminiferous tubules is also impor-
essential organ of the male reproductive system and is tant for immune privilege of the testis. The seminiferous
the site of mammalian spermatogenesis. Spermatogenesis tubules are surrounded by peritubular myoid cells, which
is a complex process in which undifferentiated hap- together with SCs help to form the basal lamina (also
loid germ cells (GCs) develop into differentiated hap- known as the basement membrane) of the seminiferous
loid spermatozoa, which takes place in the seminiferous epithelium through their secretory proteins and other
tubules of the testis. The testis protects developing GCs constituent products (e.g., laminins), providing structural
from systemic circulation via local immunoregulatory integrity to the tubules.23,24 The seminiferous epithelium is
mechanisms, peritubular myoid cells, Leydig cells, as a specialized microenvironment for spermatogenesis and
well as by the blood-testis barrier (BTB) formed by Sertoli is composed of only two cell types: SC and GC at various
cells. This cumulative defense system of the testis con- stages of their development. As previously mentioned, SCs
fers its immune-privileged nature, which is largely why have innate immunosuppressive properties, which is why

183
184  A look into the testis as a reservoir for HIV and ZIKV—A reproductive biologist’s perspective

Seminiferous
tubule

Interstitial
space

Seminferous tubules:
Pachytene Spermatogonium Myroid cell
Elongated spermatocyte
spermatid Blood-testis Spermatogonial Lymph Sertoli
barrier stem cell niche cell
Round Preleptotene Basement membrane Lymphatic
spermatid spermatocyte and type I collagen layer endothelium

Interstitial space:

Leydig Mast Dendritic Blood


Macrophage
cell cell cell vessel

Figure 14.1  Morphological features of the seminiferous epithelium in mammalian testes. The micrograph in the left panel is a
cross section of an adult rat testis. The schematic drawing in the right panel illustrates the basic morphological features of the semi-
niferous epithelium. The blood-testis barrier, constituted by coexisting actin-based tight junctions, basal ectoplasmic specialization,
and gap junction, as well as intermediate filament-based desmosome, divides the epithelium into the basal and adluminal (apical)
compartment. Both undifferentiated and differentiated type A spermatogonia and preleptotene spermatocytes derived from type
B spermatogonia are found in the basal compartment, whereas late spermatocytes (including pachytene/zygotene/diplotene sper-
matocytes, secondary spermatocytes, and spermatids [steps 1–19 in the rat testis]) are found in the adluminal compartment. Blood
vessels, Leydig cells, dendritic cells, and macrophages, however, are all found in the interstitial space, whereas only Sertoli cells and
germ cells at different stages of their development constitute the seminiferous epithelium. Seminiferous epithelium lays next to the
basement membrane (a modified form of basal lamina in the testis), underneath of which is the type 1 collagen, to be followed by
the myoid cell layer and then the lymphatic vessel, constituting the tunica propria.

they are used to cotransplant pancreatic islet cells. In addi- them except in pathological conditions.36–39 However,
tion, SC, or “nurse” cells, have numerous essential roles when these autoantigens are injected in other parts of the
in the testis, such as providing nutritional and structural body, a strong autoimmune response is directed against
support for differentiating GCs and formation of the BTB. them, which is compelling evidence to support the unique
The BTB is one of the tightest blood-tissue barriers and immunosuppressive properties of the testis.21,40 Thus,
divides the seminiferous epithelium into two compart- suppression of immunological response to expression of
ments, adluminal (also known as apical) and basal.25–28 autoantigens by either GCs outside the BTB in the basal
It is comprised of specialized junctions between adjacent compartment or by GCs in the adluminal compartment is
SCs near the base of the epithelium, serving as an immu- an indication of the highly regulated immunoregulatory
nological barrier.29–33 mechanisms of the testis.
During GC development, autoantigens are transiently
expressed by GCs undergoing meiosis as well as by devel- VIRUSES AND THE TESTIS
oping spermatids.22,34 The BTB protects GCs, postmeiotic The mammalian testis is known to have a sophisticated
spermatids in particular, from immunological response system to defend against viral infections. It has been
to their autoantigens. Interestingly, premature GCs such hypothesized that viral entry into the testis via blood is
as spermatogonia and preleptotene spermatocytes that lie subject to three lines of defense: (1) Leydig cells and mac-
outside the BTB also express autoantigens35 as well as many rophages in the interstitial space, (2) peritubular myoid
cancer/testis (CT) antigens and oncogenes,36 yet male cells that lay at the periphery of the seminiferous tubules,
rodents and humans do not develop antibodies against and (3) Sertoli cells.41 How viruses can evade the first two
Viruses and the testis  185

lines of defense is still unclear. Overcoming the third line Both the CNS and genitourinary tracts have been
of defense to invade the seminiferous epithelium proves to proposed to be reservoirs for latent HIV infection.19,45
be a daunting task. Part of the SC antiviral defense system Sanctuary sites in the body are anatomical sites that are
consists of interferon response; SCs constitutively express nearly inaccessible by drugs. The CNS is a well-established
interferons (IFN), and after exposure to virus the levels of sanctuary site due to the blood-brain barrier (BBB), a
multiple interferons increase considerably.41 As previously selective semipermeable membrane that segregates the
discussed, SCs protect developing late spermatocytes brain from the circulatory system. The brain is an ideal
and postmeiotic germ cells through the formation of the reservoir for HIV as it is an immune-privileged site and
BTB. In short, the BTB segregates the events of meiosis I/ because the BBB protects ART drugs from effectively
II and postmeiotic spermatid development from systemic entering the brain. The testis is also an immune-privileged
circulation and the host immune system. Interestingly, site and has its own blood-tissue barrier namely the BTB,
it was found that GCs per se, unlike SCs, express lower which also effectively prevents ART drugs from passing
levels of IFNs and following exposure to virus, GC IFN into the testis. Both these sites are efficiently protected
levels change minimally.42 This possibly suggests that GCs from leukocyte trafficking, making these ideal sanctuary
are more susceptible to viral infection if a virus can pass sites for HIV, where they can hide and eventually evolve
through the BTB in the first place. Behind the BTB, GCs and emerge as more virulent.46
express autoantigens during differentiation and are pro- As mentioned above, the testis is an ideal site for HIV to
tected from immune response due to their presence. Thus, hide because it is immune-privileged. Features of immune
this also suggests the possibility of a suppressed immune privilege allow the testis to facilitate fertility, such as by
response if viruses penetrate the BTB and stay behind in tolerating GC autoantigens that are transiently expressed
the adluminal compartment. On the other hand, SCs and during differentiation. HIV has been detected in semen
GCs secrete defensins, which are a family of antimicrobial but its origins in the genitourinary tract are still not clearly
peptides that may also target viruses.43,44 It is known that defined. HIV was found in semen as free viral particles,
some viruses can evade the testicular antiviral system, which proved that infected lymphocytes and macrophages
infecting GC/sperm, and remain in considerable quanti- from the blood were not the only sources of the virus.47
ties in semen; however, how they do so is still unknown. Subsequently, it was shown that free viral particles in
Sexual transmission of viruses is a public health con- semen can be different from those isolated from infected
cern. HIV/acquired immunodeficiency syndrome (AIDS) leukocytes in semen, suggesting that HIV can be compart-
is a global health pandemic and ZIKV is emerging as a mentalized in genital tissues.48
novel sexually transmitted virus. These two unrelated Studies have shown that men receiving ART have reduced
viruses do share something in common: both are found in viral load. However, some of these men can still shed HIV-1
immune-privileged sites such as the testis and brain when in their semen, suggesting that the testis is an antiretroviral
their viral RNA levels are virtually undetectable in serum. drug sanctuary.49–51 Similar results were found in simian
This chapter explores what is currently known about HIV immunodeficiency virus (SIV)-infected macaques.52 ART
and ZIKV in the testis. reduced viral load in blood but did not affect viral DNA
levels in genital tissues compared to untreated macaques.
HIV In another study, human testes samples from virally sup-
HIV is a retrovirus that causes HIV infection, and if left pressed patients were examined and confirmed primate
untreated can lead to the potentially life-threatening con- study findings.14 In this study, expression of C-C chemokine
dition, AIDS. It is a virus that attacks the immune sys- receptor 5 (CCR5), the main coreceptor of HIV, increased as
tem, specifically CD4 cells (T cells), which help the body’s compared to expression of the same coreceptor in peripheral
immune system fight off infections. The loss or a declining blood in these virally suppressed patients, suggesting that
population of T cells makes the body more prone to infec- the human testis is a sanctuary site for HIV and provides
tion and other diseases such as pneumonia. HIV is most an immune-tolerant environment for the virus. However,
commonly transmitted through contact with bodily flu- one limitation of this study is that whole testis lysates were
ids, such as blood, semen, vaginal fluids, and breast milk. used for examination, thereby preventing investigators
Even with treatment, HIV cannot be fully eliminated from identifying which testicular cell type(s) was infected
from the body and remains throughout an infected indi- or harboring the virus.
vidual’s lifetime. However, with the use of HIV medicines Since HIV viruses are detected in the testis and semen,
or antiretroviral therapy (ART), people with HIV can lead it is quite possible that spermatozoa/sperm may be carri-
longer, healthier lives while reducing the risk of transmis- ers or infected by the virus. However, whether or not HIV
sion to other individuals. The goal of ART is to achieve and viruses are detected in sperm remains controversial.53–55
maintain viral suppression. Monitoring plasma HIV RNA Depending on the technique used, HIV can be found with
(viral load) is an important indicator of positive response sperm.56 There are reports in the literature suggesting that
to ART. However, measuring viral load does not measure HIV is present on sperm. One such study detected CD4
the presence of HIV in sites protected from systemic cir- receptor, which has a high affinity for HIV, on the surface
culation, such as the central nervous system (CNS) and of sperm, indicating that it may be a vehicle for HIV.56
genitourinary tracts. Another study, using transmission electron microscopy
186  A look into the testis as a reservoir for HIV and ZIKV—A reproductive biologist’s perspective

to examine sperm obtained from AIDS patients, which Finding a suitable model to study ZIKV has proven
illustrated the presence of virus-like particles in sperm challenging. To date, a small number of studies investi-
cytoplasm, supporting the notion that viruses could infect gating the role of ZIKV and its consequences in the male
sperm.53,57 These and other studies in which GC/sperm reproductive tract have emerged. In humans, ZIKV targets
were cultured in vitro also showed that they can become transcription factor STAT2 and initiates its degradation,
infected or be hosts for HIV.53,58 In contrast, other reports disrupting type I IFN signaling.73,74 However, when wild-
challenged those findings as very low levels of CD4 recep- type (WT) mice, a typical study model, were infected with
tor were detected on sperm surface57 and that virus-like ZIKV they were found to have little or no infectious virus/
particles were in fact acrosomal reaction vesicles.54 viral RNA in their tissues.71 Interestingly, ZIKV does not
Other possible routes of HIV association or infec- target STAT2 in WT mice, which may be one of the rea-
tion of sperm may be through early infection of GCs. sons why WT mice are resistant to ZIKV infection.71,73,74
Spermatogenesis is the process by which spermatogo- Due to the general resistance of immunocompetent mice
nial stem cells or premature GCs develop into sperm. to ZIKV, scientists have used other approaches to establish
Spermatogonial stem cells reside outside the BTB as they a working ZIKV mouse model.
develop into preleptotene spermatocytes, which are the Currently, scientists use immunocompromised mice to
only GC type able to pass through the BTB and enter the study the pathogenesis of ZIKV and persistence in the male
adluminal compartment for subsequent development. reproductive tract. In one study, researchers inoculated
Infection of GCs may be a path by which HIV may harbor INF α/ß receptor-deficient mice (Ifnar1-/-) or WT mice
in the testis, a sanctuary site. In one study in which tes- treated with anti-Ifnar1 MAb (IFNα receptor 1 monoclo-
tes of AIDS patients were examined, investigators found nal antibody) to block IFN signaling. Findings from this
evidence of decreased spermatogenesis and atrophy in the study were primarily from the Ifnar1-/- mice data.75 They
testes. Investigators also detected considerable loss of GCs examined the genital tract of these mice and reported that
as well as HIV positivity in residual GCs, supporting the ZIKV was present in the testis and epididymis but not in
notion that HIV-infected GCs may be partially responsible the prostate and seminal vesicles. Progressive testicular
for sexual transmission of HIV.59 Additionally, other find- damage was observed in these mice. At 8 dpi (days postin-
ings suggest the possibility of direct infection of GC by cell- fection), mouse testes already began to display signs of
free virus as HIV-1 proviral DNA was detected in the nuclei orchitis, degeneration of GC and Leydig cells, and lym-
of GCs at all stages of differentiation.60,61 Collectively, phocyte and leukocyte infiltration inside the seminiferous
these findings suggest that GCs could be infected by HIV. tubules. By 30 dpi and longer, seminiferous tubules were
structurally damaged and testes atrophic. Additionally,
ZIKV these investigators also detected the presence of ZIKV in
ZIKV is a flavivirus that can cause microcephaly and specific testicular cell types and found it in spermatogo-
Guillain-Barré syndrome as well as other neurological nia, spermatocytes, sperm, and SCs.
complications.62–64 Typical symptoms of infected individu- A subsequent study published by a different group used
als are usually mild, characterized by fever, headache, rash, WT mice treated with anti-Ifnar1 antibody and infected
joint pain, conjunctivitis, and muscle pain (www.cdc.gov) them with a mouse-adapted ZIKV strain. They discov-
lasting for a few days to a week. Since symptoms are mild, ered that ZIKV caused significant testicular damage as
most infected individuals do not realize they are infected. well, in addition to decreased sperm count and ­fertility.76
In 2015, a large-scale outbreak of ZIKV in Brazil was cause Cell types infected in this study were SC, GC/sperm, and
for alarm. By 2016, ZIKV was declared a global health possibly Leydig cells in the testes but also in the epididymis.
emergency by the World Health Organization (WHO).65 Since then a number of studies have used similar approaches
Prior to this, small outbreaks were detected in other conti- to study the pathogenesis of ZIKV. What these studies have
nents, ranging from Africa to Southeast Asia and Oceania. in common is that they all utilize ZIKV-infected immuno-
Conventionally, ZIKV is considered an arborvirus since it compromised mice. As seen in Table 14.1, defining which
is transmitted by mosquitoes. However, recent outbreaks cell types are targeted by ZIKV is inconclusive. Results
have revealed that ZIKV can also be spread via sexual vary depending on the ZIKV strain in combination with
(both homo- and heterosexual) and vertical transmission the type of immunocompromised mice. Understanding
routes unlike most flaviviruses.17,18,66–70 Zika viruses can which cell types are susceptible to ZIKV infection can help
pass through the placenta during pregnancy and infect the to further uncover the biology of the virus, especially how
fetus to cause microcephaly as well as cause Guillain-Barré the virus is able to evade the highly immune-privileged
syndrome in adults.71 Clinical observations in male ZIKV nature of the testis and male r­ eproductive tract.
patients detected the presence of the virus in semen for up
to 6 months, suggesting the possibility of transmission long CONCLUDING REMARKS
after the appearance of symptoms.72 These findings among The clinical implications of HIV and ZIKV are drasti-
others, in addition to the declaration of a global health cally different; however, both are global health concerns.
emergency, spurred scientists worldwide to better under- Further understanding of how both viruses evade the
stand the pathogenesis of ZIKV. ZIKV and its impact on immunoregulatory mechanisms of the testis, as well as
male reproductive health is the subject of this section. other sanctuary sites like the brain, will be helpful for
Concluding remarks  187

Table 14.1  Results of selected study models investigating Zika virus infection in the male genitourinary tract.
Infected cell
Zika virus strain Study model type(s) Cell location Technique d.p.i. Ref.
From patient returning ZIKV positive semen Sperm Semen IF 56a 81

from trip to French from human patient


Guyana, 2016

ZIKA-SMGC-1 (isolated ZIKV infected Spermatogonia/GC & Testes & epididymis IF 8, 16 & 30 75

from Chinese patient Ifnar1-/- mice peritubular myoid


returning from Fiji cells; epididymal
and Samoa, 2016) epithelial cells (EEC)
Testicular & epididymal Testes & epididymis qRT-PCR 8, 16 & 30
cells

Mouse-adapted ZIKV infected C57BL6 Testicular, epididymal Testes, epididymis, & qRT-PCR 7, 14 & 42 76

Dakar 41519 mice + anti-Ifnar1 cells & sperm epididymal fluid


(Senegal, 1984) or H/ antibody Sperm Epididymis Plaque assay 7
PF/2013 (French Spermatogonia, Testes ISH 7 & 14
Polynesia, 2013)b primary
spermatocytes, & SC

Mouse-adapted Dakar ZIKV infected Axl-/- Indiscriminate All reproductive tract qRT-PCR 7
41519 (Senegal, mice + anti-Ifnar1 tissues
1984) antibody SC & GC Testes & epididymis ISH 7
ZIKV infected Rag-/- Indiscriminate All reproductive tract qRT-PCR 7
mice + anti-Ifnar1 tissues
antibody SC & GC Testes & epididymis ISH 13

American-derived ZIKV infected Leydig cells (LC) Interstitial space IHC 5 82

MEX2-81 (Mexico, Ifnar1-/- mice Sperm & EEC Epididymis IHC 5


2016) Isolated LC from WT or Interstitial space Plaque assay 1-5
Ifnar1-/- mice testesc
Testicular & epididymal Testes & epididymis qRT-PCR 5 & 21
cells

Paraiba_01/2015 ZIKV infected Rag1-/- GC (likely Seminiferous IHC & ISH 17 or 21 83

(Brazil, 2015) mice + anti Ifnar1 spermatogonia/ epithelium


antibody primary
spermatocytes)

ZIKV/Homo sapiens/ ZIKV infected Sperm Caudal epididymis qRT-PCR 7, 14 & 21 84

PRI/ Ifnar1-/- mice Sperm Caudal epididymis IF 28


PRVABC59/2015 SC & GC Seminiferous IHC 7, 14 & 21
(Puerto Rico, 2015) epithelium
EEC & cells in Epididymis IHC 7, 14 & 21
epididymal lumend

CAS-ZK01 (Asian ZIKV infected AG6 SC Seminiferous IF (in vivo & in 2, 5 & 8 85

strain) mice epithelium vitro)


Macrophages Interstitial space IF
Abbreviations: d.p.i., days post-infection; EEC, epididymal epithelial cells; SC, Sertoli cells; GC, germ cells, LC, Leydig cells; IF, immunofluorescence
analysis; qRP-PCR, quantitative reverse transcription-polymerase chain reaction; ISH, in situ hybridization; IHC,
immunohistochemistry.
a Days postappearance of symptoms.

b Dakar 41519 replicated to higher levels than H/PF/2013 strain and was used for majority of this study.

c Leydig cells (LCs) were infected after isolation.

d Cells in lumen contained sperm and degenerating/sloughed necrotic epithelial cells.


188  A look into the testis as a reservoir for HIV and ZIKV—A reproductive biologist’s perspective

developing drugs and treatments. It also remains to be 8. Dufour JM, Hemendinger R, Halberstadt CR
determined whether viruses that penetrate the SC BTB to et  al. Genetically engineered Sertoli cells are able
gain entry into the adluminal compartment of the seminif- to survive allogeneic transplantation. Gene Ther.
erous epithelium is mediated through transfection of SC 2004;11(8):694.694–700.
or is the result of paracellular transport of viral particles 9. Shamekh R, El-Badri NS, Saporta S, Pascual C,
at the BTB. Reports in the literature suggest that HIV is Sanberg PR, Cameron DF. Sertoli cells induce sys-
utilizing one of its exterior envelope glycoproteins, gp120, temic donor-specific tolerance in xenogenic trans-
to induce breakdown of tight junctions (TJs) between plantation model. Cell Transplant. 2006;15(1):45–53.
endothelial cells at the BBB.77 HIV gp120 also downregu- 10. Fallarino F, Luca G, Calvitti M et al. Therapy of
lated the expression of ZO-1 and ZO-2, two key adaptor experimental type 1 diabetes by isolated Sertoli cell
proteins that support TJ integral membrane proteins such xenografts alone. J Exp Med. 2009;206(11):2511–2526.
as occludin, claudins, and junctional adhesion molecules, 11. Mital P, Kaur G, Dufour JM. Immunoprotective Sertoli
thereby causing the TJ barrier to become “leaky,” facilitat- cells: Making allogeneic and xenogeneic transplanta-
ing their entry into the brain.77 Furthermore, HIV gp120 tion feasible. Reproduction. 2009;139(3):​495–504.
was also found to induce production of matrix metallo- 12. Yin Z, Chen D, Hu F et al. Cotransplantation with
proteinases (MMPs) in vaginal epithelial cells, leading to xenogenetic neonatal porcine Sertoli cells signifi-
degradation of TJ proteins in vaginal epithelium, thereby cantly prolongs islet allograft survival in nonim-
facilitating the passage of HIV across the vaginal epithelial munosuppressive rats. Transplantation. 2009;88(3):​
barrier.78 Additionally, human T-cell leukemia virus type 339–345.
1 (HTLV-1) was found to penetrate through a tight human 13. Garolla A, Pizzol D, Bertoldo A, Menegazzo M.
epithelial barrier via a transcytosis mechanism without Sperm viral infection and male infertility: Focus
involving any disruption or infection of the epithelium.79 on HBV, HCV, HIV, HPV, HSV, HCMV, and AAV.
This information is crucial since it presents evidence that J Reprod Immunol. 2013;100(1):20–29.
viruses have the ability to pass through tight tissue barri- 14. Jenabian M-A, Costiniuk CT, Mehraj V et al.
ers. These findings provide the foundation for developing Immune tolerance properties of the testicular tissue
future treatments to prevent viral entry through the BTB as a viral sanctuary site in ART-treated HIV-infected
and other tissue barriers. Perhaps the BTB can be tight- adults. AIDS. 2016;30(18):2777.
ened by manipulating signaling pathway(s) that regulate 15. Wu H, Zhao X, Wang F et al. Mouse testicular cell
it80 to deny entry of viral particles to the testis, eliminating type-specific antiviral response against mumps virus
a haven for these viruses. replication. Front Immunol. 2017;8:117.
16. Foresta C, Ferlin A, Bertoldo A. Human papilloma
CONFLICTS OF INTEREST AND ACKNOWLEDGEMENTS virus in the sperm cryobank: An emerging problem?
Conflicts of interest: Nothing to declare. J Androl. 2011;34(3):242–246.
This work was supported by grants from the National 17. Atkinson B, Thorburn F, Petridou C et al. Presence
Institutes of Health (NICHD R01 HD056034 to C.Y.C.; and persistence of Zika virus RNA in semen, United
U54 HD029990 Project 5 to C.Y.C.). Kingdom, 2016. Emerg Infect Dis. 2017;23(4):611–115.
18. Musso D, Roche C, Robin E, Nhan T, Teissier A, Cao-
REFERENCES Lormeau V-M. Potential sexual transmission of Zika
1. Medawar PB. Immunity to homologous grafted skin; virus. Emerg Infect Dis. 2015;21(2):359–361.
the fate of skin homografts transplanted to the brain, 19. Darcis G, Coombs RW, Lint C. Exploring the ana-
to subcutaneous tissue, and to the anterior chamber tomical HIV reservoirs: Role of the testicular tissue.
of the eye. Br J Exp Pathol. 1948:58–69. AIDS. 2016;30(18):2891.
2. Brent L. Transplantation tolerance—A historical 20. Cai L, Kato T, Nakayama M, Susa T, Murakami S.
introduction. Immunology. 2016;147(3):267–268. HSV type 1 thymidine kinase protein accumulation
3. Streilein WJ, Streilein WJ. Unraveling immune priv- in round spermatids induces male infertility by sper-
ilege. Science. 1995; 270(5239);1158–1160. matogenesis disruption and apoptotic loss of germ
4. Fijak M, Meinhardt A. The testis in immune privi- cells. Reprod Toxicol. 2009;27(1):14–21.
lege. Immunol Rev. 2006;213(1):66–81. 21. Wang T, Feng X, Han D. Mechanisms of testicular
5. Setchell BP, Setchell BP. The testis and tissue trans- immune privilege. Front Biol. 2011;6(1):19–30.
plantation: Historical aspects. J Reprod Immunol. 22. Meinhardt A, Hedger MP. Immunological, paracrine
1990;18(1):1–8. and endocrine aspects of testicular immune privi-
6. Head JR, Neaves WB, Billingham RE. Immune lege. Mol Cell Endocrinol. 2011;335(1):60–68.
privilege in the testis. Transplantation. 1983:36(4):​ 23. Dym M. Basement membrane regulation of Sertoli
423–431. cells. Endocr Rev. 1994;15:102–115.
7. Selawry HP, Cameron DF. Sertoli cell-enriched frac- 24. Siu MKY, Cheng CY. Dynamic cross-talk between
tions in successful islet cell transplantation. Cell cells and the extracellular matrix in the testis.
Transplant. 1993;2(2);123–129. BioEssays. 2004;26:978–992.
References 189

25. Dym M, Fawcett DW. The blood-testis barrier 42. Dejucq N, Chousterman S, Jegou B. The testicular
in the rat and the physiological compartmenta- antiviral defense system: Localization, expression,
tion of the seminiferous epithelium. Biol Reprod. and regulation of 2’5’ oligoadenylate synthetase,
1970;3(3):308–326. double-stranded RNA-activated protein kinase, and
26. Setchell BP. The functional significance of the blood- Mx proteins in the rat seminiferous tubule. J Cell
testis barrier. J Androl. 1980;1(1):3–10. Biol. 1997;139(4):865–873.
27. Franca LR, Auharek SA, Hess RA, Dufour JM. 43. Com E, Bourgeon F, Evrard B et al. Expression of
Morphofunctional and immunological aspects of the antimicrobial defensins in the male reproductive
blood-testis and blood-epididymal barriers. In Cheng tract of rats, mice, and humans. Biol Reprod. 2003;​
CY, ed. Biology and Regulation of Blood-Tissue Barriers. 68(1):95–104.
New York: Springer Science+Business Media, 2011. 44. Jin JY, Zhou L, Wang Y et al. Antibacterial and anti-
28. Cheng CY, Mruk DD. The blood-testis barrier and its viral roles of a fish beta-defensin expressed both in
implications for male contraception. Pharmacol Rev. pituitary and testis. PLoS One. 2010;5(12):e12883.
2012;64(1):16–64. 45. Blankson JN. Viral reservoirs and HIV-specific
29. Gilula NB, Fawcett DW, Aoki A. The Sertoli cell immunity. Curr Opin Intern Med. 2006;5(3):211–215.
occluding junctions and gap junctions in mature 46. Churchill M, Nath A. Where does HIV hide? A focus
and developing mammalian testis. Dev Biol. on the central nervous system. Curr Opin HIV AIDS.
1976;50(1):142–168. 2013;8(3):165–169.
30. Russell LD, Peterson RN. Sertoli cell junctions: 47. Le Tortorec A, Dejucq-Rainsford N. HIV infec-
Morphological and functional correlates. Int Rev tion of the male genital tract—Consequences for
Cytol. 1985;94:177–211. sexual transmission and reproduction. Int J Androl.
31. Pelletier RM, Byers SW. The blood-testis barrier 2010;33(1):108.
and Sertoli cell junctions: Structural considerations. 48. Paranjpe S, Craigo J, Patterson B et al. Subcom­
Microsc Res Techn. 1992;20(1):3–33. partmentalization of HIV-1 quasispecies between
32. Cheng CY, Mruk DD. Cell junction dynamics in the seminal cells and seminal plasma indicates their
testis: Sertoli-germ cell interactions and male contra- origin in distinct genital tissues. AIDS Res Hum
ceptive development. Physiol Rev. 2002;82(4):​825–74. Retroviruses. 2002;18(17):1271–1280.
33. Vogl AW, Vaid KS, Guttman JA. The Sertoli cell cyto- 49. Byrn RA, Kiessling AA. Analysis of human immu-
skeleton. In Cheng CY, ed. Molecular Mechanisms in nodeficiency virus in semen: Indications of a geneti-
Spermatogenesis. New York: Springer. 2009;186–211. cally distinct virus reservoir. J Reprod Immunol.
34. Fijak M, Bhushan S, Meinhardt A. Immunoprivileged 1998;41(1–2):161–176.
sites: The testes. In Cuturi M C, Anegon I, eds. 50. Baeten JM, Kahle E, Lingappa JR. Genital HIV-1
Suppression and Regulation of Immune Responses. RNA predicts risk of heterosexual HIV-1 transmis-
Methods in Molecular Biology. New York: Springer sion. Sci Transl Med. 2011;3(77):77ra29.
Science+Business Media, 2011;677:459–470. 51. Costiniuk CT, Kovacs C, Routy JP. Short commu-
35. Yule TD, Montoya GD, Russell LD, Williams TM, nication: Human immunodeficiency virus rebound
Tung KS. Autoantigenic germ cells exist outside in blood and seminal plasma following discon-
the blood testis barrier. J Immunol. 1988;141(4):​ tinuation of antiretroviral therapy. AIDS Res Hum
1161–1167. Retroviruses. 2013;29(2):266–269.
36. Cheng YH, Wong EWP, Cheng CY. Cancer/testis 52. Santangelo PJ, Rogers KA, Zurla C et al. Whole-body
(CT) antigens, carcinogenesis and spermatogenesis. immunoPET reveals active SIV dynamics in viremic
Spermatogenesis. 2011;1(3):209–220. and antiretroviral therapy-treated macaques. Nat
37. Simpson AJG, Caballero OL, Jungbluth A, Chen Y-T, Methods. 2015;12(5):427–432.
Old LJ. Cancer/testis antigens, gametogenesis and 53. Piomboni P, Baccetti B. Spermatozoon as a vehicle
cancer. Nat Rev Cancer. 2005;5(8):615–25. for HIV-1 and other viruses: A review. Mol Reprod
38. Caballero OL, Chen YT. Cancer/testis (CT) antigens: Dev. 2000;56(S2):238–242.
Potential targets for immunotherapy. Cancer Sci. 54. Pudney J, Nguyen H, Xu C, Anderson DJ. Micro­
2009;100(11):2014–2021. scopic evidence against HIV-1 infection of germ
39. Kalejs M, Erenpreisa J. Cancer/testis antigens and cells or attachment to sperm. J Reprod Immunol.
gametogenesis: A review and “brain-storming” ses- 1998;41(1–2):105–125.
sion. Cancer Cell Int. 2005;5(1):1–11. 55. Baccetti B, Benedetto A, Collodel G, Caro DA. The
40. Tung KS, Teuscher C, Meng AL. Autoimmunity debate on the presence of HIV-1 in human gametes.
to spermatozoa and the testis. Immunol Rev. 1981;​ J Reprod Immunol. 1998;41(1–2):41–67.
55(1):217–255. 56. Ashida ER, Scofield VL. Lymphocyte major histo-
41. Dejucq N, Lienard MO, Jegou B. Interferons and compatibility complex-encoded class II structures
interferon-induced antiviral proteins in the testis. may act as sperm receptors. Proc Natl Acad Sci U S A.
J Reprod Immunol. 1998;41(1–2):291–300. 1987;84(10):3395–3399.
190  A look into the testis as a reservoir for HIV and ZIKV—A reproductive biologist’s perspective

57. Bagasra O, Freund M, Weidmann J, Harley G. 72. Barzon L, Pacenti M, Franchin E et al. Infection
Interaction of human immunodeficiency virus with dynamics in a traveller with persistent shedding
human sperm in vitro. J Acquir Immune Defic Syndr. of Zika virus RNA in semen for six months after
1988;1(5):431–435. returning from Haiti to Italy, January 2016. Euro
58. Cardona-Maya W, Velilla PA, Montoya CJ. In vitro Surveill. 2016;21(32).
human immunodeficiency virus and sperm cell 73. Grant A, Ponia SS, Tripathi S et al. Zika virus targets
interaction mediated by the mannose receptor. human STAT2 to inhibit type i interferon signaling.
J Reprod Immunol. 2011;92(1–2):1–7. Cell Host Microbe. 2016;19(6):882–890.
59. Shevchuk MM, Nuovo GJ, Khalife G. HIV in testis: 74. Kumar A, Hou S, Airo AM et al. Zika virus inhibits
Quantitative histology and HIV localization in germ type-I interferon production and downstream sig-
cells. J Reprod Immunol. 1998;41(1–2):69–79. naling. EMBO Rep. 2016;17(12):1766–1775.
60. Muciaccia B, Filippini A, Ziparo E, Colelli F, Baroni 75. Ma W, Li S, Ma S et al. Zika virus causes testis
CD, Stefanini M. Testicular germ cells of HIV- damage and leads to male infertility in mice. Cell.
seropositive asymptomatic men are infected by the 2016;167(6):1511–1524.
virus. J Reprod Immunol. 1998;41(1–2):81–93. 76. Govero J, Esakky P, Scheaffer SM et al. Zika virus
61. Muciaccia B, Uccini S, Filippini A et al. Presence and infection damages the testes in mice. Nature.
cellular distribution of HIV in the testes of seroposi- 2016;540(7633):438–442.
tive subjects: An evaluation by in situ PCR hybrid- 77. Nakamuta S, Endo H, Higashi Y et al. Human
ization. FASEB J. 1998;12(2):151–163. immunodeficiency virus type 1 gp120-mediated
62. Kim K, Shresta S. Neuroteratogenic viruses and disruption of tight junction proteins by induction
lessons for Zika virus models. Trends Microbiol. of proteasome-mediated degradation of zonula
2016;24(8):622–636. occludens-1 and -2 in human brain microvascular
63. White MK, Wollebo HS, Beckham DJ, Tyler KL, endothelial cells. J Neurovirol. 2008;14:186–195.
Khalili K. Zika virus: An emergent neuropathologi- 78. Fanibunda SE, Modi DN, Gokral JS, Bandivdekar
cal agent. Ann Neurol. 2016;80(4):479–489. AH. HIV gp120 binds to mannose receptor on
64. Mlakar J, Korva M, Tul N et al. Zika virus associated vaginal epithelial cells and induces produc-
with  microcephaly. N Engl J Med. 2016;374(10):​ tion of matrix metalloproteinases. PLoS One.
951–958. 2011;6:e28014.
65. WHO. Zika situation report [Internet]. Geneva: 79. Martin-Latil S, Gnadig NF, Mallet A et al.
World Health Organization; 2016 [updated Nov 24, Transcytosis of HTLV-1 across a tight human epithe-
2016. Available from: http://www.who.int/emergencies​ lial barrier and infection of subepithelial dendritic
/zika-virus/situation-report/24-november-2016/en/. cells. Blood. 2012;120:572–580.
66. Deckard TD, Chung WM, Brooks JT et al. Male- 80. Wen Q, Tang EI, Gao Y et al. Signaling pathways
to-male sexual transmission of Zika virus—Texas, regulating blood-tissue barriers—Lesson from
January 2016. MMWR Morbid Mortal Wkly Rep. the testis. Biochim Biophys Acta—Biomembranes.
2016;65(14):372–374. 2018;1860(1):141–153. DOI:101016/jbbamem201704020.
67. Hills SL, Russell K, Hennessey M et al. Transmission 81. Mansuy JM, Suberbielle E, Chapuy-Regaud S et al.
of Zika virus through sexual contact with travelers to Zika virus in semen and spermatozoa. Lancet Infect
areas of ongoing transmission – continental United Dis. 2016;16(10):1106–1107.
States, 2016. MMWR Morbid Mortal Wkly Rep. 82. Uraki R, Hwang J, Jurado KA et al. Zika virus causes
2016;65(8):215–216. testicular atrophy. Sci Adv. 2017;3(2):e1602899.
68. Brasil P, Pereira JP Jr, Moreira ME et al. Zika virus 83. Winkler CW, Myers LM, Woods TA et al.
infection in pregnant women in Rio de Janeiro. Adaptive immune responses to Zika virus are
N Engl J Med. 2016;375(24):2321–2334. important for controlling virus infection and pre-
69. D’Ortenzio E, Matheron S, Yazdanpanah Y et al. venting infection in brain and testes. J Immunol.
Evidence of sexual transmission of Zika virus. 2017;198(9):3526–3535.
N Engl J Med. 2016;374(22):2195–2198. 84. Griffin BD, Muthumani K, Warner BM et al.
70. Russell K, Hills SL, Oster AM et al. Male-to-female DNA vaccination protects mice against Zika
sexual transmission of Zika virus—United States, virus-induced damage to the testes. Nat Commun.
January–April 2016. Clin Infect Dis. 2017;64(2):211–213. 2017;8:15743.
71. Morrison TE, Diamond. Animal models of Zika 85. Sheng ZY, Gao N, Wang ZY et al. Sertoli cells are
virus infection, pathogenesis, and immunity. J Virol. susceptible to ZIKV infection in mouse testis. Front
2017;91:e00009-17. Cell Infect Microbiol. 2017;7:272.
Cytoskeletons (F-actin)
and spermatogenesis
15
LIZA O’DONNELL and PETER G. STANTON

INTRODUCTION via association with various microtubule-associated proteins


Spermatozoa are produced during the complex and (Figure 15.1b), (reviewed in Pollard 2016).1 The complex-
elegant process of spermatogenesis. These specialized ity of actin structures is further increased by branching,
cells arise from immature diploid spermatogonia, which facilitated by the Arp2/3 complex. The Arp2/3 complex,
undergo meiosis to produce haploid gametes. The sperm in association with nucleation promoting factors such
structure develops during spermiogenesis, where a round as WASH and WASP, binds to the side of a filament and
haploid germ cell (round spermatid) is transformed into nucleates a daughter branch,5 whereas cortactin stabilizes
a spermatozoon by the development of a motile cilium, a the branches (Figure 15.1b).
streamlined nucleus, and an acrosomal cap required for Actin filaments and higher-order structures are innately
fertilization. All aspects of male germ cell development are dynamic, possessing elastic and mechanical properties
supported by the somatic Sertoli cell; this large and struc- that allow them to fulfill multiple roles within the cell;
tural complex cell simultaneously supports multiple germ such properties are influenced by actin’s association with
cells at different phases of development, providing both a wide range of binding proteins.1,6 Along with profilin,
the structural and nutritional support required to execute F-actin elongation can be regulated by formins, homodi-
the spermatogenic program. Spermatogenesis is highly meric proteins that bind to the barbed ends of F-actin
dynamic and precisely controlled in space and time and is and promote further polymerization by stabilizing actin
accompanied by extraordinary changes in each cell’s cyto- dimers. The growth of F-actin barbed ends can be inhib-
skeleton. This chapter will address the role of a particular ited by actin capping proteins, whereas tetramers of Ena/
cytoskeletal component, actin, in spermatogenesis. VASP proteins facilitate elongation by inhibiting actin
Actin is one of the most abundant proteins in cells and capping. F-actin dynamics are further regulated by sever­
indeed on Earth.1 The amino acid sequence of actin mono- ing of the filaments (Figure 15.1b) via the small protein
mers (also known as globular actin or G-actin) is highly cofilin and the larger protein complex gelsolin, whereas
conserved between species. There are three main isoforms the actin-associated protein tropomyosin can protect fila-
of actin monomers, alpha, beta, and gamma, encoded by ments from severing. Cofilin can sever filaments or inhibit
different genes. Actin monomers are polymerized into their elongation but can also promote filament assembly
filamentous actin (F-actin), which in turn is arranged into at higher concentrations.1,7 A multitude of actin cross-
higher-order structures via interactions with numerous linking proteins are also essential for the organization of
actin binding and regulatory proteins. Actin participates F-actin into higher-order structures (Figure 15.1b). These
in many cellular functions, including division, migration, linking proteins are rapidly exchanged, giving rise to the
polarity, shape, intercellular adhesion (including cell-cell unique properties of F-actin networks; these networks are
and cell-substrate adhesion), and intracellular transport. relatively stiff when rapidly deformed but can be deformed
Emerging evidence has revealed actin can also func- over a longer time scale (tens of seconds) due to exchange
tion within the nucleus, regulating gene expression and of linking proteins. This explains why cells are stiff and
responding to extracellular cues.2,3 elastic when subject to a rapid force but can be deformed
by longer periods of applied pressure.1
F-ACTIN BIOGENESIS, DYNAMICS, AND FUNCTION Actin generates forces that underlie many cellular
Actin monomers are relatively flat proteins with a cleft functions and provides a signal-responsive cytoskeletal
that  binds tightly to ATP, which is hydrolyzed during system that responds to many cues. While polymeriza-
polymerization. Monomers spontaneously dimerize then tion of actin itself can generate force, many cellular move-
polymerize into a helical single-stranded filament (Figure ments and force generation involve F-actin binding to
15.1a). Due to the orientation of actin monomers at each myosins, a family of ATPase motor proteins. Myosins are
end, F-actin has a “pointy end,” which is slow to polymer- large heterodimeric proteins consisting of heavy and light
ize, and a “barbed end,” which rapidly polymerizes and chain subunits. They possess a globular head that binds
grows in length (Figure 15.1). Profilin is a small protein to F-actin and has ATPase activity and a tail region that
that plays a major role in actin polymerization via binding confers specificity for different binding proteins. Upon
to monomers (Figure 15.1a) and to the polymerized fila- ATP hydrolysis, the head region undergoes a conforma-
ment and cytoskeletal proteins.4 Once polymerized, actin tional change, allowing it to “walk” along the actin fila-
filaments can be cross-linked into higher-order structures ment while moving vesicles or other proteins associated

191
192  Cytoskeletons (F-actin) and spermatogenesis

ATP binding
cleft

Pointed end Barbed end


(slow (rapid Profilin
Barbed end polymerization) polymerization)
groove
Actin Actin
(a) monomer filament

Cortactin
Cofilin

N-WASP

Arp2/3 Actin-
Formins capping
proteins
(b) Branching Elongation Severing Cross-linking and bundling

Figure 15.1  Actin filament biogenesis and dynamics. (a) Actin monomer and filament structure and profilin’s role in regulat-
ing actin polymerization by binding to the monomer pool. Actin monomers contain an ATP-binding cleft and a so-called barbed
end that binds to target proteins. The polymerization of the actin monomers, facilitated by profilin, creates a helical filament with
a slowly polymerizing end and a rapidly polymerizing end that can grow rapidly in length. (b) Overview of the regulation of actin
dynamics by interacting proteins. Branched filaments are created by the Arp2/3 complex in association with other nucleation pro-
moting factors such as N-WASP, and the branches are stabilized by interactions with cortactin. Actin filament elongation is promoted
by formins, but can be inhibited by actin capping proteins. Filaments can be severed (e.g., by cofilin) and arranged into various
higher-order structures by a multitude of actin-binding and bundling proteins.

with the tail region. Myosin activity is regulated by the Actin-binding formins appear to be a key player in the
phosphorylation of the regulatory light chain subunit.8,9 cross talk between the actin and microtubule cytoskel-
The forces generated by actin filaments and networks, eton, with an ability to regulate both actin polymerization
as well as those generated during myosin-mediated move- and microtubule stabilization.13
ments, create a mechanically-sensitive dynamic state, From the above it is clear that the actin cytoskeleton
reviewed in De La Cruz and Gardel 2015.10 This mechani- plays multiple roles in all cells and thus the actin cytoskel-
cally sensitive state allows the cell to respond to external eton will drive many aspects of spermatogenesis, such as
stimuli, for example to shear stress, but also to respond the extraordinary changes in Sertoli and germ cell shape
to internal cues such as forces generated within the cyto- as they proceed through the spermatogenic cycle. The fol-
skeleton itself. Depending on the type of filament cross- lowing section will provide an overview of the actin cyto-
linking and the abundance of myosin motors, the actin skeleton in spermatogenesis, highlighting the role of actin
network can be arranged into various higher-order struc- dynamics in particular processes in the seminiferous epi-
tures with different physical properties. For example, cell thelium. Other reviews14–18 also address this topic.
motility is achieved by the polymerization of branched
actin filaments in lamellipodia effectively pushing the
F-ACTIN IN SPERMATOGENIC CELLS
leading edge of a migrating cell forward. Actin networks
can be contractile; actomyosin (actin + myosin complex)- F-actin in spermatogonia and meiotic cells
mediated contractility is involved in many cellular func- Spermatogenesis originates from the spermatogonial stem
tions such as exo- and endocytosis and cell shape changes. cell (SSC) niche. SSCs both renew their own population to
Actin networks can also be organized into ring structures; provide a lifelong supply of stem cells and differentiate to
constricting actomyosin rings create the cleavage fur- become spermatogonia that ultimately develop into sper-
row between dividing cells and facilitate cytokinesis, and matozoa; the establishment and maintenance of this niche
stabilizing actin rings are components of cell adhesion is the foundation of successful spermatogenesis. Various
junctions.11 lines of evidence suggest that the actin cytoskeleton is
Finally, it should also be noted that there is extensive involved in maintaining the SSC niche. Spermatogonial
cross talk between the actin and microtubule cytoskel- transplantation experiments show that the ability of SSCs
etons.12 For example, while cell motility requires actin to colonize the niche is dependent on their ability to express
polymerization in membrane protrusions, directional cell β1-integrin, which links the actin cytoskeleton with the
movement involves growing microtubules being captured extracellular matrix.19 Glial cell line-derived neurotrophic
by the cortical actin foci at the leading edge of the cell.12 factor (GDNF), secreted by Sertoli cells, is a key regulator
F-actin in spermatogenic cells  193

of the SSC niche, regulating the migration, differentiation, profilins,34 and a novel isoform of αT-catenin35 that can
and self-renewal of SSCs. GDNF induces the expression regulate actin cytoskeleton dynamics. Therefore, it is pos-
of VASP in undifferentiated spermatogonia, suggesting sible that spermatids have evolved a novel F-actin machin-
that VASP-dependent changes in the actin cytoskeleton ery that may be functionally different to other actin-based
influences the ability of SSC to migrate into the niche.20 cytoskeletal structures.
BMP4, another factor that regulates SSC differentiation,21 Male germ cell division is incomplete and germ cells
stimulates SSCs to transcribe genes involved in the actin at various stages of development remain attached to one
cytoskeleton and actin-dependent adhesion junctions.22 another by intercellular bridges that allow the movement
AIP1, an actin disassembly factor, is required in both of cytoplasmic contents between cells. These bridges are
spermatogonia and Sertoli cells for spermatogonial migra- required for germ cell development36 and facilitate shar-
tion and establishment of the SSC niche in mice,23 and ing of gene products between adjacent cells;37 this is likely
profilin-dependent stabilization of F-actin is required for important for synchronous development of cohorts of hap-
the maintenance of SSCs in Drosophila.24 Therefore, main- loid spermatids. Intercellular bridges contain actin,18,36,38
tenance of the SSC niche involves the regulation of actin- which is essential for their maintenance, as disruption of
dependent adhesion and cytoskeletal structures. actin filaments dissolves the bridges38 ultimately resulting
Differentiating spermatogonia ultimately enter meiosis, in the formation of multinuclear symplasts followed by
the process that generates haploid, genetically diverse gam- germ cell death.
etes. Early in meiosis, preleptotene spermatocytes must Round spermatids employ sophisticated mechanisms
traverse the blood-testis-barrier to complete their develop- to control gene transcription and translation during sper-
ment within a specialized microenvironment; their trans- miogenesis.39,40 Such mechanisms are required to execute
port across this barrier is dependent on F-actin dynamics the spermiogenic program as active gene transcription
within Sertoli cells (see section “Dynamics of Sertoli cell ceases midway through spermiogenesis when the sperma-
F-actin-containing structures during spermatogenesis”). tid nucleus compacts. Interestingly, an actin-related pro-
During the first meiotic prophase, homologous chromo- tein, ARPC5, a member of the Arp2/3 complex proteins,
somes pair, form synaptonemal complexes, and undergo has been shown to participate in translational repression
recombination to ensure genetic diversity of the male during spermiogenesis,41 and the F-actin cytoskeleton is
germline. Actin and myosin-dependent processes are likely associated with translational repression mechanisms in
involved in synaptonemal complex formation. Ablation other systems.42 The chromatoid body in round spermatids
of ALKBH4, a protein that influences F-actin’s ability to is an organizing center for RNA and facilitates noncod-
associate with nonmuscular myosin II, caused failure of ing RNA-dependent translational silencing pathways.39
synaptonemal complexes to form, resulting in spermato- Although the chromatoid body contains actin,43 it remains
cytes entering apoptosis and failing to complete meiosis.25 intact after cytochalasin D-induced actin disassembly,37
Interestingly, nonmuscular myosin II is also required for suggesting actin dynamics might not be required for the
the completion of cytokinesis at the end of meiosis.26 The maintenance of chromatoid body structure. Spermatids
movement of chromosomes during prophase is thought also express novel actin proteins in the nucleus during
to be reliant on microtubule-based processes;27 however, elongation.31,44 Given the abundant evidence for actin and
there is some evidence that actin is involved in chromo- its associated proteins operating within the nucleus to reg-
some positioning in late diakinesis just prior to anaphase.28 ulate transcription, translation, and chromatin dynam-
ics,3,42,45,46 the role of nuclear F-actin in gene transcription
F-actin during spermiogenesis and chromatin remodeling during spermiogenesis war-
After the last meiotic division, haploid spermatids enter rants further study.
spermiogenesis as small, round, very transcriptionally active During early spermiogenesis, round spermatids develop
cells, yet they finish the process as long, transcriptionally the acrosome, a vesicle containing proteolytic enzymes
inert spermatozoa with dense, characteristically shaped required to penetrate and fertilize the oocyte. Acrosome
nuclei and long flagella capable of motility. This transfor- development involves F-actin dynamics, as the acrosome
mation is accompanied by extraordinary cytoskeletal and fails to spread across the nucleus after treatment with
nuclear changes, in which actin dynamics play key roles. cytochalasin D.18 Acrosome development is accomplished
There is abundant evidence that spermatids specifically by F-actin–rich structures present in the subacrosomal
express novel transcripts of components of the F-actin space; these structures have been termed the acroplax-
cytoskeleton. Spermatids specifically express novel actin ome (an actin-based scaffold that anchors the acrosome to
monomers29–31 that only share 40%–50% identity with the nuclear membrane) and the marginal ring (an actin-
conventional actin genes but retain conserved functional rich structure at the leading edge of the spreading acro-
motifs; these intron-less genes are conserved in rodents some).47,48 The acrosome arises from vesicles in the Golgi;
and humans, have unique regulatory elements, and may these vesicles coalesce and spread across the nuclear sur-
have arisen from retrotransposons during evolution.29 face in a process facilitated by the actin motor myosin Va
Sperm uniquely express novel actin-related proteins that together with its receptor RAB27A/B.49 Proteins involved
form a major constituent of the sperm head32 as well as in the regulation of F-actin dynamics are present in the
a unique actin-capping protein,33 novel testis-specific subacrosomal space and acroplaxome and may regulate
194  Cytoskeletons (F-actin) and spermatogenesis

acrosome development, including FerT, profilin III and spermiation. These actin-based structures within Sertoli
IV, and phosphorylated cofilin, reviewed in Kierszenbaum cells perform various essential functions required for suc-
et al. 2007.47 cessful sperm production.
As spermiogenesis progresses, the nucleus polarizes to
one side and the spermatid begins to elongate, with the F-actin in ectoplasmic specializations
nucleus undergoing extensive condensation and remod- The Sertoli cell ectoplasmic specialization (ES) consists
eling. The perinuclear theca is a narrow, condensed layer of a layer of hexagonally packed actin filaments recog-
of cytoskeleton surrounding the spermatid nucleus as it nizable by electron microscopy61,62 located between the
elongates,50 extending beyond the acrosome as the post- plasma membrane and a cistern of endoplasmic reticu-
acrosomal sheath or calyx.51 This calyx contains short lum. Opposing ES structures are found at intercellular
F-actin filaments bound to the protein calicin, thought to junctions between Sertoli cells near their basal aspect and
impart a structural rigidity,52 and other actin-associated are referred to as basal ES (Figure 15.2); these comprise
proteins, reviewed in Toshimori 2003.53 The spermatid an important part of the BTB (see section “Dynamics of
nucleus is remodeled into its species-specific shape via the Sertoli cell F-actin-containing structures during sper-
microtubule-based manchette. 54,55 However, F-actin- matogenesis”). In contrast, single ES structures are seen in
based mechanisms operating within the manchette and Sertoli cells opposite elongating spermatids (Figure 15.2);
acroplaxome, accompanied by “sculpting forces” from the these are referred to as apical ES.
actin-based Sertoli cell ectoplasmic specialization (see The ES has adhesive properties because the intercellular
section “F-actin in ectoplasmic specializations”), are gap is narrow and contains adhesion proteins such as α6β1
also proposed to participate in nuclear shaping during integrin,63 nectin-2,64 and N-cadherin.65 The basal ES forms
spermiogenesis.17,47 Toward the end of spermatid develop- part of the inter-Sertoli–cell basal junctional complex; var-
ment and in epididymal sperm, F-actin is found in specific ious other junction types (tight-, gap-, ­desmosome-like-)
regions of the perinuclear theca in the sperm head56 along are also present at this site and have links to the actin cyto-
with a testis-specific isoform of an actin-­bundling protein, skeleton66,67 (Figure 15.2). The adhesive function of this
FSCN3.57 As spermatids continue through spermiogen- junctional complex is spatiotemporally regulated to allow
esis, however, F-actin may depolymerize to G-actin and restructuring when basal spermatocytes translocate into
become redistributed.18 Interestingly, F-actin in the sperm the adluminal c­ ompartment (Figure 15.2).
head increases during capacitation and the acrosome reac- Various actin-binding proteins are found at the basal ES,
tion, suggesting that polymerization of monomeric actin with functions including actin bundling and cross-linking
is involved in the activation of mature spermatozoa.58 (espin, ezrin, fascin 1, palladin, plastin family), branched
Therefore, F-actin is present in the sperm head and may actin polymerization (Arp2/3 complex, N-WASP), end-
play multiple roles in sperm head development and fertil- capping (Eps8), and branch stabilization (cortactin).68,69
izing ability. Selective RNAi-mediated knockdown of individual actin-
Finally, F-actin may play a role in the development of binding protein expression tends to disturb Sertoli cell
the spermatid flagella. Flagella biogenesis begins in round junction function in vitro as well as F-actin and associ-
spermatids, with the formation and extension of the ated junction protein organization (e.g., targeting fascin
microtubule-based axoneme. Sperm-specific accessory 170). However, under these circumstances ES function is
structures such as outer dense fibers and the mitochon- not completely ablated, suggesting that multiple proteins
drial sheath are then assembled around the axoneme in contribute to actin dynamics at the basal ES. In contrast,
elongating spermatids. This latter stage of development targeted disruption of the branched actin polymerization
is proposed to involve the transport of proteins along the pathway by knockout of N-Wasp (which acts upstream
manchette to the developing flagellum.59 While the man- of Arp2/3) in mouse Sertoli cells caused spermatogenic
chette is largely microtubule-based, small tracks of F-actin arrest, loss of expression of connexin-43 and occludin
are visible within the microtubule bundles and contain (markers for gap and tight junctions, respectively), and
myosin-based vesicles,49 testis-specific profilins34 and the loss of BTB function.71 These studies clearly show con-
actin regulatory protein palladin.60 Current theories sug- trol of branched actin polymerization to be an essential
gest that the transport of vesicles along the manchette component of the maintenance of seminiferous epithelial
microtubule and F-actin tracks delivers at least some of structure and function.
the proteins required for flagella assembly.59 In contrast to basal inter-Sertoli cell junctions, apical
ES junctions between Sertoli cells and advanced sperma-
F-ACTIN IN SERTOLI CELLS tids are heterotypic and consist of ES located only on the
The cytoskeleton in Sertoli cells is comprised of actin fila- Sertoli cell side; no junctional structures are discernible
ments, intermediate filaments, and microtubules. F-actin in the spermatid. The apical ES has much the same struc-
tends to be concentrated at intercellular junctions between ture as the basal ES, comprised of hexagonally packed
adjacent Sertoli cells where they contribute to the blood- actin filaments located between the Sertoli cell plasma
testis barrier (BTB), and at junctions between Sertoli cells membrane and an underlying endoplasmic reticulum.
and elongating spermatids (step 8 and onward in rats In the rat, the apical ES first appears in Sertoli cells adja-
and mice) prior to their release from the epithelium at cent to round spermatids as they polarize at step 8, and
F-actin in Sertoli cells  195

5
6

3
Adluminal
4 4
4 7
2 Sertoli cell
8
9

1 Basal

10

Figure 15.2  Summary of the major roles of F-actin structures (green) in the seminiferous epithelium. In certain Sertoli cell struc-
tures, F-actin is located near cisternae of endoplasmic reticulum (denoted in red). 1. Actin dynamics are involved in the migration of
spermatogonia and the establishment of the spermatogonial stem cell niche. 2. Actin dynamics are proposed to be involved in the
formation of synaptonemal complexes between meiotic chromosomes. 3. In early spermiogenesis, actin is involved in formation of
the acrosome and its attachment to the nucleus, as well as in maintaining intercellular bridges. 4. During the elongation phase of
spermiogenesis, F-actin gradually disappears from the perinuclear region of the spermatid head, but abundant F-actin is present
in large bundles in the Sertoli cell cytoplasm adjacent to the spermatid head in an adhesion structure known as the ectoplasmic
specialization (ES). 5. At the end of spermiogenesis, the actin bundles and the ES are disassembled, coincident with the formation
of tubulobulbar complexes (TBCs). TBCs are surrounded by dendritic actin and are involved in the removal of adhesion junction
components and remodeling of the spermatid. 6. Just prior to sperm release, spermatids remain attached to the Sertoli cell by an
integrin-based focal adhesion type junction that may associate with cytoplasmic actin filaments. 7. Transport and phagocytosis of
the residual body (the remainder of the spermatid cytoplasm) is thought to rely on actin dynamics. 8. Inter-Sertoli cell junctions
include ES junctions with opposing bundles of actin, TBCs, and other adhesion junctions (gray bars) including desmosome-like
and occluding junctions. 9. As early spermatocytes translocate into the adluminal compartment, the inter-Sertoli cell junctions are
remodeled, disappearing above and reappearing below the spermatocytes as they migrate. 10. Peritubular myoid cells surrounding
the seminiferous tubules contain abundant actin filaments; the actin-based contractility of these cells is important for the contrac-
tion of the tubules as they propel released sperm toward the epididymis. The tight inter-Sertoli cell junctions (8) effectively divide
the seminiferous epithelium into a basal and adluminal compartment (denoted by the blue lines); germ cells in the basal compart-
ment have free access to factors from the interstitium whereas germ cells in the adluminal compartment develop in a specialized
microenvironment determined by the Sertoli cell.

remains associated with elongating spermatids until it is so-called bulbous structure, surrounded by endoplasmic
removed during the process of spermiation.72 The apical reticulum but not actin, which extends into a clathrin-
ES functions to firmly attach the elongating spermatid coated pit. Many studies have shown that TBCs are a
to Sertoli cells, which in turn draws them deep into the novel endocytic structure similar to podosomes, and their
epithelium as part of the spermatid maturation process.62 major function is the removal and recycling of intercel-
The orientation and polarity of adluminal spermatids is lular junctions.69 TBCs are readily found at apical junc-
likely maintained by F-actin dynamics at the apical ES, tions between Sertoli cells and advanced spermatids and
as knockdown of various proteins involved in F-actin are well described at this site.69 These structures form on
dynamics leads to the loss of correct positioning sper- the ventral side of the spermatid nucleus at the begin-
matid in the epithelium and ultimately in their failure to ning of spermiation (stage VII in rodents) and they persist
complete spermatogenesis.73,74 throughout spermiation, where they are involved in the
removal of ES and other junction-associated proteins as
F-actin in tubulobulbar complexes well as remodeling the spermatid plasma membrane prior
The tubulobulbar complex (TBC) is a novel structure to its release into the lumen.69
comprised of a tubular extension of plasma membranes TBCs are also present in the basal junctional complex
surrounded by a cuff of cross-linked actin (Figure 15.2) between Sertoli cells 69; see Figure 15.2. Here, the long
to which various actin-binding proteins are localized. At tubular extension from one Sertoli cell projects into an
the end of this F-actin-associated tubular extension is a invagination in the neighboring cell, and the distal region
196  Cytoskeletons (F-actin) and spermatogenesis

including the clathrin-coated pit enlarges to form a bul- cell F-actin organization. Interestingly, palladin, which
bous structure before it buds off from the complex and is functions as a scaffold to bundle actin fibers, is found at
internalized via endocytic mechanisms.75 Various junc- the basal ES in vivo86 but FSH-mediated intercellular sig-
tion proteins, including claudin-11, connexin-43, and naling directs its localization to the Sertoli cell nucleus
nectin-2 are found within basal TBCs, and indeed whole where it may regulate transcription, potentially providing
tight and gap junction fragments have been observed a direct link between nuclear function and the status of
there by electron microscopy,75 highlighting the role of the F-actin cytoskeleton.87
basal TBCs in remodeling of inter-Sertoli-cell junctions. Remodeling of the BTB and inter-Sertoli-cell junctions
TBC formation and function depends on F-actin is required for early spermatocytes to enter the adlumi-
dynamics. Various actin-binding proteins are present in nal compartment and complete their development (Figure
the proximal cuff region of the TBC, including espin, cor- 15.2). This remodeling is accompanied by marked changes
tactin, cofilin, paxillin, vinculin, zyxin, Eps8, N-WASP, in F-actin-based structures, and the disturbance of vari-
and Arp2/3.69 These proteins are likely important for the ous proteins involved in F-actin organization can impair
formation, elongation, and maintenance of the tubu- both the structure and function of inter-Sertoli-cell junc-
lar portion of the TBC76,77; however, relatively little is tions.68,88 TBCs are most prevalent between Sertoli cells
known about their regulation. Knockdown of Arpc1b, prior to inter-Sertoli-cell junctions being remodeled to
an e­strogen-responsive subunit of the Arp2/3 complex allow germ cell translocation.89 The fact that these TBCs
involved in actin nucleation and branching, compro- contain morphologically discernible fragments of gap
mised BTB integrity in vivo.78 Both FSH and testosterone and tight junctions, as well as internalized junction-
regulate the expression of a panel of Sertoli cell miRNAs associated molecules, supports the hypothesis that TBC
in vitro that target proteins in the actin cytoskeleton and formation between Sertoli cells is important for the
focal adhesions.79 Importantly, disruption of F-actin net- remodeling of inter-Sertoli-cell junctions prior to germ
work using cytochalasin D caused the tubular regions of cell translocation.69
basal TBCs to become swollen with disorganized actin TBCs are also involved in junction removal between
foci, confirming the importance of actin networks in nor- Sertoli cells and spermatids during spermiation. Sper­
mal TBC function.76 miation is the process by which mature spermatids
undergo a final remodeling, including removal of their
Dynamics of Sertoli cell F-actin-containing extensive cytoplasm and remodeling of the nucleus, prior
structures during spermatogenesis to their disengagement or release from the Sertoli cell.72
The BTB is a functional barrier that sequesters meiotic In order for spermiation to occur, the extensive ES junc-
and postmeiotic germ cells into a controlled adlumi- tion present around the spermatid (Figure 15.2) needs to
nal environment separate from the immune system that be removed. The removal of ES likely involves the disas-
would otherwise see them as foreign. In broad terms the sembly of the extensive actin bundles by proteins involved
BTB has three components: the inter-Sertoli-cell junctions in F-actin dynamics (e.g., formin 190). In addition, TBCs
(mentioned above) that provide a physical barrier, plus likely remove ES structures. TBCs form and internal-
Sertoli cell transporters that control substance movement ize both Sertoli cell and spermatid plasma membranes
and create a physiological barrier, and an immunologi- together with the junction-associated proteins.69,91 The
cal barrier contributed by immunoregulatory factors and bulbar portion of the TBC then buds off and becomes
tolerance mechanisms.80,81 This barrier effectively divides internalized into the Sertoli cell as an endocytic vesicle; in
the seminiferous epithelium into two compartments: the this way TBCs are able to remove ES and associated adhe-
basal compartment, where germ cells “below” the inter- sion junctions between the Sertoli cell and the spermatid
Sertoli-cell junctions have free access to factors from the in preparation for its final disengagement.69,91 There are
interstitium and circulation, and the adluminal compart- now many lines of evidence to show that F-actin dynamics
ment, where germ cells reside “above” the barrier and control TBC formation and their ability to remove ES and
develop within a restricted environment (Figure 15.2). The other junction-associated proteins to ready the sperma-
barrier also enables maturing germ cells to develop within tid for disengagement. For example, disruption of F-actin
a unique specialized environment that is strictly regulated with cytochalasin D prevents TBC formation and normal
by the Sertoli cell. spermiation.92 The F-actin “cuffs” around the tubular por-
It is well known that the Sertoli cell responds to the hor- tion of TBCs contains various proteins involved in actin
mones FSH and testosterone with multiple structural and branching, including the Arp2/3 complex, N-WASP and
functional changes.40 The formation of the BTB and basal paxillin.69 Estradiol treatment reduces F-actin-associated
ES occurs concomitant with the pubertal rise in FSH and proteins within TBCs, causing defective TBC formation
LH.82 Androgen action on Sertoli cells is required for BTB and spermiation failure.78 Importantly, knockdown of
formation, whereas FSH action appears to be permissive cortactin, the protein that stabilizes actin branches, pre-
but not mandatory.82 However, FSH stimulation of F-actin vents normal TBC formation and impairs spermiation.93
organization into ES structures in Sertoli cells has consis- The release of mature sperm into the lumen is mediated
tently been observed in vitro83,84 and in vivo,85 highlight- by a focal adhesion type junction containing integrins and
ing the importance of FSH in promoting proper Sertoli focal adhesion kinase (FAK).73 Focal adhesions in other
References 197

systems link to, and are regulated by, the actin cytoskel- 8. Sackmann E. How actin/myosin crosstalks guide
eton, mediating instantaneous loss of adhesion events. the adhesion, locomotion and polarization of cells.
Thus, the actin cytoskeleton within the Sertoli cell at the Biochim Biophys Acta. 2015;1853(11 Pt B):3132–3142.
time of disengagement likely participates in spermatid 9. Woolner S, Bement WM. Unconventional myo-
disengagement, perhaps explaining why many inhibitors sins acting unconventionally. Trends Cell Biol.
of actin dynamics can impair sperm release.72,94 Finally, 2009;19(6):245–252.
when spermatids disengage, the remnants of their cyto- 10. De La Cruz EM, Gardel ML. Actin mechan-
plasm are left behind as the residual body (Figure 15.2). ics and fragmentation. J Biol Chem. 2015;290(28):​
The transport and phagocytosis of residual bodies may 17137–17144.
also rely on F-actin dynamics.95 11. Schwayer C, Sikora M, Slovakova J, Kardos R,
Heisenberg CP. Actin rings of power. Dev Cell. 2016;​
ROLE OF F-ACTIN IN SEMINIFEROUS TUBULE 37(6):493–506.
CONTRACTION 12. Goode BL, Drubin DG, Barnes G. Functional coop-
The seminiferous tubules are surrounded by one or more eration between the microtubule and actin cytoskel-
continuous layers of peritubular myoid cells.96 These cells etons. Current Opin Cell Biol. 2000;12(1):​63–71.
facilitate the contraction of seminiferous tubules to pro- 13. Chesarone MA, DuPage AG, Goode BL. Unleashing
pel sperm and fluid along the tubules toward the rete tes- formins to remodel the actin and microtubule cyto-
tis.96 Consistent with their contractile nature, peritubular skeletons. Nat Rev Mol Cell Biol. 2010;11(1):62–74.
myoid cells contain layers of actin filaments (along with 14. Cheng CY, Wong EW, Lie PP et al. Polarity proteins
myosin motors) oriented in different directions (longi- and actin regulatory proteins are unlikely partners
tudinal and circular) in rodent97,98 and human testis.99 that regulate cell adhesion in the seminiferous epi-
Contraction of the peritubular myoid cells is accompanied thelium during spermatogenesis. Histol Histopathol.
by marked rearrangements in the F-actin cytoskeleton,100 2011;26(11):1465–1474.
highlighting the fact that F-actin dynamics are important 15. Li N, Mruk DD, Tang EI et al. Formins: Actin nuclea-
for seminiferous tubule contractility. tors that regulate cytoskeletal dynamics during sper-
matogenesis. Spermatogenesis. 2015;5(2):​e1066476.
CONCLUSIONS 16. Su W, Mruk DD, Cheng CY. Regulation of actin
This chapter has highlighted the major roles for the F-actin dynamics and protein trafficking during spermato-
cytoskeleton during spermatogenesis. F-actin dynamics genesis—Insights into a complex process. Critical
are involved in many processes within germ cells, par- Rev Biochem Mol Biol. 2013;48(2):153–172.
ticularly in the dramatic remodeling of spermatids as they 17. Sun X, Kovacs T, Hu YJ, Yang WX. The role of actin
develop into spermatozoa. Sertoli cells contain numerous and myosin during spermatogenesis. Mol Biol Rep.
actin-based structures, some of which are unique to the 2011;38(6):3993–4001.
testis, that maintain the integrity of the seminiferous epi- 18. Vogl AW. Distribution and function of organized
thelium and support male germ cells through their dra- concentrations of actin filaments in mammalian
matic transformation into motile spermatozoa. spermatogenic cells and Sertoli cells. Int Rev Cytol.
1989;119:1–56.
REFERENCES 19. Kanatsu-Shinohara M, Takehashi M, Takashima S
1. Pollard TD. Actin and actin-binding proteins. Cold et al. Homing of mouse spermatogonial stem cells to
Spring Harb Perspect Biol. 2016;8(8). germline niche depends on beta1-integrin. Cell Stem
2. Louvet E, Percipalle P. Transcriptional control of Cell. 2008;3(5):533–542.
gene expression by actin and myosin. Int Rev Cell 20. Dovere L, Fera S, Grasso M et al. The niche-derived
Mol Biol. 2009;272:107–147. glial cell line-derived neurotrophic factor (GDNF)
3. Plessner M, Grosse R. Extracellular signaling cues induces migration of mouse spermatogonial stem/
for nuclear actin polymerization. Eur J Cell Biol. progenitor cells. PloS One. 2013;8(4):e59431.
2015;94(7–9):359–362. 21. Nagano M, Ryu BY, Brinster CJ, Avarbock MR,
4. Yarmola EG, Bubb MR. How depolymerization can Brinster RL. Maintenance of mouse male germ
promote polymerization: The case of actin and pro- line stem cells in vitro. Biol Reprod. 2003;68(6):​
filin. BioEssays. 2009;31(11):1150–1160. 2207–2214.
5. Pizarro-Cerda J, Chorev DS, Geiger B, Cossart P. The 22. Carlomagno G, van Bragt MP, Korver CM, Repping
diverse family of Arp2/3 complexes. Trends Cell Biol. S, de Rooij DG, van Pelt AM. BMP4-induced differ-
2017;27(2):93–100. entiation of a rat spermatogonial stem cell line causes
6. Dominguez R, Holmes KC. Actin structure and changes in its cell adhesion properties. Biol Reprod.
function. Annu Rev Biophys. 2011;40:169–186. 2010;83(5):742–749.
7. Andrianantoandro E, Pollard TD. Mechanism of 23. Xu J, Wan P, Wang M et al. AIP1-mediated actin
actin filament turnover by severing and nucleation disassembly is required for postnatal germ cell
at different concentrations of ADF/cofilin. Mol Cell. migration and spermatogonial stem cell niche estab-
2006;24(1):13–23. lishment. Cell Death Dis. 2015;6:e1818.
198  Cytoskeletons (F-actin) and spermatogenesis

24. Shields AR, Spence AC, Yamashita YM, Davies 37. Ventela S, Toppari J, Parvinen M. Intercellular
EL, Fuller MT. The actin-binding protein pro- organelle traffic through cytoplasmic bridges in early
filin is required for germline stem cell mainte- spermatids of the rat: Mechanisms of haploid gene
nance and germ cell enclosure by somatic cyst cells. product sharing. Mol Biol Cell. 2003;14(7):2768–2780.
Development. 2014;141(1):73–82. 38. Russell LD, Vogl AW, Weber JE. Actin localization
25. Nilsen A, Fusser M, Greggains G, Fedorcsak P, in male germ cell intercellular bridges in the rat and
Klungland A. ALKBH4 depletion in mice leads to ground squirrel and disruption of bridges by cyto-
spermatogenic defects. PloS One. 2014;9(8):e105113. chalasin D. Am J Anat. 1987;180(1):25–40.
26. Yang F, Wei Q, Adelstein RS, Wang PJ. Non-muscle 39. de Mateo S, Sassone-Corsi P. Regulation of sper-
myosin IIB is essential for cytokinesis during matogenesis by small non-coding RNAs: Role of the
male meiotic cell divisions. Dev Biol. 2012;369(2):​ germ granule. Semin Cell Devl Biol. 2014;29:​84–92.
356–361. 40. O’Donnell L, Stanton P, de Kretser DM. Endocrinology
27. Shibuya H, Morimoto A, Watanabe Y. The dissection of the male reproductive system and spermatogene-
of meiotic chromosome movement in mice using sis. In De Groot LJ, Chrousos G, Dungan K et al., eds.
an in vivo electroporation technique. PLoS Genet. Endotext [Internet]. South Dartmouth, MA: MDText.
2014;10(12):e1004821. com, Inc.; 2000. Available from: https://www.ncbi​
28. LaFountain JR, Jr., Siegel AJ, Rickards GK. Chromo­ .nlm.nih.gov/books/NBK279031/.
some movement during meiotic prophase in crane- 41. Chang YF, Lee-Chang JS, Imam JS et al. Interaction
fly spermatocytes: IV. Actin and the effects of between microRNAs and actin-associated protein
cytochalasin D. Cell Motil Cytoskeleton. 1999;43(3):​ Arpc5 regulates translational suppression during
199–212. male germ cell differentiation. Proc Natl Acad Sci
29. Hisano M, Yamada S, Tanaka H, Nishimune Y, U S A. 2012;109(15):5750–5755.
Nozaki M. Genomic structure and promoter activ- 42. Besse F, Ephrussi A. Translational control of local-
ity of the testis haploid germ cell-specific intron- ized mRNAs: Restricting protein synthesis in space
less genes, Tact1 and Tact2. Mol Reprod Dev. 2003;​ and time. Nat Rev Mol Cell Biol. 2008;9(12):​971–980.
65(2):148–156. 43. Walt H, Armbruster BL. Actin and RNA are compo-
30. Oh SD, Park SY, Park JI, Chun SY, Ryu TH, Soh J. nents of the chromatoid bodies in spermatids of the
The novel, actin-like protein Tact3 is expressed in rat. Cell Tissue Res. 1984;236(2):487–490.
rodent testicular haploid germ cells. Mol Reprod Dev. 44. Hara Y, Yamagata K, Oguchi K, Baba T. Nuclear
2013;80(12):988–999. localization of profilin III-ArpM1 complex in mouse
31. Tanaka H, Iguchi N, Egydio de Carvalho C, spermiogenesis. FEBS Lett. 2008;582(20):2998–3004.
Tadokoro Y, Yomogida K, Nishimune Y. Novel actin- 45. Kristo I, Bajusz I, Bajusz C, Borkuti P, Vilmos P.
like proteins T-ACTIN 1 and T-ACTIN 2 are dif- Actin, actin-binding proteins, and actin-related
ferentially expressed in the cytoplasm and nucleus proteins in the nucleus. Histochem Cell Biol. 2016;​
of  mouse haploid germ cells. Biol Reprod. 2003;​ 145(4):373–388.
69(2):​475–482. 46. Misu S, Takebayashi M, Miyamoto K. Nuclear actin
32. Heid H, Figge U, Winter S, Kuhn C, Zimbelmann in development and transcriptional reprogramming.
R, Franke W. Novel actin-related proteins Arp-T1 Front Genet. 2017;8:27.
and Arp-T2 as components of the cytoskeletal calyx 47. Kierszenbaum AL, Rivkin E, Tres LL. Molecular
of the mammalian sperm head. Exp Cell Res. 2002;​ biology of sperm head shaping. Soc Reprod Fertil
279(2):177–187. Suppl. 2007;65:33–43.
33. Tokuhiro K, Miyagawa Y, Tanaka H. Characterizing 48. Kierszenbaum AL, Rivkin E, Tres LL. Acroplaxome,
mouse male germ cell-specific actin capping protein an F-actin-keratin-containing plate, anchors the
alpha3 (CPalpha3): Dynamic patterns of expression acrosome to the nucleus during shaping of the sper-
in testicular and epididymal sperm. Asian J Androl. matid head. Mol Biol Cell. 2003;14(11):4628–4640.
2008;10(5):711–718. 49. Kierszenbaum AL, Rivkin E, Tres LL. The actin-based
34. Behnen M, Murk K, Kursula P et al. Testis-expressed motor myosin Va is a component of the acroplaxome,
profilins 3 and 4 show distinct functional charac- an acrosome-nuclear envelope junctional plate, and
teristics and localize in the acroplaxome-manchette of manchette-associated vesicles. Cytogenet Genome
complex in spermatids. BMC Cell Biol. 2009;10:34. Res. 2003;103(3–4):337–344.
35. Goossens S, Janssens B, Vanpoucke G, De Rycke R, 50. Oko R, Sutovsky P. Biogenesis of sperm perinuclear
van Hengel J, van Roy F. Truncated isoform of mouse theca and its role in sperm functional competence
alphaT-catenin is testis-restricted in expression and and fertilization. J Reprod Immunol. 2009;83(1–2):2–7.
function. FASEB J. 2007;21(3):647–655. 51. Longo FJ, Krohne G, Franke WW. Basic proteins of
36. Greenbaum MP, Yan W, Wu MH et al. TEX14 is essen- the perinuclear theca of mammalian spermatozoa
tial for intercellular bridges and fertility in male mice. and spermatids: A novel class of cytoskeletal ele-
Proc Natl Acad Sci U S A. 2006;103(13):4982–4987. ments. J Cell Biol. 1987;105(3):1105–1120.
References 199

52. Lecuyer C, Dacheux JL, Hermand E, Mazeman E, 67. Pelletier RM. The blood-testis barrier: The junc-
Rousseaux J, Rousseaux-Prevost R. Actin-binding tional permeability, the proteins and the lipids. Prog
properties and colocalization with actin during Histochem Cytochem. 2011;46(2):49–127.
spermiogenesis of mammalian sperm calicin. Biol 68. Li N, Tang EI, Cheng CY. Regulation of blood-­
Reprod. 2000;63(6):1801–1810. testis barrier by actin binding proteins and protein
53. Toshimori K, Ito C. Formation and organization kinases. Reproduction. 2016;151(3):R29–R41.
of the mammalian sperm head. Arch Histol Cytol. 69. Vogl AW, Du M, Wang XY, Young JS. Novel clathrin/
2003;66(5):383–396. actin-based endocytic machinery associated with
54. O’Donnell L, O’Bryan MK. Microtubules and sper- junction turnover in the seminiferous epithelium.
matogenesis. Semin Cell Dev Biol. 2014;30:45–54. Semin Cell Dev Biol. 2014;30:55–64.
55. Russell LD, Russell JA, MacGregor GR, Meistrich 70. Gungor-Ordueri NE, Celik-Ozenci C, Cheng CY.
ML. Linkage of manchette microtubules to the Fascin 1 is an actin filament-bundling protein that
nuclear envelope and observations of the role of the regulates ectoplasmic specialization dynamics in
manchette in nuclear shaping during spermiogenesis the rat testis. Am J Physiol Endocrinol Metabol. 2014;​
in rodents. Am J Anat. 1991;192(2):97–120. 307(9):E738–E753.
56. Vogl AW, Genereux K, Pfeiffer DC. Filamentous 71. Rotkopf S, Hamberg Y, Aigaki T, Snapper SB, Shilo
actin detected in rat spermatozoa. Tissue Cell. 1993;​ BZ, Schejter ED. The WASp-based actin polymeriza-
25(1):33–48. tion machinery is required in somatic support cells
57. Tubb B, Mulholland DJ, Vogl W et al. Testis fascin for spermatid maturation and release. Development.
(FSCN3): A novel paralog of the actin-bundling 2011;138(13):2729–2739.
protein fascin expressed specifically in the elongate 72. O’Donnell L, Nicholls PK, O’Bryan MK, McLachlan
spermatid head. Exp Cell Res. 2002;275(1):92–109. RI, Stanton PG. Spermiation: The process of sperm
58. Brener E, Rubinstein S, Cohen G, Shternall K, release. Spermatogenesis. 2011;1(1):14–35.
Rivlin J, Breitbart H. Remodeling of the actin cyto- 73. Gao Y, Xiao X, Lui WY, Lee WM, Mruk D, Cheng
skeleton during mammalian sperm capacitation CY. Cell polarity proteins and spermatogenesis.
and acrosome reaction. Biol Reprod. 2003;68(3):​ Semin Cell Dev Biol. 2016;59:62–70.
837–845. 74. Li N, Wong CK, Cheng CY. Plastins regulate ecto-
59. Kierszenbaum AL, Rivkin E, Tres LL. Cytoskeletal plasmic specialization via its actin bundling activity
track selection during cargo transport in sperma- on microfilaments in the rat testis. Asian J Androl.
tids is relevant to male fertility. Spermatogenesis. 2016;18(5):716–722.
2011;1(3):221–230. 75. Du M, Young J, De Asis M et al. A novel subcellular
60. Niedenberger BA, Chappell VK, Kaye EP, Renegar machine contributes to basal junction remodeling
RH, Geyer CB. Nuclear localization of the actin reg- in the seminiferous epithelium. Biol Reprod. 2013;​
ulatory protein Palladin in Sertoli cells. Mol Reprod 88(3):60.
Dev. 2013;80(5):403–413. 76. Sriram A, Lyon KR, Ho CD, Huynh N, Vogl AW.
61. Russell LD, Peterson RN. Sertoli cell junctions: Actin disruption results in altered morphology of
Morphological and functional correlates. Int Rev basal tubulobulbar complexes in rat seminiferous
Cytol. 1985;94:177–211. epithelium. Anat Rec. 2016;299(10):1449–1455.
62. Vogl AW, Vaid KS, Guttman JA. The Sertoli cell cyto- 77. Lyon KR, Bosseboeuf E, Vogl AW. An alternative
skeleton. Adv Exp Med Biol. 2008;636:186–211. model of tubulobulbar complex internalization dur-
63. Salanova M, Ricci G, Boitani C, Stefanini M, De Grossi ing junction remodeling in the seminiferous epithe-
S, Palombi F. Junctional contacts between Sertoli lium of the rat testis. Biol Reprod. 2015;​93(1):12.
cells in normal and aspermatogenic rat seminiferous 78. Kumar A, Dumasia K, Deshpande S, Gaonkar R,
epithelium contain alpha6beta1 integrins, and their Balasinor NH. Actin related protein complex subunit
formation is controlled by follicle-­stimulating hor- 1b controls sperm release, barrier integrity and cell
mone. Biol Reprod. 1998;58(2):371–378. division during adult rat spermatogenesis. Biochim
64. Ozaki-Kuroda K, Nakanishi H, Ohta H et al. Biophys Acta. 2016;1863(8):1996–2005.
Nectin couples cell-cell adhesion and the actin scaf- 79. Nicholls PK, Harrison CA, Walton KL, McLachlan
fold at heterotypic testicular junctions. Curr Biol. RI, O’Donnell L, Stanton PG. Hormonal regula-
2002;12(13):1145–1150. tion of sertoli cell micro-RNAs at spermiation.
65. Jiang X, Ma T, Zhang Y et al. Specific deletion of Endocrinology. 2011;152(4):1670–1683.
Cdh2 in Sertoli cells leads to altered meiotic pro- 80. Mital P, Hinton BT, Dufour JM. The blood-testis and
gression and subfertility of mice. Biol Reprod. 2015;​ blood-epididymis barriers are more than just their
92(3):79. tight junctions. Biol Reprod. 2011;84(5):851–858.
66. Gerber J, Heinrich J, Brehm R. Blood-testis barrier 81. Ploen L, Setchell BP. Blood-testis barriers revis-
and Sertoli cell function: Lessons from SCCx43KO ited. A homage to Lennart Nicander. Int J Androl.
mice. Reproduction. 2016;151(2):R15–R27. 1992;15(1):1–4.
200  Cytoskeletons (F-actin) and spermatogenesis

82. Stanton PG. Regulation of the blood-testis barrier. 92. Russell LD, Saxena NK, Turner TT. Cytoskeletal
Semin Cell Dev Biol. 2016;59:166–173. involvement in spermiation and sperm transport.
83. Cameron DF, Muffly KE. Hormonal regulation of Tissue Cell. 1989;21(3):361–379.
spermatid binding. J Cell Sci. 1991;100 (Pt 3):​623–633. 93. Young JS, De Asis M, Guttman J, Vogl AW. Cortactin
84. Sluka P, O’Donnell L, Bartles JR, Stanton PG. depletion results in short tubulobulbar complexes
FSH regulates the formation of adherens junc- and spermiation failure in rat testes. Biol Open.
tions and ectoplasmic specialisations between rat 2012;1(11):1069–1077.
Sertoli cells in vitro and in vivo. J Endocrinol. 2006;​ 94. Qian X, Mruk DD, Cheng YH et al. Actin bind-
189(2):381–395. ing proteins, spermatid transport and spermiation.
85. Muffly KE, Nazian SJ, Cameron DF. Effects of Semin Cell Devl Biol. 2014;30:75–85.
­follicle-stimulating hormone on the junction-related 95. Tang EI, Lee WM, Cheng CY. Coordination of
Sertoli cell cytoskeleton and daily sperm production actin- and microtubule-based cytoskeletons sup-
in testosterone-treated hypophysectomized rats. Biol ports transport of spermatids and residual bodies/
Reprod. 1994;51(1):158–166. phagosomes during spermatogenesis in the rat testis.
86. Qian X, Mruk DD, Wong EW, Lie PP, Cheng CY. Endocrinology. 2016;157(4):1644–1659.
Palladin is a regulator of actin filament bundles at 96. Maekawa M, Kamimura K, Nagano T. Peritubular
the ectoplasmic specialization in adult rat testes. myoid cells in the testis: Their structure and func-
Endocrinology. 2013;154(5):1907–1920. tion. Arch Histol Cytol. 1996;59(1):1–13.
87. Niedenberger BA, Chappell VA, Otey CA, Geyer CB. 97. Vogl AW, Soucy LJ, Lew GJ. Distribution of actin in
Actin dynamics regulate subcellular localization of isolated seminiferous epithelia and denuded tubule
the F-actin-binding protein PALLD in mouse Sertoli walls of the rat. Anat Rec. 1985;213(1):63–71.
cells. Reproduction. 2014;148(4):333–341. 98. Losinno AD, Morales A, Fernandez D, Lopez LA.
88. Li N, Mruk DD, Cheng CY. Actin binding proteins in Peritubular myoid cells from rat seminiferous
blood-testis barrier function. Curr Opin Endocrinol tubules contain actin and myosin filaments distrib-
Diabetes Obes. 2015;22(3):238–247. uted in two independent layers. Biol Reprod. 2012;​
89. Russell LD. Observations on the inter-relationships 86(5):150–151.
of Sertoli cells at the level of the blood-testis barrier: 99. Holstein AF, Maekawa M, Nagano T, Davidoff MS.
Evidence for formation and resorption of Sertoli- Myofibroblasts in the lamina propria of human
Sertoli tubulobulbar complexes during the spermato- semi-niferous tubules are dynamic structures of
genic cycle of the rat. Am J Anat. 1979;155(2):259–279. heterogeneous phenotype. Arch Histol Cytol. 1996;​
90. Li N, Mruk DD, Wong CK, Han D, Lee WM, Cheng 59(2):109–125.
CY. Formin 1 regulates ectoplasmic specialization in 100. Losinno AD, Sorrivas V, Ezquer M, Ezquer F, Lopez
the rat testis through its actin nucleation and bun- LA, Morales A. Changes of myoid and endothe-
dling activity. Endocrinology. 2015;156(8):2969–2983. lial cells in the peritubular wall during contraction
91. Young JS, Guttman JA, Vaid KS, Vogl AW. of the seminiferous tubule. Cell Tissue Res. 2016;​
Tubulobulbar complexes are intercellular podosome- 365(2):425–435.
like structures that internalize intact intercellular
junctions during epithelial remodeling events in the
rat testis. Biol Reprod. 2009;80(1):162–174.
Roles of mTOR signaling
in spermatogenesis
16
LAN YE and KE ZHENG

INTRODUCTION transplantation. Substantial evidence suggests that the


mTOR complexes and its inhibitor rapamycin dysregulation of the mTOR pathway is associated with
cancer pathologies, and hyperactivation of mTORC1 is
The mechanistic target of rapamycin (mTOR) (TOR1/
highly prevalent in human cancers. In support of this view,
TOR2 in yeast) was originally identified in budding yeast
the mutations of genes encoding for many tumor suppres-
through a genetic screen in 1991,1,2 and it is the target of
sors promote mTORC1 activation.25,26 For example, P53,
a small molecule called rapamycin. mTOR is a large con-
a well-known tumor suppressor and a major checkpoint
served serine/threonine protein kinase that belongs to the
protein, regulates mTOR activity through AMP-activated
phosphoinositide 3-kinase (PI3K)-related kinase family,
kinase (AMPK) and the tuberous sclerosis (TSC) 1/TSC2
and it forms two complexes in mammals called mTORC1
complex.27 PTEN, a putative protein tyrosine phospha-
and mTORC2.3–6 mTORC1 consists of mTOR, Raptor,
tase gene mutated in many types of human cancers, lies
mLST8/GβL, PRAS40, and DEPTOR, and mTORC2 con-
upstream of the mTOR complexes and negatively regulates
sists of mTOR, Rictor, mLST8/GβL, mSIN1, Protor1/2, and
the PI3K pathway.25,28 Additionally, mTORC2 is required
DEPTOR. Raptor and PRAS40 are specific components
for the development of tumors from human prostate epi-
of mTORC1 complex that regulates ribosomal biogenesis,
thelial cells caused by Pten deletion.29 LKB1 is a critical
translation of mRNAs containing pyrimidine-rich 5’TOP
upstream kinase for AMPK activation and is also involved
or TOP-like motifs, and autophagy through downstream
in cancer development. Mutations in the Lkb1 gene are
targets including ribosomal protein S6 kinase (S6K),
associated with a cancer-prone disease, Peutz-Jeghers
eukaryotic initiation factor 4E binding protein-1 (4E-BP1),
syndrome (PJS).30,31 Previous studies demonstrated LKB1
and ULK1/Atg13/FIP200 complex (unc-51-like kinase-1/
phosphorylates and activates TSC2, which further regu-
mammalian autophagy-related gene 13/focal adhesion
lates the small GTPase Rheb and subsequently inhibits
kinase-interacting protein of 200 kDα).7–11 In skeletal mus-
mTORC1.26,32 This evidence suggests that modulating of
cle tissues and cells, Raptor is required for maintaining
the mTOR pathway by rapamycin and its analogs (rapa-
mitochondrial respiration.3,12,13 In contrast, Rictor, mSIN1,
logs) may have great potential to treat cancer pathogenesis.
and Protor1/2 are specific components of mTORC2 com-
So far, there are two mTOR inhibitors approved by the U.S.
plex, which is involved in metabolism through AKT and
Food and Drug Administration (FDA) for treating specific
serum/glucocorticoid induced kinase (SGK).14,15 It also sus-
cancer types. Temsirolimus, the first approved rapalog, is
tains cytoskeleton through protein kinase C (PKC-α).16
an intravenous drug for advanced stage of renal cell car-
Rapamycin (clinically referred to as sirolimus) was dis-
cinoma. The second approved rapalog is everolimus for
covered in the soil of Easter Island as a macrolide compound
treatment of renal cell cancer and also for a genetic disease
produced by Streptomyces bygroscopicus.17 Mechanistically,
caused by Tsc1/Tsc2 mutation.26
rapamycin forms an intracellular complex with FKBP12 to
In addition to the critical role of the deregulated mTOR
inhibit mTOR kinase through binding its FRB domain.18,19
pathway implicated in cancer progression, the dysregu-
Rapamycin is known as a potent immunosuppressant
lation of this pathway is also involved in many patho-
for the prevention of transplant rejection20,21 and it also
logical conditions, including obesity, type 2 diabetes,
inhibits the growth and proliferation of mammalian cells
neurodegenerative disorders, as well as aging processes.
with the therapeutic potential in treating human can-
For example, the dysfunction of autophagy is impli-
cers.22 mTORC1 complex is sensitive to rapamycin and
cated in neurodegenerative diseases, such as Parkinson’s
nutrient signals, whereas mTORC2 was originally consid-
disease, Alzheimer’s disease, and Huntington’s dis-
ered a rapamycin-resistant complex. However, prolonged
ease.26 mTORC1 regulates autophagy and inhibition of
rapamycin treatment also disrupts mTORC2 assembly and
mTORC1 by rapamycin reduces the accumulation of
inhibits phosphorylation of mTORC2 downstream effec-
protein aggregates and subsequently has a beneficial role
tors in cultured cell lines and tissues.23,24
in alleviating neurodegenerative-associated symptoms.33
Importantly, reduced TOR signaling either by genetic
PHARMACOLOGICAL APPLICATIONS OF RAPAMYCIN manipulation or small inhibitors extends the lifespan
AND RAPALOGS IN HUMAN DISEASE of model organisms including yeast, worms, and flies. 34
In 1999, rapamycin received FDA approval as an immu- In mammals, rapamycin is the small molecule that
nosuppressant agent to prevent rejection in organ robustly extends lifespans in mice across three sites by

201
202  Roles of mTOR signaling in spermatogenesis

the National Institute on Aging Interventions Testing ROLES OF mTOR SIGNALING IN MAINTAINING
Program (ITP).35,36 Supportingly, genetic attenuation SPERMATOGONIAL FUNCTION
of mTORC1 signaling or deletion of the mTORC1 sub- The lifelong spermatogenesis in mammals is founded by
strate S6 kinase 1 (S6K1) is sufficient to extend lifespan the self-renewal potential of the spermatogonial popu-
in mice.23,37 In contrast, depletion of Rictor, an essential lation, which resides on the basement of seminiferous
protein component mTORC2, decreases the lifespan in tubules. In mouse testes, spermatogonial population con-
male mice.38 These studies indicates that rapalogs of spe- tains A, intermediate, and B spermatogonia, and type A
cifically targeting mTORC1 may promote longevity and spermatogonia are further divided into undifferentiated
have beneficial effects in treating aging-associated dis- (As, Apr, and Aal) and differentiated spermatogonia (A1-4)
eases in humans. with a distinct gene expression pattern that predicts dif-
ferential self-renewal potential.78 One of the questions in
CONNECTING mTOR SIGNALING TO MALE FERTILITY stem cell biology is, What is the fundamental mechanism
As mentioned above, LKB1 is an essential tumor sup- that coordinates various stimuli to regulate the balance
pressor and mutation of Lkb1 is associated with human between spermatogonial stem cell maintenance and dif-
cancers. LKB1 is a serine/threonine kinase that directly ferentiation? Recently, genetic studies in mouse models
phosphorylates AMPK, which further phosphory- demonstrated that the signaling network anchored by the
lates and activates the negative upstream regulator of mTOR kinase is one of the mechanisms.
mTORC1, TSC2. 39 Mutation of Lkb1 was identified in The glial cell line-derived neurotrophic factor (GDNF)
patients with PJS and is believed to have a causal role in is a crucial growth factor required for self-renewal of sper-
this disease. Importantly, clinical evidence shows that matogonial stem cells (SSCs).79 Mechanistically, GDNF
PJS patients have defects in spermatogenesis, includ- binds the cell surface receptor GFRA1/RET, which are
ing substantial loss of germ cells, and are at high risk expressed in a subset of undifferentiated spermatogo-
of developing Sertoli cell tumors.40,41 Moreover, male nia (spermatogonial progenitor cells, SPCs).80 Feng and
patients who are prescribed with sirolimus in organ colleagues first indicated that P70 S6 kinase, one of the
transplantation and cancer therapies have high preva- downstream targets of mTORC1, mediates the role of stem
lence of infertility.42 This evidence indicates that the cell factor (SCF) on the proliferation of cultured spermato-
mTOR pathway regulates multiple physiological pro- gonial cells.81 PLZF is an essential transcriptional factor
cesses, and dysregulation of the mTOR pathway is asso- for SPCs maintenance, evidenced by compromised self-
ciated with pathological conditions such as cancer and renewal potential in Plzf-/- testes. Hobbs and colleagues
infertility in humans. demonstrated that mTORC1 signaling is enhanced in
A close connection between the mTOR pathway and cultured SPCs isolated from prepubertal Plzf knockout
male fertility has also demonstrated in lower model testes, and the aberrant mTORC1 activation inhibits the
organisms. In Caenorhabditis elegans (C. elegans), Vellai response of SPCs to GDNF through suppressing expres-
and colleagues found that depletion of mTOR homo- sion of GDNF receptor components GFRα and c-Ret.82 The
log let-363/CeTor by RNA interference reduces fertility.43 RNA-binding protein Nanos2, another essential factor
The reproductive defects are also observed in mutants of for spermatogonial self-renewal, marks the undifferenti-
daf-2 gene, which encodes the C. elegans insulin/IGF-like ated spermatogonial populations in testes. Similar to the
receptor.44–47 Insights into the regulation of the mTOR observations in Plzf-/- testes, hyperactivation of mTORC1
pathway on fertility have also been provided by studies in was also apparent in germline stem cells from Nanos2 con-
Drosophila. In Drosophila, the release of insulin-like pep- ditional knockout testes. Indeed, Nanos2 interacts with
tides (ILPs) responds to changes in nutritional availability other mRNP components to promote SPCs self-renewal
and couples metabolism, longevity, and fertility through through repressing mTORC1 by sequestrating mTOR in
a TOR/Raptor-dependent mechanism.48 Loss of function mRNPs.83 Together, these findings indicate that mTORC1
of PRAS40, an inhibitory subunit of TORC1, completely signaling mediates the function of several essential
rescues the sterility of insulin receptor substrate (chico) growth factors or transcriptional factors in maintaining
mutant flies.49,50 In contrast, Drosophila mutants of Rictor SPCs homeostasis, but they did not show genetic evidence
or Sin1, two members of TORC2, grows normally and to support a direct role of mTOR signaling in spermato-
are fertile.51 Therefore, the mTORC1 pathway integrates gonial self-renewal and differentiation. Hobbs and col-
nutrient status in animals to regulate fertility in a man- leagues further define the mTORC1 signaling network in
ner that is highly conserved from flies to mammals (Table controlling SPCs function by conditionally deleting TSC2,
16.1). More recently, an accumulating body of evidence a key upstream regulator of mTORC1.63 TSC2 negatively
suggests an essential role of the mTOR pathway in mouse regulates mTORC1 by inactivating Rheb, which stimu-
spermatogenesis. Genetic manipulation for inhibition/­ lates mTORC1 activity.3 Intriguingly, different subpopula-
activation of mTOR signaling in mice is complicated, tions of undifferentiated spermatogonia exhibits distinct
and both mTORC1 and mTORC2 play important roles in staining intensity of mTORC1 activity characterized by its
multiple aspects of testicular function as shown in greater downstream targets phospho-S6 and 4EBP1.63,65 mTORC1
detail in Table 16.1. activity is lower in SSCs with high self-renewal potential
Roles of mTOR signaling in maintaining spermatogonial function  203

Table 16.1  Genetic evidence of TOR pathway in reproduction.


Species Genetic strategies Phenotypes References
Drosophia dTorA948V mutant Abnormal testes with enlarged apical tip 52

dTORp (P insertions in the dTOR) Infertile 53

dTorW1251R, dTorP2293L mutants GSCs maintenance defects 54

Tsc1Q87X mutant GSCs loss 54

chico (insulin receptor substrate) mutants Infertile 50,55

PRAS40 null Fertile, but rescues the infertility of chico 49

mutants
InR (insulin-like receptor) mutants Sterile 52

Tsc1 mutant Precocious differentiation of GSCs and 56

loss
Tsc2 mutant GSCs loss 56

Rictor mutant Fertile 51

Sin1 mutant Fertile 51

C. elegans CeTor RNAi Reduced fertility 43

Daf-15 (Raptor) mutant Sterile 57

CeTor mutant Gonadal developmental arrest 58

rict-1 (Rictor) mutant Reproductive defects 59

rsks-1(S6K) mutant Reduced fertility 60

rsks-1 mutant Germline progenitors loss 61

CeTor RNAi Reduced germline progenitors 61

Daf-15 (Raptor) RNAi Reduced germline progenitors 61

ife-1 (eIF4E) deletion Reduced germ cells 61

Daf-18 (Pten) mutant Reproductive defects 62

Daf-2 (insulin receptor family) Reproductive defects 44

M. musculus Tsc2fl/-Stra8-Cre Normal testicular phenotype 63

Tsc2fl/-Vasa-Cre SPCs maintenance defects 63

Tsc2fl/-Amh-Cre Defective spermatogenesis in juvenile 63

but recovered in adulthood


Rheb fl/-Vasa-Cre Oligoasthenoteratozoospermia 64

Tsc1fl/-Vasa-Cre Precocious spermatogonial 65

differentiation
mTOR fl/-Vasa-Cre Arrested at differentiated spermatogonia 66

Constitutive activation of AKT Promotes proliferation of SSCs 67

Foxo1fl/-Vasa-Cre SSC defects, sterile 68

Foxo1/3/4 triple knockout Severe spermatogenic defects 68

Pten fl/-Vasa-Cre Depletion of germ cells 68

Pdk1 fl/-Vasa-Cre Normal spermatogonia 68

Raptor fl/-Vasa-Cre Spermatogonial defects 69

Raptor fl/-Ngn3-Cre Meiotic arrest 70

Rictor fl/-Ngn3-Cre Spermatogonial and BTB integrity 71

defects
Rictor RNAi BTB integrity defects 72

LKB1S mutant Abnormal spermiogenesis and 73,74

spermiation
Lkb1 fl/-Amh-Cre Sertoli-cell-only tubule 75

Tsc1fl/-Amh-Cre Sertoli-cell-only tubule 75

Tsc2fl/-Amh-Cre Sertoli-cell-only tubule 75

Ptenfl/-Amh-Cre Normal testicular phenotype 75

Rictorfl/-Amh-Cre BTB integrity defects 76

Raptor fl/-Ksp-Cre Disrupted spermatozoa 77


204  Roles of mTOR signaling in spermatogenesis

but induced in differentiation-committed progenitor cells. regulation takes place in a distinct subset of SPCs with
In their study, three mouse models were made with con- a differential dependency on mTORC1 activity. Shortly
ditional deletion of Tsc2 in germ cells and somatic cells after this work, Busada and colleagues showed the role of
within testes by using Vasa-Cre, Stra8-Cre, and Amh- mTORC1 in SSCs by treating neonatal mice with rapamy-
Cre strategies, respectively. Through genetic and cell bio- cin, a well-known mTOR inhibitor.84 They showed that
logical analyses, they demonstrate that the self-renewal activation of mTORC1 is required for the spermatogonial
potential of undifferentiated spermatogonia is impaired differentiation. Specially, inhibition of mTOR signaling
in Vasa-Cre-mediated Tsc2-deleted testes (Figure 16.1), by rapamycin promotes the self-renewal potential of sper-
while the spermatogonial function appears unaffected matogonia, since rapamycin treatment causes an accu-
in Tsc2-deficient testes using Stra8-Cre and Amh-Cre.63 mulation of undifferentiated spermatogonia, and blocks
TSC1, another upstream regulator of mTORC1, inter- spermatogonial differentiation partially through transla-
acts with TSC2 to form a complex to negatively regulate tional control of a subset mRNAs including Kit, Sohlh1,
mTORC1 activity. Wang and colleagues observed preco- and Sohlh2 (Figure 16.1).84 Recently, they provided direct
cious spermatogonial differentiation at the expense of genetic evidence of mTOR signaling in spermatogenesis by
germline maintenance and subsequently testes atrophy specifically inactivating mTOR using Vasa-Cre. In agree-
were observed in Tsc1 knockout testes (Figure 16.1).65 ment with their previous studies, mTOR deficiency sub-
These findings suggest mTORC1 acts cell-autonomously stantially blocks spermatogonial differentiation; hence,
in the maintenance of the germline stem cells, and this spermatocytes are absent at different ages (Figure 16.1).66

PI3K
mTORC2
Deptor
mSin1 PIP3 PTEN Depletes germ cells
mTOR mLST8
BTB integrity defects
Impaired spermatogonial Rictor PDK1 No defects in SSCs maintenance
differentiation Protor

Constitutive
activation
Promotes SSCs proliferation Akt Foxo1 SSCs maintenance defects

Precocious spermatogonial TSC1 TSC2 Impaired undifferentiated spermatogonia


differentiation

GTP-Rheb GDP-Rheb

mTORC1
mTOR Blocked spermatogonial differentiation
Deptor mLST8
Raptor
Meiotic arrest, MSCI defects
PRAS40
Rapamycin Inhibits spermatogonial differentiation
Activate Inhibit

Figure 16.1  mTOR signaling and deletion of its components influences spermatogenesis. mTOR forms two distinct protein
complexes, mTORC1 and mTORC2. mTORC2 phosphorylates Akt kinase at its serine 473 site. Conditional deletion of Rictor in Sertoli
cells disrupts cytoskeletal organization and cell polarity. Conditional deletion of Rictor in germ cells impairs spermatogonial dif-
ferentiation and disrupts intercellular adhesion and BTB integrity. PTEN, a negative regulator of the PI3K/AKT signaling, inactivation
of Pten results in AKT hyperphosphorylation and severe defects in SSCs self-renewal, while inactivation of Pdk1 has no obvious
effects on SSCs maintenance. AKT phosphorylates and inactivates FOXO1 while conditional deletion of Foxo1 causes defects in SSCs
maintenance. TSC1 interacts with TSC2 and forms a TSC complex, which inactivates Rheb by converting GTP-Rheb into its inactive
GDP-Rheb. Deletion of either Tsc1 or Tsc2 activates mTORC1 signaling and results in precocious spermatogonial differentiation and
impaired SSCs maintenance. In contrast, specifically knocking out Rheb caused defects in meiotic progression and spermatogenesis.
Inhibition of mTORC1 by acute rapamycin treatment blocks spermatogonial differentiation, and conditional knockout of mTOR using
Vasa-cre also results in defects in spermatogonial differentiation. Specifically inactivating Raptor using Ngn3-Cre leads to derepres-
sion of sex chromosomes-linked genes and meiotic arrest.
Roles of mTOR signaling in maintaining spermatogonial function  205

Interestingly, a subset of undifferentiated spermatogonia, mTOR signaling in meiosis


PLZF and GFRα1 positive, remained in mTOR-deficient As discussed above, mTOR signaling is an essential
testes, indicating spermatogonial self-renewal potential pathway in the organism to coordinate the balance
is not compromised. Consistently, Xiong et al. revealed of spermatogonial self-renewal and differentiation.
that enhanced mTORC1 activity, induced by P53 loss, Spermatogenesis begins with spermatogonial mitotic
also promotes spermatogonial differentiation.85 Taken proliferation, followed by two meiotic divisions, and
together, mTORC1 is involved in the GDNF response of mature into haploid spermatozoa through spermiogen-
SSCs through crosstalk with transcriptional factors, and it esis. Meiosis is a specialized division process in germ
regulates the balance of spermatogonial self-renewal and cells that contains several key stages including chro-
differentiation. mosomal pairing, chromosomal recombination, and
Compared to mTORC1, mTORC2 complex is less accurate segregation. Recently, we showed that Raptor,
sensitive to rapamycin. However, prolonged rapamy- an essential component of mTORC1, expresses in iso-
cin inhibits mTORC2 signaling in vitro and in vivo.23,24 lated spermatogonia, pachytene spermatocytes, and
mTORC2 directly activates AKT at its hydrophobic motif round spermatids, especially with high abundance in
(Ser473) and with phosphorylation at other sites further pachytene spermatocytes, suggesting the potential of
phosphorylates substrates including forkhead box O1/3a mTOR signaling functions beyond the spermatogonial
(Foxo1/3a), TSC1/2, and glycogen synthase kinase alpha/ development. To characterize the role of mTORC1 dur-
beta (GSK3α/β). Through a germline stem cell culture ing meiosis, we generated Raptor conditional knock-
system, Lee and colleagues have reported that activa- out mice using Neurogenin Ngn3-Cre, which has been
tion of AKT kinase is observed in GDNF-treated SSCs.67 shown to fully disrupt protein expression in testes at
Consistent with our recent study, phosphorylated AKT postnatal day 9. Indeed, Raptor-deficient testes arrest
is one of the dramatically increased phosphorylated at pachytene stage with a particularly high frequency
proteins upon GDNF stimuli identified by quantitative of X-Y asynapsis and defects in recruiting silencing fac-
mass-spectrometry-based proteomic and phosphopro- tors and epigenetic modifiers into the entire sex chro-
teomic analyses.69 Constitutive activation of AKT by matin, which is illustrated in Figure 16.2.70 However,
transfecting with myr-Akt-Mer plasmid substantially conditional ablation of mTORC2 component Rictor by
promotes proliferation of SSCs even in the absence of employing the same knockout strategy shows Rictor
GDNF (Figure 16.1).67 Goertz and colleagues generated is dispensable for meiotic progression and recombina-
germline conditional Foxo1 knockout and Foxo1/3/4 tri- tion, suggesting the nonoverlapping functionality of
ple knockout mice to investigate the function of Foxos in mTORC1 and mTORC2 during meiosis.71
the mouse testes, particularly in SSCs self-renewal and
differentiation.68 Conditional Foxo1-deficient mice are
mTOR signaling in Sertoli cells
sterile, with distinct defects in SSCs maintenance and
initiation of spermatogenesis (Figure 16.1). Although Spermatogenesis relies largely on an exclusive type of
Foxo3 and Foxo4 are dispensable for germ cell devel- nongerm cells called Sertoli cells that provide nutritional
opment, Foxo1/3/4 triple knockout mice exhibit more and physical support for germ cells at different stages of
severe spermatogenic defects, indicating the members of their development within seminiferous tubules.86 These
Foxo family compensate their functions. These findings somatic cells, extending from the basement membrane
indicate the transcriptional factor FOXO1, one down- almost throughout the seminiferous epithelium, keep
stream target of AKT and AKT kinase itself are required in frequent contact with differentiating germ cells and
for maintaining SSCs. Goertz and colleagues further direct their migration toward the epithelia lumen, for
conditionally inactivated Pten or 3-phosphoinositide- which Sertoli-Sertoli and Sertoli-germ cell interactions
dependent protein kinase 1 (Pdk1) to determine whether are critical.87,88 The blood-testis barrier (BTB) is formed
Foxos regulate SSCs function through PI3K-AKT signal- between adjacent Sertoli cells with the structures of tight
ing. Indeed, Pten mutant gradually depletes germ cells in junction (TJ), basal ectoplasmic specialization (basal
an age-dependent manner, which indicates Pten shares ES), gap junction (GJ), and desmosome (DS).89–91 As an
similar roles in maintaining SSCs function with FOXOs immunological barrier, the BTB protects germ cells at
(Figure 16.1).68 Pdk1 loss attenuates AKT phosphoryla- the basal compartment against immune responses from
tion and subsequently constitutive Foxo1 activation, and those at the apical compartment. An extensive network
no obvious defects in SSCs self-renewal and differentia- of actin filament bundles that lie perpendicular to the
tion were observed in Pdk1 mutant testes (Figure 16.1).68 Sertoli cell plasma membrane at above adhesion junc-
These results strongly suggest Foxo1 acts through PI3K- tions except for DS is recognized as the major force by
AKT to regulate SSCs self-renewal and PI3K-AKT sig- which BTB is formed as one of the tightest BTBs.72,86,92 In
naling is crucial for SSCs maintenance. Moreover, Rictor order to facilitate the translocation of preleptotene sper-
has a germline-specific role in maintaining normal sper- matocytes to initiate meiosis, BTB undergoes remodel-
matogonial differentiation, suggesting mTORC1 and ing via cyclic junctional events during stage VIII–XI of
mTORC2 functions may overlap during spermatogonial the epithelial cycle.92 Spermatocytes continuously travel
differentiation.71 toward the luminal edge of the seminiferous tubule,
206  Roles of mTOR signaling in spermatogenesis

(a) WT (b) Raptorcko signaling participates in actin cytoskeleton reorganiza-


tion and BTB remodeling.92 mTOR is also involved in the
extensive reorganization of F-actin network to modulate
the BTB dynamics. Disrupting mTOR in Sertoli cells
affects meiotic progression probably due to improper gap
junction alpha-1 protein distribution.99 A component of
the basement membrane laminin α2 is essential for the
SYCP3 SYCP3
rH2AX rH2AX Sertoli cell TJ barrier through mTORC1 signaling.100
Phospho-S6 is a downstream signaling molecule  of
(c) WT (d) Raptorcko mTORC1 that colocalizes with zonula occludens-1 (ZO-1),
a BTB adaptor protein that connects TJ to actin cytoskel-
eton and TJ-barrier function.101 A master upstream pro-
tein kinase LKB1 has a critical role in spermiation due
in part to defects in the breakdown of the junctions of
ESs.73,74 Specific loss of Lkb1 in Sertoli cells causes dra-
SYCP3 SYCP3 matic defects in Sertoli cell polarity and testicular junc-
H3K9me3 H3K9me3 tional complexes by ectopic activation of mTOR, leading
to Sertoli-only phenotype in spermatogenesis.75 As LKB1
(e) WT pachynema (f ) Raptorcko pachynema
positively regulates mTOR signaling that is mediated via
Silencing of Impaired silencing of
sex chromosomes sex chromosomes TSC1/2,102 conditional deletion of Tsc1 and Tsc2, but not
Pten, phenocopys the Lkb1 mutant.75 Clinically, patients
X
X of Peutz-Jeghers syndrome, relevant to gene mutations
in the LKB1-mTOR signaling, often develop Sertoli cell
Y
Y tumors impairing spermatogenesis. 30,31,40,41 These studies
together reinforce the importance of the mTOR signal-
ing for ensuring normal Sertoli cell function and male
fertility.
Figure 16.2  Mechanisms responsible for meiotic arrest Raptor and Rictor localize in the seminiferous epithe-
in conditional Raptor knockout testes. (a) In wild-type lium near the basement membrane. Rictor is a crucial BTB
pachynema, γH2AX disappeared from synapsed auto- regulator, protecting the Sertoli cell TJ-barrier function
somes and accumulated on sex chromatin. (b) In Raptorcko in vitro and the BTB integrity in vivo.72 RNAi-mediated
pachynema, γH2AX was diminished on sex chromatin and par- knockdown of Rictor in cultured Sertoli cells led to a
tially remained on autosomes. (c) In wild-type pachynema, the reduced PKC-α phosphorylation and actin reorganization,
repressive epigenetic maker H3K9m3 is enriched on sex chro- with reduced F-actin to support the BTB TJ structure.72 In
matin and centromeric regions of autosomes. (d) In Raptorcko mouse testis, Sertoli cell-specific ablation of Rictor led to
pachynema, sex chromosomes were largely exposed except disregulation of cytoskeleton and disruption of BTB integ-
for the centromeric region of X chromosome. (e) In wild-type rity resulting in azoospermia.76 Our recent study demon-
pachynema, autosomes are synapsed except for XY chromo- strates an equivalent essentiality of the germline-specific
somes, which only pair at a short region and are silenced. Rictor in protection of BTB structure and function71
(f) In Raptorcko pachynema, extensive asynapsis including implicating an interconnection of  mTORC2-mediated
autosomes and sex chromosomes takes place, and sex chro- BTB regulatory pathway between somatic and germinal
mosomes are not efficiently silenced. cells. A “yin” and “yang” model has been proposed to
explain the stage-specific expression of the components of
mTOR complexes Raptor and Rictor during the epithelial
synchronizing with the dynamic Sertoli-germ cell inter- cycle and how mTORC1 and mTORC2 exert antagonistic
action. Compelling evidence has implicated the cen- regulation on BTB.92 BTB dynamics is regulated by both
tral involvement of the mTOR pathway in Sertoli cell mTORC1 and mTORC2 signaling. Coinciding with the
physiology and function during spermatogenesis.93,94 timing of BTB restructuring at stage VIII–IX of the epi-
Follicle-stimulating hormone (FSH) is the major Sertoli thelial cycle, ribosomal protein S6 (S6) is phosphorylated
cell mitogen and regulates Sertoli cell proliferation via and specifically activated at the BTB site to facilitate the
a PI3K/Akt/mTORC1 pathway.95 17b-estradiol pro- opening of BTB, possibly coupled with transient downreg-
motes Sertoli cell proliferation via mTOR activation.96 ulation of the TJ protein claudin-11.101 The mTORC2 sig-
Human Sertoli cells, when stimulated with rapamycin, naling might be involved in maintaining the BTB integrity
present accelerative glucose consumption, increased during all the stages of the epithelial cycle of spermato-
sensitivity to oxidative stress, and altered mitochon- genesis except at stage VIII–IX when Rictor is downreg-
drial bioenergetics.97,98 In prepubertal mice, short-term ulated and the BTB is under restructuring.72 Therefore,
in vivo rapamycin treatment does not affect the state of the “assembly” and “disassembly” of the BTB is precisely
Sertoli cells.84 Recent studies have reported that mTOR coordinated by mTORC2 and mTORC1, respectively.
References 207

CONCLUSION Mengrou Liu for helpful discussions and providing refer-


The signaling anchored by mTOR kinase is one of the ences. The Zheng lab is supported by National Key R&D
fundamental pathways responding to the nutritional Program of China (2016YFA0500902), National Natural
state of the organisms to regulate cell growth, metabo- Science Foundation of China (31471228, 31771653), Jiangsu
lism, and reproduction. In this review, we have described Science Foundation for Distinguished Young Scholars
­reproduction-associated observations in low model organ- (BK20150047), and Natural Science Foundation of Jiangsu
isms with mutation/deletion in mTOR pathway genes and Province (BK20140897, 14KJA180005, 14KJB310004).
further detailed the current advances on the function of The Ye lab is supported by National Natural Science
the mTOR pathway during mouse spermatogenesis, espe- Foundation of China Grant (81471502), Innovative and
cially in the process of spermtogonial self-renewal and Entrepreneurial Program of Jiangsu Province, and Natural
differentiation, meiosis, and Sertoli cell physiology. These Science Foundation of Jiangsu Province (15KJA180006).
findings suggest mTOR signaling is a highly evolutionally
conserved pathway for germ cell development. REFERENCES
Notably, inhibition of mTOR by rapamycin treatment 1. Cafferkey R, Young PR, McLaughlin MM et al.
significantly promotes spermatogonial self-renewal poten- Dominant missense mutations in a novel yeast pro-
tial and restored the functional capacity in SPCs from Plzf tein related to mammalian phosphatidylinositol
knockout mice. On the other hand, hyperactivation of 3-kinase and VPS34 abrogate rapamycin cytotoxic-
mTORC1 signaling due to deletion of Tsc1, Tsc2, or Pten ity. Mol Cell Biol. 1993;13(10):6012–6023.
impairs the functional capacity of SPCs. This phenomenon 2. Kunz J, Henriquez R, Schneider U et al. Target of
is also observed in somatic cells. For example, reduced rapamycin in yeast, TOR2, is an essential phosphati-
mTORC1 signaling in Paneth cells mediates the beneficial dylinositol kinase homolog required for G1 progres-
effects of calorie restriction on intestinal stem-cell (ISC) sion. Cell. 1993;73(3):585–596.
function, and rapamycin increases the proliferation of 3. Laplante M, Sabatini DM. mTOR signaling in growth
ISC.103 These results suggest inhibition of mTOR signal- control and disease. Cell. 2012;149(2):274–293.
ing has a protective effect on stem cells, and hyperactive 4. Wullschleger S, Loewith R, Hall MN. TOR signaling
mTORC1 promotes excessive proliferation or precocious in growth and metabolism. Cell. 2006;124(3):471–484.
differentiation, which subsequently depletes stem cell pop- 5. Jacinto E, Loewith R, Schmidt A et al. Mammalian
ulation. Helsel and colleagues recently reported that SSCs TOR complex 2 controls the actin cytoskeleton and
are prone to utilize lactate through glycolysis as their pri- is rapamycin insensitive. Nat Cell Biol. 2004;6(11):
mary bioenergetics process, and glycolysis-optimized cul- 1122–1128.
ture media boost the regenerative capacity of the SSCs.104 6. Sarbassov DD, Ali SM, Kim DH et al. Rictor, a novel
Interestingly, it was reported that mTOR inhibition by ­binding partner of  mTOR, defines a rapamycin-­
rapamycin reduced mitochondrial oxidative phosphory- insensitive and raptor-independent pathway that
lation and enhanced glycolysis and hence produced more regulates the cytoskeleton. Curr Biol. 2004;​14(14):​
lactate.105 This suggests the metabolic shift from oxidative 1296–1302.
phosphorylation to glycolysis by inhibition of mTOR may 7. Blommaart EF, Luiken JJ, Blommaart PJ et al.
mediate its effects on maintaining SSCs. However, the Phosphorylation of ribosomal protein S6 is inhibi-
downstream effectors of mTORC1 or the molecular link tory for autophagy in isolated rat hepatocytes. J Biol
that mediates this effect remains to be identified. It is impor- Chem. 1995;270(5):2320–2326.
tant to note that rapamycin robustly extends life spans in 8. Hara K, Yonezawa K, Weng QP et al. Amino acid
genetically heterogeneous mice,36 and reduced mTORC1 sufficiency and mTOR regulate p70 S6 kinase and
signaling through genetic manipulation in mouse model eIF-4E BP1 through a common effector mechanism.
is sufficient to increase longevity without any side effects J Biol Chem. 1998;273(23):14484–14494.
on glucose metabolism.23 Future studies will be needed to 9. Ganley IG, Lam du H, Wang J et al. ULK1.
explore the mechanism of rapamycin used in the longevity ATG13.FIP200 complex mediates mTOR signal-
study on spermatogenesis, and ultimately provide implica- ing and is essential for autophagy. J Biol Chem.
tions for designing new molecules for targeting mTOR to 2009;284(18):12297–12305.
confer their beneficial effects on human health. 10. Hosokawa N, Hara T, Kaizuka T et al. Nutrient-
dependent mTORC1 association with the ULK1-
DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST Atg13-FIP200 complex required for autophagy. Mol
No potential conflicts of interest were disclosed. Biol Cell. 2009;20(7):1981–1991.
11. Jung CH, Jun CB, Ro SH et al. ULK-Atg13-FIP200
ACKNOWLEDGMENTS complexes mediate mTOR signaling to the autoph-
We would like to thank C. Yan Cheng at the Center for agy machinery. Mol Biol Cell. 2009;20(7):1992–2003.
Biomedical Research (Population Council, New York) for 12. Bentzinger CF, Romanino K, Cloëtta D et al.
critical reading of this manuscript. We thank all members Skeletal muscle-specific ablation of raptor, but not
of the Zheng and Ye labs, particularly Mengneng Xiong, of rictor, causes metabolic changes and results in
Shun Bai, Ruping Zhu, Suwen Tian, Shuya Zhang, Xin Li, and muscle dystrophy. Cell Metab. 2008;8(5):411–424.
208  Roles of mTOR signaling in spermatogenesis

13. Morita M, Gravel SP, Chénard V et al. mTORC1 29. Guertin DA, Stevens DM, Saitoh M et al. mTOR
controls mitochondrial activity and biogenesis complex 2 is required for the development of pros-
through 4E-BP-dependent translational regula- tate cancer induced by Pten loss in mice. Cancer Cell.
tion. Cell Metab. 2013;18(5):698–711. 2009;15(2):148–159.
14. Sarbassov DD, Guertin DA, Ali SM et al. 30. Hemminki A, Markie D, Tomlinson I et al. A ­serine/
Phosphoryla­ t ion and regulation of Akt/PKB threonine kinase gene defective in Peutz-Jeghers
by the rictor-mTOR complex. Science. 2005;​ syndrome. Nature. 1998;391(6663):184–187.
307(5712):1098–1101. 31. Jenne DE, Reimann H, Nezu J et al. Peutz-Jeghers
15. Garcia-Martinez JM, Alessi DR. mTOR c­ omplex  2 syndrome is caused by  mutations in a novel serine
(mTORC2) controls hydrophobic motif phosphory­ threonine kinase. Nat Genet. 1998;18(1):38–43.
lation and activation of serum- and glucocorticoid- 32. Mihaylova MM, Shaw RJ. The AMPK signalling
induced protein kinase 1 (SGK1). Biochem J. 2008;​ pathway coordinates cell growth, autophagy and
416(3):375–385. metabolism. Nat Cell Biol. 2011;13(9):1016–1023.
16. Guertin DA, Stevens DM, Thoreen CC et al. Ablation 33. Sarkar S, Rubinsztein DC. Small molecule enhancers
in mice of the mTORC components raptor, rictor, or of autophagy for neurodegenerative diseases. Mol
mLST8 reveals that mTORC2 is required for signal- Biosyst. 2008;4(9):895–901.
ing to Akt-FOXO and PKCalpha, but not S6K1. Dev 34. Lamming DW, Ye L, Sabatini DM et al. Rapalogs
Cell. 2006;11(6):859–871. and mTOR inhibitors as anti-aging therapeutics.
17. Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY- J Clin Invest. 2013;​123(3):980–989.
22,989), a new antifungal antibiotic. I. Taxonomy 35. Wilkinson JE, Burmeister L, Brooks SV et al.
of the producing streptomycete and isolation of the Rapamycin slows aging in mice. Aging Cell.
active principle. J Antibiot. 1975;28(10):721–726. 2012;11(4):675–682.
18. Brown EJ, Albers MW, Shin TB et al. A mammalian 36. Harrison DE, Strong R, Sharp ZD et al. Rapamycin
protein targeted by G1-arresting rapamycin-receptor fed late in life extends lifespan in genetically hetero-
complex. Nature. 1994;369(6483):756–758. geneous mice. Nature. 2009;460(7253):392–395.
19. Sabatini DM, Erdjument-Bromage H, Lui M et al. 37. Selman C, Tullet JM, Wieser D et al. Ribosomal pro-
RAFT1: A mammalian protein that binds to FKBP12 tein S6 kinase 1 signaling regulates mammalian life
in a rapamycin-dependent fashion and is homolo- span. Science. 2009;326(5949):140–144.
gous to yeast TORs. Cell. 1994;78(1):35–43. 38. Lamming DW, Mihaylova MM, Katajisto P et al.
20. Martel RR, Klicius J, Galet S. Inhibition of the Depletion of Rictor, an essential protein component
immune response by rapamycin, a new antifungal of mTORC2, decreases male lifespan. Aging Cell.
antibiotic. Can J Physiol Pharmacol. 1977;55(1):48–51. 2014;13(5):911–917.
21. Ingle GR, Sievers TM, Holt CD. Sirolimus: 39. Shackelford DB, Shaw RJ. The LKB1-AMPK path-
Continuing the evolution of transplant immunosup- way: Metabolism and growth control in tumour sup-
pression. Ann Pharmacother. 2000;34(9):1044–1055. pression. Nat Rev Cancer. 2009;9(8):563–575.
22. Eng CP, Sehgal SN, Vezina C. Activity of rapamycin 40. Young S, Gooneratne S, Straus FH 2nd et al.
(AY-22,989) against transplanted tumors. J Antibiot. Feminizing Sertoli cell tumors in boys with Peutz-
1984;37(10):1231–1237. Jeghers syndrome. Am J Surg Pathol. 1995;19(1):50–58.
23. Lamming DW, Ye L, Katajisto P et al. Rapamycin- 41. Ulbright TM, Amin MB, Young RH. Intratubular
induced insulin resistance is mediated by mTORC2 large cell hyalinizing sertoli cell neoplasia of the
loss and uncoupled from longevity. Science. testis: A report of 8 cases of a distinctive lesion of
2012;335(6076):1638–1643. the Peutz-Jeghers syndrome. Am J Surg Pathol.
24. Sarbassov DD, Ali SM, Sengupta S et al. Prolonged 2007;31(6):827–835.
rapamycin treatment inhibits mTORC2 assembly 42. Huyghe E, Zairi A, Nohra J et al. Gonadal impact of
and Akt/PKB. Mol Cell. 2006;22(2):159–168. target of rapamycin inhibitors (sirolimus and evero-
25. Inoki K, Corradetti MN, Guan KL. Dysregulation limus) in male patients: An overview. Transpl Int.
of the TSC-mTOR pathway in human disease. Nat 2007;20(4):305–311.
Genet. 2005;37(1):19–24. 43. Vellai T, Takacs-Vellai K, Zhang Y et al. Genetics:
26. Saxton RA, Sabatini DM. mTOR Signaling in Influence of TOR kinase on lifespan in C. elegans.
growth, metabolism, and disease. Cell. 2017;168(6):​ Nature. 2003;426(6967):620.
960–976. 44. Gems D, Sutton AJ, Sundermeyer ML et al. Two
27. Feng Z, Zhang H, Levine AJ et al. The coordinate pleiotropic classes of daf-2 mutation affect larval
regulation of the p53 and mTOR pathways in cells. arrest, adult behavior, reproduction and longevity in
Proc Natl Acad Sci U S A. 2005;102(23):8204–8209. Caenorhabditis elegans. Genetics. 1998;150(1):129–155.
28. Li J, Yen C, Liaw D et al. PTEN, a putative pro- 45. Tissenbaum HA, Ruvkun G. An insulin-like sig-
tein tyrosine phos­phatase gene mutated in human naling pathway affects both longevity and repro-
brain, breast, and prostate cancer. Science. duction in Caenorhabditis elegans. Genetics.
1997;275(5308):1943–1947. 1998;148(2):703–717.
References 209

46. Malone EA, Thomas JH. A screen for nonconditional 61. Korta DZ, Tuck S, Hubbard E.J. S6K links cell fate, cell
dauer-constitutive mutations in Caenorhabditis ele- cycle and nutrient response in C. elegans germline stem/
gans. Genetics. 1994;136(3):879–886. progenitor cells. Development. 2012;139(5):859–870.
47. Patel DS, Garza-Garcia A, Nanji M et al. Clustering 62. Cid VJ, Rodríguez-Escudero I, Andrés-Pons A et al.
of genetically defined allele classes in the Assessment of PTEN tumor suppressor activity
Caenorhabditis elegans DAF-2 insulin/IGF-1 recep- in nonmammalian models: The year of the yeast.
tor. Genetics. 2008;178(2):931–946. Oncogene. 2008;27(41):5431–5442.
48. Geminard C, Rulifson EJ, Leopold P. Remote control 63. Hobbs RM, La HM, Mäkelä JA et al. Distinct
of insulin secretion by fat cells in Drosophila. Cell germline progenitor subsets defined through Tsc2-
Metab. 2009;10(3):199–207. mTORC1 signaling. EMBO Rep. 2015;16(4):467–480.
49. Pallares-Cartes C, Cakan-Akdogan G, Teleman AA. 64. Baker MD, Ezzati M, Aloisio GM et al. The small
Tissue-specific coupling between insulin/IGF and GTPase Rheb is required for spermatogenesis but not
TORC1 signaling via PRAS40 in Drosophila. Dev oogenesis. Reproduction. 2014;147(5):615–625.
Cell. 2012;22(1):172–182. 65. Wang C, Wang Z, Xiong Z et al. mTORC1 Activation
50. Bohni R, Riesgo-Escovar J, Oldham S et al. Promotes Spermatogonial Differentiation and Causes
Autonomous control of cell and organ size by Sub­fertility in Mice. Biol Reprod. 2016;95(5):97.
CHICO, a Drosophila homolog of vertebrate IRS1-4. 66. Serra ND, Velte EK, Niedenberger BA et al. Cell-
Cell. 1999;97(7):865–875. autonomous requirement for mammalian target of
51. Hietakangas V, Cohen SM. Re-evaluating AKT rapamycin (Mtor) in spermatogonial proliferation
regulation: Role of TOR complex 2 in tissue growth. and differentiation in the mousedagger. Biol Reprod.
Genes Dev. 2007;21(6):632–637. 2017;96(4):816–828.
52. Chen C, Jack J, Garofalo RS. The Drosophila 67. Lee J, Kanatsu-Shinohara M, Inoue K et al. Akt medi-
insulin receptor is required for normal growth. ates self-renewal division of mouse spermatogonial
Endocrinology. 1996;137(3):846–856. stem cells. Development. 2007;134(10):1853–1859.
53. Zhang H, Stallock JP, Ng JC et al. Regulation of cel- 68. Goertz MJ, Wu Z, Gallardo TD et al. Foxo1 is
lular growth by the Drosophila target of rapamycin required in mouse spermatogonial stem cells for
dTOR. Genes Dev. 2000;14(21):2712–2724. their maintenance and the initiation of spermato-
54. LaFever L, Feoktistov A, Hsu HJ et al. Specific roles of genesis. J Clin Invest. 2011;121(9):3456–3466.
Target of rapamycin in the control of stem cells and 69. Wang M, Guo Y, Wang M et al. The glial cell-derived
their progeny in the Drosophila ovary. Development. neurotrophic factor (GDNF)-responsive phospho-
2010;​137(13):2117–2126. protein landscape identifies raptor phosphorylation
55. Richard DS, Rybczynski R, Wilson TG et al. Insulin required for spermatogonial progenitor cell prolif-
signaling is necessary for vitellogenesis in Drosophila eration. Mol Cell Proteomics. 2017;16(6):982–997.
melanogaster independent of the roles of juvenile 70. Xiong M, Zhu Z, Tian S et al. Conditional ablation
hormone and ecdysteroids: Female sterility of the of Raptor in the male germ line causes infertility
chico1 insulin signaling mutation is autonomous to due to meiotic arrest and impaired inactivation of
the ovary. J Insect Physiol. 2005;51(4):455–464. sex chromosomes. FASEB J. 2017;31(9):3934–3949.
56. Sun P, Quan Z, Zhang B et al. TSC1/2 tumour 71. Bai S, Cheng L, Zhang Y et al. A germline-specific
suppressor complex maintains Drosophila germ- role for the mTORC2 component Rictor in maintain-
line stem cells by preventing differentiation. ing spermatogonial differentiation and intercellular
Development. 2010;137(15):2461–2469. adhesion in mouse testis. Mol Hum Reprod. 2018.
57. Jia K, Chen D, Riddle D.L. The TOR pathway inter- doi: 10.1093/molehr​/gay009. [Epub ahead of print]
acts with the insulin signaling pathway to regulate 72. Mok KW, Mruk DD, Lee WM et al. Rictor/mTORC2
C. elegans larval development, metabolism and life regulates blood-testis barrier dynamics via its effects
span. Development. 2004;131(16):3897–3906. on gap junction communications and actin filament
58. Long X, Spycher C, Han ZS et al. TOR deficiency in network. FASEB J. 2013;27(3):1137–1152.
C. elegans causes developmental arrest and intesti- 73. Denison FC, Smith LB, Muckett PJ et al. LKB1 is
nal atrophy by inhibition of mRNA translation. Curr an essential regulator of spermatozoa release dur-
Biol. 2002;​12(17):1448–1461. ing spermiation in the mammalian testis. PLoS One.
59. Webster CM, Wu L, Douglas D et al. A non-canonical 2011;6(12):e28306.
role for the C. elegans dosage compensation complex 74. Towler MC, Fogarty S, Hawley SA et al. A novel
in growth and metabolic regulation downstream short splice variant of the tumour suppressor
of TOR complex 2. Development. 2013;140(17):​ LKB1 is required for spermiogenesis. Biochem J.
3601–3612. 2008;416(1):1–14.
60. Saxe JP, Chen M, Zhao H et al. Tdrkh is essen- 75. Tanwar PS, Kaneko-Tarui T, Zhang L et al. Altered
tial for spermatogenesis and participates in pri- LKB1/AMPK/TSC1/TSC2/mTOR signaling causes
mary piRNA biogenesis in the germline. EMBO J. disruption of Sertoli cell polarity and spermatogen-
2013;32(13):1869–1885. esis. Hum Mol Genet. 2012;21(20):4394–4405.
210  Roles of mTOR signaling in spermatogenesis

76. Dong H, Chen Z, Wang C et al. Rictor regulates sper- 91. Wong CH, Cheng CY. The blood-testis barrier: Its
matogenesis by controlling Sertoli cell cytoskeletal biology, regulation, and physiological role in sper-
organization and cell polarity in the mouse testis. matogenesis. Curr Top Dev Biol. 2005;71:263–296.
Endocrinology. 2015;156(11):4244–4256. 92. Mok KW, Mruk DD, Cheng CY. Regulation of
77. Schell C, Kretz O, Liang W et al. The Rapamycin- blood-testis barrier (BTB) dynamics during sper-
Sensitive Complex of Mammalian Target of matogenesis via the “Yin” and “Yang” effects of mam-
Rapamycin Is Essential to Maintain Male Fertility. malian target of rapamycin complex 1 (mTORC1) and
Am J Pathol. 2016;186(2):​324–336. mTORC2. Int Rev Cell Mol Biol. 2013;301:291–358.
78. Oatley JM, Brinster RL. The germline stem cell 93. Jesus TT, Oliveira PF, Sousa M et al. Mammalian target
niche unit in mammalian testes. Physiol Rev. of rapamycin (mTOR): A central regulator of male fer-
2012;92(2):577–595. tility? Crit Rev Biochem Mol Biol. 2017;52(3):235–253.
79. Meng X, Lindahl M, Hyvönen ME et al. Regulation 94. Oliveira PF, Cheng CY, Alves MG. Emerging Role
of cell fate decision of undifferentiated spermatogo- for  Mammalian Target of Rapamycin in Male Fer­
nia by GDNF. Science. 2000;287(5457):1489–1493. tility. Trends Endocrinol Metab. 2017;28(3):165–167.
80. Oatley JM, Avarbock MR, Brinster RL. Glial cell 95. Riera MF, Regueira M, Galardo MN et al. Signal
line-derived neurotrophic factor regulation of genes transduction pathways in FSH regulation of rat
essential for self-renewal of mouse spermatogonial Sertoli cell proliferation. Am J Physiol Endocrinol
stem cells is dependent on Src family kinase signal- Metab. 2012;302(8):E914–E923.
ing. J Biol Chem. 2007;282(35):25842–25851. 96. Yang WR, Wang Y, Wang Y et al. mTOR is involved
81. Feng LX, Ravindranath N, Dym M. Stem cell factor/ in 17beta-estradiol-induced, cultured immature
c-kit up-regulates cyclin D3 and promotes cell cycle boar Sertoli cell proliferation via regulating the
progression via the phosphoinositide 3-kinase/p70 expression of SKP2, CCND1, and CCNE1. Mol
S6 kinase pathway in spermatogonia. J Biol Chem. Reprod Dev. 2015;82(4):305–314.
2000;275(33):25572–25576. 97. Nogueira V, Hay N. Molecular pathways: Reactive
82. Hobbs RM, Seandel M, Falciatori I et al. Plzf regu- oxygen species homeostasis in cancer cells and
lates germline progenitor self-renewal by opposing implications for cancer therapy. Clin Cancer Res.
mTORC1. Cell. 2010;142(3):468–479. 2013;19(16):4309–4314.
83. Zhou Z, Shirakawa T, Ohbo K et al. RNA bind- 98. Jesus TT, Oliveira PF, Silva J et al. Mammalian tar-
ing protein Nanos2 organizes post-­t ranscriptional get of rapamycin controls glucose consumption and
buffering system to retain primitive state of redox balance in human Sertoli cells. Fertil Steril.
mouse spermatogonial stem cells. Dev Cell. 2015;​ 2016;105(3):825–833 e3.
34(1):96–107. 99. Boyer A, Girard M, Thimmanahalli DS et al. mTOR
84. Busada JT, Niedenberger BA, Velte EK et al. regulates gap junction Alpha-1 protein trafficking in
Mammalian target of rapamycin complex 1 Sertoli cells and is required for the maintenance of
(mTORC1) is required for mouse spermatogonial spermatogenesis in mice. Biol Reprod. 2016;95(1):13.
differentiation in vivo. Dev Biol. 2015;407(1):90–102. 100. Gao Y, Chen H, Lui WY et al. Basement mem-
85. Xiong M, Ferder IC, Ohguchi Y et al. Quantitative brane laminin alpha2 regulation of BTB dynam-
analysis of male germline stem cell differen- ics via its effects on f-actin and microtubule
tiation reveals a role for the p53-mTORC1 path- cytoskeletons is mediated through mTORC1 signal-
way in spermatogonial maintenance. Cell Cycle. ing. Endocrinology. 2017;158(4):963–978.
2015;14(18):2905–2913. 101. Mok KW, Mruk DD, Silvestrini B et al. rpS6 regu-
86. Lie PP, Mruk DD, Lee WM et al. Cytoskeletal lates blood-testis barrier dynamics by affecting F-actin
dynamics and spermatogenesis. Philos Trans R Soc organization and protein recruitment. Endocrinology.
Lond B Biol Sci. 2010;365(1546):1581–1592. 2012;153(10):5036–5048.
87. Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli- 102. Shaw RJ, Bardeesy N, Manning BD et al. The LKB1
germ cell interactions and their significance in tumor suppressor negatively regulates mTOR signal-
germ cell movement in the seminiferous epi- ing. Cancer Cell. 2004;​6(1):91–99.
thelium during spermatogenesis. Endocr Rev. 103. Yilmaz OH, Katajisto P, Lamming DW et al.
2004;25(5):747–806. mTORC1 in the Paneth cell niche couples intes-
88. Holembowski L, Kramer D, Riedel D et al. TAp73 is tinal stem-cell function to calorie intake. Nature.
essential for germ cell adhesion and maturation in 2012;486(7404):490–495.
testis. J Cell Biol. 2014;204(7):1173–1190. 104. Helsel AR, Oatley MJ, Oatley JM. Glycolysis-optimized
89. Mruk DD, Cheng CY. Tight junctions in the testis: conditions enhance maintenance of regenerative
New perspectives. Philos Trans R Soc Lond B Biol Sci. integrity in mouse spermatogonial stem cells during
2010;365(1546):1621–1635. long-term culture. Stem Cell Rep. 2017;8(5):1430–1441.
90. Mruk DD, Cheng CY. The mammalian blood-­testis 105. Ramanathan A, Schreiber SL. Direct control of
barrier: Its biology and regulation. Endocr Rev. mitochondrial function by mTOR. Proc Natl Acad
2015;36(5):564–591. Sci U S A. 2009;106(52):22229–22232.
Does planar cell polarity matter during
spermatogenesis?
17
LINXI LI, HAIQI CHEN, QINGQUAN LIAN, REN-SHAN GE, and C. YAN CHENG

INTRODUCTION classified into (1) PCP core proteins (e.g., Van Gogh-like
Cell polarity is important to spermatogenesis since it con- [Vangl] protein such as Vangl2, dishevelled [Dvl] such as
fers apico-basal (A/B) polarity in Sertoli cells and devel- Dvl2, and frizzled [Fzd] class receptor such as Fzd 3 and
oping spermatids in the seminiferous epithelium during Fzd5), (2) PCP ligands (e.g., Wnt5a), (3) PCP effectors (e.g.,
the epithelial cycle of spermatogenesis1–3 (Figure 17.1). fuzzy, multiple wing hair [Mwh], Fritz, inturned), and
This is important and necessary so that the maximal PCP signaling proteins (e.g., dachsous cadherin-related
number of developing germ cells can be packed within protein [Dchs] such as Dchs1).7,11,12 Due to the indispens-
the limited space of the seminiferous epithelium in semi- able role of PCP proteins to confer axial versus vectorial
niferous tubules, such that millions of sperm can be pro- asymmetry during embryonic development (Figure 17.2),
duced in the testes of an adult male in rodents or humans. PCP proteins are very well conserved and are found from
Furthermore, cell polarity also confers A/B polarity to worms, flies, and mammals, and their sequences are well
developing cells and tissues during embryogenesis and conserved. While PCP proteins are important to confer
morphogenesis.4 However, besides cell polarity, studies PCP alignment of developing spermatids in the Sertoli cell
have shown that planar cell polarity (PCP) is essential to epithelium, PCP proteins have not been studied in the tes-
many cellular processes, in particular convergent exten- tis regarding their role in spermatogenesis until recently.
sion during embryogenesis such as gastrulation when tis- Interestingly, virtually all major PCP proteins in each of
sue narrows (i.e., converges) along one axis alongside with the four classes of PCP have been detected in the testis.13
elongation (i.e., extension) along a perpendicular axis due Herein, we sought to carefully evaluate the currently avail-
to polarized cell movement to generate antero-posterial able data in the field, which will provide important guides
(A/P) axis.5–7 In short, PCP proteins are important to and insightful information for future studies so that the
establish cell polarity within an epithelial plane and reg- role of PCP proteins in spermatogenesis can be carefully
ulates tissue morphogenesis during embryonic and neo- delineated.
natal development.8 Earlier studies have mostly focused
on the role of PCP proteins in regulating directional cell
PCP PROTEINS AND SPERMATOGENESIS
movement during embryogenesis, such as neutral tube
formation, and asymmetric orientation of cell projections, Background
such as cilia in cochlear hair cells.8 PCP proteins are also Much of the information regarding PCP proteins is
important to the assembly of cilia in multiciliated cells of based on studies in cuticular hair in Drosophila. Table
the skin and on neuroepithelial cells of the neural tube 17.1 summarizes the best-studied core PCP proteins/
during development.9 In adult animals, PCP is most nota- genes found in vertebrates, the corresponding homologs
bly detected in cells and/or tissues wherein PCP refers to in Drosophilia. Table 17.1 also summarizes their func-
the alignment of a field of polarized cells (e.g., elongating/ tion in rodents including their role in the testis regard-
elongated spermatids) within the plane of a cell epithe- ing spermatogenesis/fertility based on studies using gene
lium such as the Sertoli cell epithelium in the seminiferous knockdown approach by RNAi in rats. Also listed are
tubule, mostly notably in stage VII-VIII of the epithelial diseases caused by their mutations and/or deletions in
cycle (Figure 17.1)10 versus hair cell alignment on skin or humans (Table 17.1). As shown in Figure 17.2, alignment
cochlea. Without the coordinated efforts of polarity pro- of PCP proteins can be either axial (or bipolar) asymme-
teins and PCP proteins in the seminiferous epithelium try or vectorial (or unipolar) asymmetry.7,11 It is generally
to confer the proper alignments of polarized spermatids accepted that axial asymmetry is conferred by integral
across the plane of Sertoli cells, the tremendous cellular membrane proteins Celsr (or flamingo [Fmi] in inverte-
output of generating millions of spermatozoa per testis brates) and furrowed (Fw) (no equivalent homolog has
on a daily basis is not sustainable. In this context, it is of been found in vertebrates thus far) in which they local-
interest to note that spermatid PCP is highly sensitive to ize to both poles along one planar axis (Figure 17.2).7,11
toxicants such as environmental toxicants (e.g., cadmium Vectorial asymmetry is conferred by integral membrane
chloride) or male contraceptives (e.g., adjudin) since a proteins frizzled (Fzd) and Van Gogh (Vang, or Vangl,
brief exposure of the testis to these toxicants can induce such as Vangl1, Vangl2) in which they localize prefer-
rapid loss of PCP (Figure 17.1). Studies in other mam- entially to one pole of each cell (Figure 17.2).7,11 Also,
malian epithelia have shown that PCP proteins can be Diego (Dgo) and dishevelled (Dsh) are the cytoplasmic

211
212  Does planar cell polarity matter during spermatogenesis?

Toxicants

CdCl2
(3 mg/kg b.w., i.p.)

Control adult rat testis CdCl2 (16 hr)

Adjudin
(50 mg/kg b.w., oral gavage)

Control adult rat testis Adjudin (12 hr)

Figure 17.1  A schematic drawing that illustrates features of PCP regarding the alignment of a field of polarized developing
spermatids within the plane of the Sertoli cell epithelium in adult rat testes. In the top left panel, PCP is noted in this schematic
drawing in which a field of polarized spermatid heads are aligned across the plane of a Sertoli cell epithelium. This is also supported
by fluorescence microscopy and histological analysis using cross sections of adult rat testes on the middle left and lower left panel,
respectively. In the corresponding right panel, spermatids had lost their PCP in which spermatid heads displayed polarity defects
following treatment of rats with either environmental toxicants (e.g., cadmium chloride, 3 mg/kg b.w. via i.p.) or a male contraceptive
(e.g., adjudin, 50 mg/kg b.w. by oral gavage). Besides defects in spermatid polarity (spermatids with polarity defects are annotated
by yellow arrowheads), many elongated spermatids remained trapped deep inside the epithelium (such as following adjudin treat-
ment) due to a disorganization of actin microfilaments and also microtubules (MTs) across the epithelium, and failure to support
proper transport of spermatids to the adluminal edge of the apical compartment to prepare these spermatids for their release at
spermiation in late stage VIII tubules. Scale bar, 40 µm in fluorescence images; 50 µm in hematoxylin and eosin stained images.
PCP proteins and spermatogenesis  213

Mwh Actin

Rho GTPase

BTB
Sertoli cell

Vangl2 Prickle (pk)

Frizzled (Fzd) Dishevelled (dvl)

Celsr Inversion (invs)


Axial asymmetry Vectorial asymmetry
Actin filament bundles

Figure 17.2  Asymmetric distribution of core PCP proteins. Asymmetrical distribution of core PCP proteins confers either axial
asymmetry or vectorial asymmetry in cells, such as in cuticular hair cells in Drosophila or cochlea hair cells in mammals. It is noted that
Celsr (cadherin EGF LAG seven-pass G-type receptor including Celsr 1, Celsr 2, and Celsr 3) is the mammalian homolog of Flamingo
(Fmi, also known as starry night [stan]) found in Drosophila, which displays axial asymmetry in which Celsr can be localized to inter-
cellular junctions along one tissue axis but excluded from orthogonal junctions. However, Van Gogh (Vang, such as Van Gogh-like
2, Vangl2)-Prickle (Pk) and Frizzled (Fzd or Fz in Drosophila)-Dishevelled (Dsh in Drosophila or Dvl in vertebrates)-Diego (Dgo, its
homolog is Inversin [Invs] in mammals) protein complexes display vectorial asymmetry in which each protein complex localizes to
the opposite end of the cell. The enlarged image depicts the Fzd/PCP core component interactions across a two-cell border in mam-
malian cells such as in Sertoli cells. Based on studies in Drosophila and also in Sertoli cells (see text for details), it is now known that
the Vangl2/Pk complex exerts its regulatory effects to promote actin filament disorganization via an inhibition of actin polymeriza-
tion through multiple wing hairs (Mwh). However, the homolog of Mwh remains unknown in the testis. In contrast, the Fzd/Dvl/Invs
protein complex promotes actin organization through its action on Rho GTPase by inducing actin polymerization. It is known that
Vangl2 and Celsr or Vangl2 and Fzd can antagonize each other as depicted herein.

components of Fz, whereas prickle (Pk) is the cytoplas- effectors (e.g., Rho-associated kinase [ROK])7,11,46,47
mic component of Vang. It is of interest to note that Dgo/ (Figure 17.2). These findings thus suggest that the Vang/
Dsh and Pk antagonize each other and are capable of acti- Pk complex and the Fz/Dgo/Dsh complex have antago-
vating separate intracellular signaling pathways, playing nistic effects on cellular F-actin polymerization.
an important role in maintaining the vectorial asymme-
try (Figure 17.2). The Vang/Pk complex and the Fz/Dgo/ Study in vitro
Dsh complex are working in concert to regulate vectorial In this context, it is of interest to note that in a recent
asymmetry through their corresponding action on actin- report, Vangl2 (a homolog of Vang) was shown to inhibit
based cytoskeletal function (Figure 17.2). It is now gener- actin-based cytoskeletal function in the testis13 similar to
ally accepted that the Vang/Pk complex recruits Mwh (a the function of Vang/Pk complex in Drosophila, by pro-
PCP effector molecule12)45 to exert its regulatory effects moting F-actin disorganization in Sertoli cells, making
by inhibiting polymerization of actin microfilaments, the Sertoli cell tight junction (TJ) barrier “leaky.”13 For
whereas the Fz/Dgo/Dsh complex promotes polymeriza- instance, following overexpression of Vangl2 in Sertoli
tion of actin microfilaments through activation and/or cell epithelium with an established functional TJ perme-
recruitment of Rho-family GTPases and the downstream ability barrier that mimicked the Sertoli cell blood-testis
214  Does planar cell polarity matter during spermatogenesis?

Table 17.1  Core PCP proteins/genes in Drosophilia and vertebrates.


Drosophila Vertebrate
genes genes Function in rodents Disease found in humans via mutation
Disheveled Dvl1 Regulates gut microbiota composition to Autosomal-dominant Robinow syndrome15
(dsh) confer intestinal homeostasis14 Inflammatory bowel disease16
Dvl2 Involves in depression using a mouse social Inflammatory bowel disease18
model17
Dvl3 Involves in heart development19 Autosomal-dominant Robinow syndrome20
Frizzled (fz) Fzd1 Oocyte maturation and cumulus cell function Inflammatory bowel disease,18 diabetic kidney
in mice21 disease22
Fzd2 Diabetic kidney disease22
Fzd3 Axonal development23 Inflammatory bowel disease18
Fzd4 Inflammatory bowel disease18
Fzd5 Inflammatory bowel disease,18 diabetic kidney
disease22
Fzd6 Diabetic kidney disease22
Fzd7 Serves as Clostridium difficile toxin B (TcdB) Diabetic kidney disease22
receptor in mice24
Fzd8 Involved in arthritis pathogenesis based on an Involved in drug resistance in breast cancer
adjuvant-induced arthritis (AIA) model in pathogenesis26
rats25
Fzd9 Hippocampal development,27 osteoblast
function28
Fzd10 Breast cancer metastasis29
Van Gogh-like Vangl1 Neural tube development 30 Neural-tube defects31
(Vangl) Vangl2 Neural tube development, lung 30 Neural-tube defects33
morphogenesis, and a regulator of
32

ectoplasmic specialization dynamics in adult


rat testes by promoting blood-testis barrier
remodeling13
Flamingo (fmi) Celsr1 Lung morphogenesis during embryogenesis32 Induces ischemic stroke,34 and involved in
Huntington’s disease pathogenesis35
Celsr2 Facilitates circulation of the cerebrospinal An increase in blood low-density lipoprotein
fluid36 cholesterol37 or an increase in serum
cholesterol,38 involved in Huntington’s disease
pathogenesis35
Celsr3 Facilitates circulation of the cerebrospinal Involved in Huntington’s disease
fluid36 and axonal development23 pathogenesis35
Prickle (pk) Pk1 Neuronal morphogenesis, its deletion leads Involved in progressive myoclonus epilepsy40
39

to seizure in mice40
Pk2 Susceptible to seizures after knockout in mice40 Involved in progressive myoclonus epilepsy40
Diego (dgo) Diversin Deletion causes PCP defects in inner ear Neural tube defects42
(ankrd6) sensory organs 41

inversin (invs) Deletion leads to defects in kidney and urinary Involved in nephronophthisis pathogenesis44
tract development43

barrier (BTB) in vivo, long stretches of actin microfila- and ß-catenin all became dislocalized, and this thus
ments across the Sertoli cell cytosol were grossly disrupted contributed to the TJ permeability barrier disruption.13
since they were defragmented.13 The truncation of actin Most importantly, the overexpression of Vangl2-induced
microfilaments, in turn, perturbed Sertoli cell adhesive F-actin disorganization was shown to be mediated by
function. This is because BTB-associated adhesion protein two actin regulatory proteins wherein (1) Eps8 (an actin
complexes such as claudin 11-ZO-1 and N-cadherin-ß- barbed end capping and bundling protein) was found to
catenin all utilized F-actin for attachment. The disrup- redistribute from the cell-cell interface to the Sertoli cell
tion of F-actin thereby impeded the function of these cell cytosol, and (2) Arp3 (an actin barbed end nucleation pro-
adhesion proteins. Thus, claudin 11, ZO-1, N-cadherin, tein that effectively converts bundled actin filaments to an
Concluding remarks and future perspectives  215

unbundled/branched configuration) was shown to become the epithelium. For instance, phagosomes derived from the
more intensely localized to the cell cortical zone.13 These engulfment of residual bodies by Sertoli cells that should
changes thus contribute to extensive truncation of actin have been transported into the base of the seminiferous
microfilaments at the Sertoli cell cortical zone following epithelium for their eventual degradation through the lyso-
overexpression of Vangl2, making the TJ-barrier “leaky.” somal degradation pathway  48,49 were persistently found near
In contrast, a knockdown of Vangl2 in the Sertoli cell epi- the luminal edge of the adluminal compartment in stages
thelium by RNAi to transiently silence Vangl2 expression IX and X tubules in testes following Vangl2 knockdown.13
led to (1) a redistribution of Eps8, which became intensely Additionally, the defects in organizing the F-actin network
localized to the Sertoli cell-cell interface, and (2) a reduced at the Sertoli-spermatid interface in response to changes in
expression of Arp3 at the Sertoli cell cortical zone.13 These the epithelial cycle following Vangl2 knockdown also per-
changes contribute to a strengthening of the actin micro- turbed apical ES plasticity, perturbing spermatid adhesion
filaments at the Sertoli cell basal ES/BTB, “tightening” and spermatid PCP so that considerable spermatids with
the TJ barrier. As such, a knockdown of Vangl2 in Sertoli defects in polarity were noted13 (Figure 17.3). In this context,
cell epithelium by RNAi was indeed found to promote the it is of interest to note that environmental toxicants (e.g.,
Sertoli cell TJ permeability barrier, making the TJ bar- cadmium chloride) or contraceptives (e.g., adjudin) are also
rier tighter.13 Studies by immunofluorescence microscopy shown to perturb spermatid PCP (Figure 17.1) since these
also support the concept that Vangl2 per se in the testis toxicants, similar to Vangl2, also exert their disruptive
promotes actin microfilament disorganization since its effects by altering the organization of actin microfilaments
knockdown by RNAi indeed was found to promote the at the ES, thereby perturbing PCP function.
recruitment of TJ protein complex (e.g., claudin 11/ZO-1) In short, Vangl2 promotes BTB disruption since a
and basal ES protein complex (e.g., N-cadherin/ß-catenin) knockdown of Vangl2 in Sertoli cells in vitro makes the
at the Sertoli cell-cell interface due to better maintenance Sertoli cell TJ barrier “tighter,” whereas its overexpres-
of actin filament organization at the cell cortical zone.13 In sion causes the TJ barrier to be “leaky” through changes
short, these findings thus suggest that Vangl2 in the nor- in the organization of actin microfilaments in Sertoli
mal testis likely promotes actin filament disorganization cells13 (Figure 17.3). In brief, Vangl2, the integral mem-
during the epithelial cycle of spermatogenesis, such as in brane protein of the Vangl2/Pk complex, is a core PCP
stage VIII tubules to facilitate the transport of prelepto- protein in the seminiferous epithelium, which exerts
tene spermatocytes across the immunological barrier. its regulatory effects through the actin-based cytoskel-
eton. F-actin is not only crucial to spermatid and Sertoli
Study in vivo cell adhesion in the seminiferous epithelium, but also
Studies in vitro as briefly discussed above have illustrated intracellular trafficking events, such as the transport of
the significance of Vangl2, and also other PCP proteins, to residual bodies and phagosomes during the epithelial
the organization of actin microfilaments in the seminifer- cycle of spermatogenesis.50,51 Therefore, it is not entirely
ous epithelium during the epithelial cycle. However, it is unexpected that a knockdown of Vangl2 in the testis in
envisioned that a delicate balance of Vangl2 across the sem- vivo by RNAi also perturbed the transport of residual
iniferous epithelium is needed to support spermatogenesis, bodies/phagosomes across the epithelium at stage VIII
in particular through its changes in spatiotemporal expres- of the cycle13 so that they can be degraded via lysosomes
sion, but also with the involvement of other PCP proteins near the base of the stage IX tubules. In this context, it is
that contribute to the proper organization of actin micro- of interest to note that the frequency of meiosis I/II was
filaments at different stages of the epithelial cycle. When also grossly reduced at stage XIV of the epithelial cycle.13
Vangl2 was knocked down by ~50% using Vangl2-specific Since meiosis is also an F-actin-dependent cellular event
siRNA duplexes for transfection in the adult rat testis in by supporting the segregation of the genetic material
vivo, considerable changes in the status spermatogenesis across the cell cytosol at anaphase, thus a Vangl2 knock-
were noted13 (Figure 17.3). For instance, following Vangl2 down that impedes F-actin organization would perturb
knockdown in the testis in vivo, defects in spermatid and meiosis. Based on the available data and information
phagosome transport were found in ~40% of the stages in the literature, we propose that Vangl2 is working in
IX-X tubules.13 For instance, many step 19 spermatids were concert with other core PCP proteins, such as Fzd/Dvl/
found embedded deep inside the epithelium in both stage Invs and Celsr, to induce BTB restructuring and changes
IX and X tubules when spermiation had occurred.13 This is in apical ES function to support the transport of prelep-
due to the persistent presence of residual F-actin network at totene spermatocytes across the immunological barrier
the apical ES to support spermatid adhesion when it should and the release of sperm at spermiation, respectively, as
have been degenerated completely to facilitate the release noted in Figure 17.3.
of sperm at spermiation.13 This observation is also con-
sistent with the findings in vitro that Vangl2 knockdown CONCLUDING REMARKS AND FUTURE PERSPECTIVES
supported the actin filament organization at the basal ES/ In brief, Vangl2 regulates PCP function in the testis
BTB to make the BTB barrier “tighter”13 (Figure 17.3). This through its effects on the actin-based cytoskeleton. It is
unwanted persistence of the F-actin network at the ES in the noted that Sertoli cell adhesion that contributes to the
epithelium also perturbed proper organelle transport across BTB integrity is mediated predominantly by the actin
216  Does planar cell polarity matter during spermatogenesis?

Spermiation

Degenerating
apical ES

Sertoli cells

Germ cell
transport
Intact
apical ES

Intact Remodeling
BTB BTB

Sertoli cells
Preleptotene
Preleptotene spermatocytes
spermatocytes

Vangl2 Prickle (Pk) Actin filament bundles

Frizzled (Fzd) Dishevelled (Dvl) Actin filaments


Celsr Inversin (Invs) Branched actin network

Figure 17.3  A schematic drawing that illustrates the likely mechanism by which core PCP proteins regulate ectoplasmic special-
ization (ES) dynamics to support germ cell transport in the testis. Actin microfilament organization at the apical and the basal ES
integrity is maintained by Vangl2, since its overexpression promotes Sertoli cell TJ-barrier disruption. In contrast, Vangl2 silencing
by RNAi in the testis in vivo also impedes spermatid transport due to changes in actin microfilament organization at the ES (see text
for details). In short, an upregulation of Vangl2 promotes ES disruption, which in turn facilitates the transport of preleptotene sper-
matocytes across the immunological barrier and the release of spermatids at spermiation.

microfilament bundles at the basal ES (which together testis as they are in Drosophila. It is likely that the Fz/Dvl
with TJ create the BTB).52–57 Additionally, spermatid complex is found in the testis to promote F-actin polym-
(steps 8–19 or 8–16 spermatids in the rat or mouse testis, erization to maintain the Sertoli TJ barrier integrity by
respectively) adhesion to the Sertoli cell in the seminif- making the barrier “tighter” and to maintain spermatid
erous epithelium is also supported by the microtubule adhesion during spermiogenesis (Figure 17.3). As such,
(MT)-based cytoskeleton at the apical ES.58,59 Thus, it the Vangl2/Pk and the Fz/Dsh complexes are working
remains to be determined regarding the role of Vangl2 partners to regulate apical and basal ES dynamics (i.e.,
in MT dynamics in the testis. Furthermore, a number of disassembly, reassembly, and stabilization), support-
issues remain to be investigated: (1) if Pk is a functional ing not just adhesion of Sertoli cells and spermatids,
partner of Vangl2 in the testis, (2) if Mwh is involved in but also intracellular trafficking events through their
the Vangl2/Pk complex functionally downstream, and effects on F-actin organization. This speculation is not
(3) if other protein partners and/or signaling proteins are entirely unexpected since Dsh has been shown to work
involved in the Vangl2-induced ES remodeling. Based with clathrin AP-2 to modulate Fz endocytosis60 and is
on studies of the other PCP core proteins, such as the involved in the endocytosis of the Wnt/ß-catenin signal-
Fz/Dsh complex, which is known to play an important ing complex,61 illustrating its role in endocytic protein
role to promote actin polymerization,7,11 it remains to be trafficking. In fact, studies have shown that the PCP
determined if the Fz/Dvl (note that dishevelled is sym- pathway plays a crucial role in endocytic protein traffick-
bolized as Dvl in vertebrates versus Dsh in Drosophila) ing,62 which is crucial to support spermatogenesis so that
complex is working in concert with the Vangl2/Pk com- proteins, in particular apical and basal ES proteins, can
plex to confer vectorial asymmetry in the mammalian be recycled without overwhelming the limited number
References 217

of Sertoli cells in the testis.59,63 In short, protein traffick- 13. Chen H, Mruk DD, Lee WM, Cheng CY. Planar cell
ing events must be tightly regulated since it is estimated polarity (PCP) protein Vangl2 regulates ectoplasmic
that a single Sertoli cell provides nutritional, hormonal, specialization dynamics via its effects on actin micro-
paracrine, and structural support to ~30 to 50 develop- filaments in the testes of male rats. Endocrinology.
ing germ cells during spermatogenesis.64 These possi- 2016;157:2140–2159.
bilities must be carefully evaluated in future studies. In 14. Belinson H, Savage AK, Fadrosh D et al. Dual epithe-
short, better understanding of the role of PCP proteins in lial and immune cell function of Dvl1 regulates gut
the seminiferous epithelium during the epithelial cycle microbiota composition and intestinal homeostasis.
is crucial to unravel the biology of spermatogenesis, to JCI Insight. 2016;1.
develop new approaches for nonhormonal male contra- 15. White J, Mazzeu JF, Hoischen A et al. DVL1 frame-
ception, and to restore spermatogenesis in infertile men. shift mutations clustering in the penultimate exon
cause autosomal-dominant Robinow syndrome. Am
ACKNOWLEDGMENTS J Hum Genet. 2015;96:612–622.
This work was supported by grants from the National 16. You XJ, Bryant PJ, Jurnak F, Holcombe RF. Expression
Institutes of Health, NICHD R01 HD056034 to C.Y.C., of Wnt pathway components frizzled and disheveled
and U54 HD029990 Project 5 to C.Y.C.; National in colon cancer arising in patients with inflamma-
Natural Science Foundation of China (NSFC 81601264 tory bowel disease. Oncol Rep. 2007;18:691–694.
to L.L.), Zhejiang Provincial Natural Science Foundation 17. Wilkinson MB, Dias C, Magida J et al. A novel role of
(LQ16H040004 to L.L.), Department of Education of the WNT-dishevelled-GSK3beta signaling cascade
Zhejiang Province (Y201534170 to L.L.). in the mouse nucleus accumbens in a social defeat
model of depression. J Neurosci. 2011;31:9084–9092.
REFERENCES 18. You J, Nguyen AV, Albers CG, Lin F, Holcombe RF.
1. Gao Y, Cheng CY. Does cell polarity matter during Wnt pathway-related gene expression in inflamma-
spermatogenesis? Spermatogenesis. 2016;6:e1218408. tory bowel disease. Digest Dis Sci. 2008;53:1013–1019.
2. Wong EWP, Cheng CY. Polarity proteins and cell- 19. Etheridge SL, Ray S, Li SD et al. Murine dishevelled
cell interactions in the testis. Int Rev Cell Mol Biol. 3 functions in redundant pathways with Dishevelled
2009;278:309–353. 1 and 2 in normal cardiac outflow tract, cochlea, and
3. Brill JA, Yildirim S, Fabian L. Phosphoinositide sig- neural tube development. PLoS Genet. 2008;4.
naling in sperm development. Semin Cell Dev Biol. 20. White JJ, Mazzeu JF, Hoischen A et al. DVL3 alleles
2016;59:2–9. resulting in a -1 frameshift of the last exon medi-
4. Roman-Fernandez A, Bryant DM. Complex polarity: ate autosomal-dominant Robinow syndrome. Am J
Building multicellular tissues through apical mem- Hum Genet. 2016;98:553–561.
brane traffic. Traffic. 2016;17:1244–1261. 21. Lapointe E, Boyer A, Rico C et al. FZD1 regulates
5. Davenport D. The cell biology of planar cell polarity. cumulus expansion genes and is required for normal
J Cell Biol. 2014;207:171–179. female fertility in mice. Biol Reprod. 2012;87:104.
6. Sokol SY. Spatial and temporal aspects of Wnt sig- 22. Woroniecka KI, Park AS, Mohtat D, Thomas DB,
naling and planar cell polarity during vertebrate Pullman JM, Susztak K. Transcriptome analy-
embgyronic development. Semin Cell Dev Biol. sis of human diabetic kidney disease. Diabetes.
2015;42:78–85. DOI:101016/jsemcdb201505002. 2011;60:2354–2369.
7. Aw WY, Devenport D. Planar cell polarity: Global 23. Tissir F, Bar I, Jossin Y, Goffinet AM. Protocadherin
inputs establishing cellular asymmetry. Curr Opin Celsr3 is crucial in axonal tract development. Nat
Cell Biol. 2016;44:110–116. Neurosci. 2005;8:451–457.
8. Goodrich LV, Strutt D. Principles of planara 24. Tao L, Zhang J, Meraner P et al. Frizzled proteins are
polarity in animal development. Development. colonic epithelial receptors for C. difficile toxin B.
2011;138:1877–1892. Nature. 2016;538:350–355.
9. Park TJ, Haigo SL, Wallingford JB. Ciliogenesis 25. Miao CG, Shi WJ, Xiong YY et al. miR-375 regulates
defects in embryos lacking inturned or fuzzy the canonical Wnt pathway through FZD8 silenc-
function are associated with failure of planar ing in arthritis synovial fibroblasts. Immunol Lett.
cell polarity and Hedgehog signaling. Nat Genet. 2015;164:1–10.
2006;38:303–311. 26. Yin S, Xu L, Bonfil RD et al. Tumor-initiating cells
10. Chen H, Cheng CY. Planar cell polarity (PCP) pro- and FZD8 play a major role in drug resistance in
teins and spermatogenesis. Semin Cell Dev Biol. triple-negative breast cancer. Mol Cancer Ther.
2016;59:99–109. 2013;12:491–498.
11. Yang Y, Mlodzik M. Wnt-Frizzled/planar cell polar- 27. Zhao C, Aviles C, Abel RA, Almli CR, McQuillen P,
ity signaling: Cellular orientation by facing the wind Pleasure SJ. Hippocampal and visuospatial learn-
(Wnt). Annu Rev Cell Dev Biol. 2015;31:623–646. ing defects in mice with a deletion of frizzled 9, a
12. Maung SM, Jenny A. Planar cell polarity in gene in the Williams syndrome deletion interval.
Drosophila. Organogenesis. 2011;7:165–179. Development. 2005;132:2917–2927.
218  Does planar cell polarity matter during spermatogenesis?

28. Albers J, Schulze J, Beil FT et al. Control of bone for- 43. Negrisolo S, Carraro A, Fregonese G et al. Inversin
mation by the serpentine receptor Frizzled-9. J Cell (INVS) as a candidate gene in renal hypodysplasia
Biol. 2011;192:1057–1072. identified by whole exome sequencing approach.
29. Gong C, Qu S, Lv XB et al. BRMS1L suppresses breast Nephrol Dial Transplant. 2016;31(Suppl 1):i90–i99
cancer metastasis by inducing epigenetic silence of (Abstract; DOI:10.1093/ndt/gfw156.30).
FZD10. Nat Commun. 2014;5:5406. 44. Otto EA, Schermer B, Obara T et al. Mutations in
30. Torban E, Patenaude AM, Leclerc S et al. Genetic INVS encoding inversin cause nephronophthisis
interaction between members of the Vangl family type 2, linking renal cystic disease to the function of
causes neural tube defects in mice. Proc Natl Acad primary cilia and left-right axis determination. Nat
Sci U S A. 2008;105:3449–3454. Genet. 2003;34:413–420.
31. Kibar Z, Torban E, McDearmid JR et al. Mutations 45. Lu Q, Schafer DA, Adler PN. The Drosophila pla-
in  VANGL1 associated with neural-tube defects. nar polarity gene multiple wing hairs directly regu-
New Engl J Med. 2007;356:1432–1437. lates the actin cytoskeleton. Development. 2015;142:​
32. Yates LL, Schnatwinkel C, Murdoch JN et al. The 2478–2486.
PCP genes Celsr1 and Vangl2 are required for nor- 46. Mlodzik M. Planar cell polarization: Do the same
mal lung branching morphogenesis. Hum Mol Genet. mechanisms regulate Drosophila tissue polar-
2010;19:2251–2267. ity and vertebrate gastrulation? Trends Genet.
33. Lei YP, Zhang T, Li H, Wu BL, Jin L, Wang HY. 2002;18:564–571.
VANGL2 mutations in human cranial neural-tube 47. Mlodzik M. The Dishevelled protein family: Still
defects. N Engl J Med. 2010;362. rather a mystery after over 20 years of molecular
34. Yamada Y, Fuku N, Tanaka M et al. Identification of studies. Curr Top Dev Biol. 2016;117:75–91.
CELSR1 as a susceptibility gene for ischemic stroke 48. Clermont Y. Kinetics of spermatogenesis in mam-
in Japanese individuals by a genome-wide associa- mals: Seminiferous epithelium cycle and spermato-
tion study. Atherosclerosis. 2009;207:144–149. gonial renewal. Physiol Rev. 1972;52:198–235.
35. The HD IPSC Consortium. Induced pluripotent stem 49. Clermont Y, Morales C, Hermo L. Endocytic activi-
cells from patients with Huntington’s disease show ties of Sertoli cells in the rat. Ann NY Acad Sci.
CAG-repeat-expansion-associated phenotypes. Cell 1987;513:1–15.
Stem Cell. 2012;11:264–275. 50. Lie PPY, Mruk DD, Lee WM, Cheng CY. Cytoskeletal
36. Tissir F, Qu YB, Montcouquiol M et al. Lack of cad- dynamics and spermatogenesis. Philos Trans R Soc
herins Celsr2 and Celsr3 impairs ependymal cilio- Lond B Biol Sci. 2010;365:1581–1592.
genesis, leading to fatal hydrocephalus. Nat Neurosci. 51. Cheng CY, Mruk DD. Biochemistry of Sertoli
2010;13:700–707. cell/germ cell junctions, germ cell transport, and
37. Kathiresan S, Melander O, Guiducci C et al. Six spermiation in the seminiferous epithelium. In
new loci associated with blood low-density lipo- Griswold MD, ed. Sertoli Cell Biology, 2nd Edition.
protein cholesterol, high-density lipoprotein cho- Amsterdam: Elsevier; 2015:333–383. DOI: http://
lesterol or  triglycerides in humans. Nat Genet. dx.doi.org/10.1016/B978-0-12-417047-6.00012.0.
2008;40:189–​197. 52. Cheng CY, Mruk DD. Cell junction dynamics
38. Samani NJ, Braund PS, Erdmann J et al. The novel in the testis: Sertoli-germ cell interactions and
genetic variant predisposing to coronary artery dis- male contraceptive development. Physiol Rev.
ease in the region of the PSRC1 and CELSR2 genes 2002;82:825–874.
on chromosome 1 associates with serum cholesterol. 53. Cheng CY, Mruk DD. The blood-testis barrier and its
J Mol Med. 2008;86:1233–1241. implication in male contraception. Pharmacol Rev.
39. Liu CQ, Lin C, Whitaker DT et al. Prickle1 is 2012;64:16–64.
expressed in distinct cell populations of the central 54. Pelletier RM. The blood-testis barrier: The junc-
nervous system and contributes to neuronal mor- tional permeability, the proteins and the lipids. Prog
phogenesis. Hum Mol Genet. 2013;22:2234–2246. Histochem Cytochem. 2011;46:49–127.
40. Tao H, Manak JR, Sowers L et al. Mutations in prickle 55. Franca LR, Auharek SA, Hess RA, Dufour JM,
orthologs cause seizures in flies, mice, and humans. Hinton BT. Blood-tissue barriers: Morphofunctional
Am J Hum Genet. 2011;88:138–149. and immunological aspects of the blood-testis
41. Jones C, Qian D, Kim SM et al. Ankrd6 is a mam- and blood-epididymal barriers. Adv Exp Med Biol.
malian functional homolog of Drosophila planar cell 2012;763:237–259.
polarity gene diego and regulates coordinated cel- 56. Stanton PG. Regulation of the blood-testis barrier.
lular orientation in the mouse inner ear. Dev Biol. Semin Cell Dev Biol. 2016;59:166–173.
2014;395:62–72. 57. Li N, Tang EI, Cheng CY. Regulation of blood-­
42. Allache R, Wang MQ, De Marco P, Merello E, Capra V, testis barrier by actin binding proteins and protein
Kibar Z. Genetic Studies of ANKRD6 as a molecular kinases. Reproduction. 2016;151:R29–R41.
switch between Wnt signaling pathways in human neu- 58. Vogl AW, Vaid KS, Guttman JA. The Sertoli cell cyto-
ral tube defects. Birth Defects Res A. 2015;103:20–26. skeleton. Adv Exp Med Biol. 2008;636:186–211.
References 219

59. Cheng CY, Mruk DD. A local autocrine axis in 62. Babayeva S, Rocque B, Aoudjit L et al. Planar cell polar-
the testes that regulates spermatogenesis. Nat Rev ity pathway regulates nephrin endocytosis in develop-
Endocrinol. 2010;6:380–395. ing podocytes. J Biol Chem. 2013;288:24035–24048.
60. Yu A, Rual JF, Tamai K et al. Association of 63. Cheng CY, Mruk DD. An intracellular trafficking
Dishevelled with the clathrin AP-2 adaptor is pathway in the seminiferous epithelium regulating
required for Frizzled endocytosis and planar cell spermatogenesis: A biochemical and molecular per-
polarity signaling. Dev Cell. 2007;12:129–141. spective. Crit Rev Biochem Mol Biol. 2009;44:245–263.
61. Hagemann AI, Kurz J, Kauffeld S et al. In vivo anal- 64. Weber JE, Russell LD, Wong V, Peterson RN. Three
ysis of formation and endocytosis of the Wnt/b- dimensional reconstruction of a rat stage V Sertoli
catenin signaling complex in zebrafish embryos. cell: II. Morphometry of Sertoli-Sertoli and Sertoli-
J Cell Sci. 2014;127:3970–3982. germ cell relationships. Am J Anat. 1983;167:163–179.
Computational characterization and
integrative analysis of proteins involved
18
in spermatogenesis
PRANITHA JENARDHANAN, MANIVEL PANNEERSELVAM,
and PREMENDU P. MATHUR

INTRODUCTION in identification of suitable biomarkers and drug targets.


Increasing population and declining male fertility rates In this chapter, we discuss selected applications of these
are two different but corelated serious issues being faced techniques in identification of gene, miRNA, and protein
among humans. Lack of sex education and reproduction biomarkers for diagnosis and treatment of male infertility.
control measures remain causes for increasing population, Later we discuss the application of reproductive toxicants
while diverse genetic, environmental, and unidentified fac- and contraceptive-based models in identification of drug
tors also affect fertility rates. While reports forecast that targets that can be used for diagnosis and design of potent
the world will have a human population of about 10.1 bil- nonhormonal male contraceptives.
lion in 2050,1 other reports also state that both developed
and developing nations will face a decline in fertility rates ROLE OF OMICS AND BIOINFORMATICS METHODS IN
far below 2.1, the average number considered to withstand TRANSLATIONAL RESEARCH OF MALE REPRODUCTION
population size.2 Approximately 15% of couples worldwide Translational research in reproduction involves a com-
are observed to face infertility problems, with about half bination of multiple areas beginning with understand-
of the total count falling under male infertility factors.3–6 ing reproductive developmental biology, reproductive
Either for understanding the causes for male infertility tract biology, reproductive genetics, reproductive endo-
or for design of male contraceptives to achieve population crinology, and reproductive medicine. The output of all
control, it becomes necessary to understand the funda- these disciplines is used for understanding the molecular
mental process, spermatogenesis. Spermatogenesis is the mechanisms associated with successful reproduction and
complex, precisely coordinated, and regulated event that understanding the causes for its failure and for identifica-
results in the production of sperms. Success of spermato- tion of novel biomarkers for developing proper diagnosis
genesis lies in the proper regulation and coordination of and treatment methods. Genomics, transcriptomics, pro-
events that dictates (1) proper renewal of stem cells, pro- teomics, and bioinformatics methods are proven to pro-
liferation by mitosis, and differentiation, (2) generation vide successful understanding of reproductive disorders
of preleptotene spermatocytes from type B spermatogo- specifically for male infertility.12 Profiling of gene/miRNA/
nia, (3) transit of preleptotene spermatocytes across the protein expression profiling associated targeted inhibition
blood-testis barrier (BTB), (4) division of preleptotene to of proteins and understanding their structure and interac-
zygotene and diplotene spermatocytes that enters meiotic tion mechanism helps in high throughput screening, iden-
divisions I and II to form haploid spermatids, (5) contin- tification, and characterization of potential biomarkers.13
ued association of spermatids with Sertoli cells and their To understand the effect of mutations, copy number
maturation by spermiogenesis, and (6) precise removal of variation, single nucleotide polymorphisms, and altera-
mature spermatozoa from seminiferous epithelium into tion pattern of genes related to spermatogenesis, various
the lumen. Deregulation at any of these stages either by sequence-related methods like DNA microarray, next gen-
genetic or environmental factors affects the successful eration sequencing methods, miRNA-based microarray,
production of mature fertile sperms.7–11 real-time polymerase chain reaction (RT-PCR), and small
Besides understanding the molecular mechanism of RNA sequencing methods are employed. Proteomics
spermatogenesis through basic research, it is also of ben- methods use two-dimensional electrophoresis (2-DE),
efit to translate the findings for medical applications such ­differential-in-gel electrophoresis (DIGE), matrix-assisted
as identification of biomarkers for designing effective laser desorption mass spectrometry (MALDI-MS),
noninvasive methods for diagnosis of male infertility as surface-­enhanced desorption/ionization mass spectrom-
well as to identify drug targets for designing therapeu- etry (SELDI-MS), multidimensional protein identifica-
tic strategies to create nonhormonal contraceptives. The tion technology (MUDPIT), and liquid chromatography
combined use of genomics, transcriptomics, metabolo- and mass spectrometry (LC–MS).14 The net result of these
mics, and bioinformatics methods has been significantly techniques provides the list of genes and proteins that are
beneficial for understanding the basic mechanisms as well differentially regulated in the given condition.

220
Genomic and transcriptomic biomarkers  221

With the identification of differentially expressed genes/ can be used as biomarkers for diagnosis and treatment of
proteins, it becomes necessary to understand their bio- male infertility.
logical significance. Application of bioinformatics finds its
place in this step, where several integrated bioinformatics GENOMIC AND TRANSCRIPTOMIC BIOMARKERS
tools are employed. The basic understanding of biologi- Lin et al. first characterized testis-specific genes in humans
cal significance aims at understanding different aspects and identified Septin 4, zinc metallopeptidase, Ca+/
of gene products such as their cellular location, func- calmodulin protein kinase IV (CaMKIV) and calspermin
tional classification, structural classification, and analy- (CAS) as some of the important genes found to be specific
sis of their domain, structures, relationship to biological to the testis and play an important role in spermatogen-
pathways, and diseases. Several integrated ­bioinformatics esis.25 Later, Feig et al. obtained testis from normal and
tools like the Database for Annotation, Visualization and azoospermic patients and identified testis-specific genes
Integrated Discovery (DAVID), gene ontology, and Kyoto in humans. Results identified about 348 genes in premei-
Encyclopedia of Genes and Genomes (KEGG) are being otic, 81 genes in postmeiotic, and 38 genes in termination
used for this purpose. DAVID tool performs gene-list- phase in testis.26 Liu et al. identified testis-specific genes
based annotations15,16 that takes gene list as the input in humans, mouse, and rat and with their findings cre-
and identifies data related to gene ontology17 and meta- ated a precise testis-specific gene database. In their study,
bolic pathway analysis from KEGG.18 With identifica- authors employed digital differential analysis where they
tion of protein targets it becomes essential to understand screened Unigene libraries, retrieved testis-specifc genes,
their interaction partners, which can be achieved using and used RT-PCR studies to validate their findings. They
web-based protein-protein interaction databases such found about 317, 449, and 147 testis-specific genes in
as String.19 Later, structural analysis of predicted targets human, mouse, and rat, respectively.27
is achieved by three-dimensional structure prediction of In addition to approaches to identify testis- and sperm-
proteins by the MODELLER suite20 and the Rosetta suite.21 specific genes from databases, several other studies have
With analysis of individual protein structures it becomes been involved in analyzing genes/miRNA that are dif-
essential to understand how selected protein interacts ferentially expressed in different conditions pertaining
with its targets using protein-protein docking software to male infertility. Malcher et al.28 attempted to charac-
such as HADDOCK 22 and protein-ligand docking such as terize the genetic causes of male infertility and in their
Schrodinger Glide.23 Finally, the conformational changes study compared the gene expression profiles between
induced by complex formation are evaluated using molec- normal and oligozoospermic patients. Results identi-
ular dynamics simulation studies that use software such as fied differential expression of about 4946 genes, wherein
the Gromacs suite.24 genes A-kinase anchoring protein (AKAP4), ubiquilin 3
With the application of diverged protein sequence (UBQLN3), calpain 11 (CAPN11), gametogenetin (GGN),
and structure-based analysis tools, the overall function sperm acrosome associated 4 (SPACA4), apermatogen-
of the identified targets can be postulated. Finally, based esis associated 3 (SPATA3), and family with sequence
on the significance of the role played by the identified similarity 71, member F1 (FAM71F1) were the most sig-
targets, the  potent biomarkers and drug targets can be nificantly downregulated in infertile patients. Expression
identified. analysis revealed that AKAP4 is expressed in spermato-
zoa29 UBQLN3, CAPN11, and SPATA3 were expressed in
APPLICATION OF GENOMIC, TRANSCRIPTOMIC, spermatocyte,30,31 and GGN was expressed in early round
AND PROTEOMIC APPROACHES IN IDENTIFICATION spermatids.32 Molecular characterization of all these pro-
OF POTENT BIOMARKERS teins shows that their knockout impairs spermatogenesis,
The conventional techniques of diagnosing male infertil- thus favoring their selection as potential biomarkers for
ity depends on semen analysis, which can be segregated diagnosing male infertility.
into macroscopic and microscopic factors that include In addition to the genomic studies, miRNA profiling
coagulation, color, viscosity, and agglutination, sperm has also helped in identifying the potential biomarkers
count, and concentration, respectively. Indeed the analy- for diagnosis of male infertility. Abu-Halima et al.33 com-
sis of semen suffers from serious challenges. These factors pared microRNA expression profiles in normozoosper-
potentiate the need for identification of novel biomarkers mic, asthenozoospermic, and oligoasthenozoospermic
that can pave the way for easy identification and diag- donors. Nearly 50 miRNAs were upregualted in astheno-
nosis of diseases. Genes, miRNAs, and proteins are such zoospermic males and 42 miRNAs were upregulated in
potential biomarkers, the presence of which can be easily oligoasthenozoospermic men in comparison with normal
identified, further promoting their selection as potent bio- men. In a similar study,34 the same authors compared the
markers. In addition, with an understanding of their func- miRNA expression profile in normal men with infer-
tion and structure, potent therapeutic strategies can also tile men characterized with Sertoli-cell-only (SCO) syn-
be designed. The following sections discuss the application drome, mixed atrophy (MA), and germ cell arrest (GA)
of genomics, transcriptomics, and proteomics in identifi- phenotypes and 197, 68, and 46 miRNAs were observed to
cation of potent biomarkers and a list of a few proteins that have differential expression in these groups, respectively.
222  Computational characterization and integrative analysis of proteins involved in spermatogenesis

Both these studies identified miR-141, miR-200a, miR- characterizing the proteomic fingerprint of testes. The tes-
122, miR-34b, miR-34c-5p, and miR-16, and hsa-mir-34b*, tis is also unique in that it has a high level of alternative
hsa-mir-34b, hsa-mir-34c-5p, hsa-mir-449a, and hsa-mir- splicing at the transcriptional level, resulting in heteroge-
449b as potential miRNA-based biomarkers for diagnosis neous proteome. This was analyzed by Guo et al., where
of male infertility. miR-122 is involved in posttranscrip- the authors employed two-dimensional gel electrophore-
tional downregulation of transition protein 2 (TNP2).35 sis to analyze testis-specific proteins, alternative isoforms
miR-34b and miR-34c regulate Notch gene homologue 1 of testis-enriched proteins, and those that exhibit unique
(NOTCH1) that is found to regulate survival and differen- expression between normozoospermic and azoospermic
tiation of germ cells.36 miR-449a in turn regulates BCL-2 patients.41 They identified 462 unique proteins and charac-
and miR-26a regulates the function of estrogen receptors. terized 52 proteins that showed heterogeneous forms. They
These miRNAs and their target mRNAs were validated also found 27 proteins that expressed differential expres-
by RT-PCR studies and are shown to be the effective bio- sion between normal and infertile donors. As well, these
markers for the diagnosis of male infertility. results were embedded and developed into a testis-specific
The combined expression profiling of both miRNA and proteome. Following this study, numerous authors have
their target mRNAs in normal versus impaired sperm been involved in characterizing the testis proteome under
donors was performed by Zhuang et al. In their study,37 different conditions. Recently, Kathrein et al. character-
they collected the testes from normal, obstructive azo- ized the expression of proteins in testis in the presence of
ospermia, and nonobstructive azoospermic patients. spermatogonia and compared them to testis with an SCO
The differential expression of the miRNAs and mRNAs data set.42 SCO appearance is the characteristic form of
obtained was studied and results found that about 93 male factor infertility and the results of this study identi-
miRNAs and 4172 mRNAs were differentially expressed fied 239 specific genes that showed differential expression.
in obstructive azoospermia and nonobstructive azoosper- The results postulated that the proteins fibroblast growth
mic patients. Functional classification of these miRNA/ factor receptor 3 (FGFR3) and desmoglein 2 (DSG2) are
mRNA pairs revealed that they are highly associated with specific biomarkers of spermatogonia.
spermatogenesis, cell meiosis, cell cycle, and development In another study, Djureinovic et al. characterized the
of secondary sexual characteristics. Interestingly, identi- testis-specific proteome based on transcriptomics and
fied miRNAs were found to target multiple genes such as antibody-based profiling and identified more than 1000
hsa-miR-199a-5p that targets spermatid-associated pro- gene products that were testis-specific and further charac-
tein (SPERT), SPACA4, actin-like protein 7A (ACTL7A), terized them based on their location, such as in spermato-
testis-specific serine kinase 6 (TSSK6), and UBQLN3. Of gonia, spermatocytes, spermatids, sperm, Sertoli cells, and
these, the functional role of TSSK6 and UBQLN3 has been Leydig cells.43 The results reveal that doublesex- and mab-
well studied. TSSK6 belongs to the testis-specific serine/ 3-related transcription factor 1 (DMRT1) and P antigen
threonine kinase family, and the downregulation asso- family member 1 (PAGE1) are specific to spermatogonia.
ciated with TSSK6 is observed to result in production of DMRT1 protein is involved in male sex determination and
abnormal sperm morphology and is also associated with differentiation,44 while the function of PAGE1 is unknown.
affecting sperm-egg fusion.38,39 UBQLN3, on the other Similarly, the deleted-in-azoospermia-like (DAZL) gene
hand, belongs to the family of ubiquitin-like proteins, and product was reported to show high enrichment in sper-
is a testis-specific gene essential for sperm function.40 matocytes, while the transition protein 1 (TNP1) gene
and sperm mitochondria-associated cysteine-rich protein
PROTEOMIC MARKERS AND DRUG TARGETS (SMCP) and actin-like protein 7B (ACTL7B) were specific
The systematic analysis of protein expression and charac- to spermatid. Meanwhile, defensin, beta 119 (DEFB119)
terization in a given tissue/cell under a particular condition was found to be specific to Sertoli cell, the A kinase (PRKA)
or at a given time, called proteomics, plays a crucial role in anchor protein 4 (AKAP4) was specific to Sertoli cell and
understanding the expression of the pattern of vital pro- glyceraldehyde-3-phosphate dehydrogenase (GAPDHS)
teins that function as cohort to ensure progression of spe- gene products are specific to sperm. These findings provide
cific functions. Proteomic approaches are highly beneficial useful insights into understanding the proteomic signature
in characterizing the protein signature of germ cells specific of testis.
to each stage of spermatogenesis and in comparison to their Like Djureinovic et al., several other authors have also
expression between fertile and infertile patients. They also characterized the proteomic signature in sperm-milieu,45,46
prove vital in identifying a specific set of proteins during seminal plasma,47 and sperm tail.48 However, over time,
the exposure of testis/Sertoli cells/sperm to a wide range of applications of proteomics has drifted toward identifica-
environmental factors. The results of all these approaches tion of biomarkers that exhibit differential expression
are crucial to understanding the underlying mechanisms between normal and infertile patients. Hashemitabar et al.
of fertile sperm production and to understanding the compared the expression of proteins in sperm tail between
molecular basis for development of male infertility. normozoospermic and asthenozoospermic donors.48
The testis is a unique somatic counterpart that pos- Asthenozoospermia, a common form of male infertility,
sesses endocrine and exocrine factors that regulate sper- is characterized by reduced sperm motility, and the pro-
matogenesis and proteomics has played a crucial role in teomic study conducted by Hashemitabar et al. identified
Models based on exogenous compounds to identify potential biomarkers in spermatogenesis  223

14 unique proteins that exhibited adistinct expression They observed decreased expression of tektin, myosin,
­pattern.48 Of the total 14 proteins, 11 proteins [A-kinase and septin, which are involved in ­acrosome formation
anchor protein 4 (AKAP4), Tubulin beta 2B (TUBB2B); and sperm motility, and therefore, their low abundance in
outer dense fiber protein 2 (ODF2), cytochrome c oxidase obesity-associated asthenozoospermic patients renders the
subunit 6B (COX6B) phospholipid hydroperoxide gluta- sperm immotile.55 Low ­expression of proteins involved in
thione peroxidasemitochondrial (PHGPx), glutathione glucose metabolism, such as ­fructose-bisphosphatealdolase,
S-transferase Mu 3 (GSTMu3);glyceraldehyde-3-phosphate glyceraldehyde-3-­phosphate dehydrogenase, aldose reduc-
dehydrogenase, testis-specific (GAPDS), voltage-depen- tase, glyoxylatereductase­, cytochrome c oxidase, and pyru-
dent anionselective channel protein 2 (VDAC2), heat vate dehydrogenase, were also observed, which directly
shock-related 70 kDa protein 2 (HSPA2); stress-70 protein, implies that their low expression affects energy metabo-
mitochondrial (HSPA9), and sperm protein associated lism in sperms. As well, for the first time they reported
with the nucleus on the X chromosome B (SPANXB)] are the association with the decreased expression of two novel
specific to normal sperm tails. proteins ERp57 and actin-related protein ACTRT2 with
On the other hand, proteins such as keratin, type II the progression of obesity-associated asthenozoospermia,
cytoskeletal 1 (KRT1), isoaspartyl peptidase/lasparaginase which they proposed as the potential biomarkers for diag-
(ASRGL1), and clusterin (CLU) were enriched in astheno- nosis and therapeutic intervention.
zoospermia data set. Besides the ASRGL1 and SPANXB Proteomics study has also been involved in analyzing
proteins, the remaining 12 proteins were assigned to five the impact of environmental factors in impairing repro-
functional classes: (1) sperm movement and structural ductive abilities in humans. In a study conducted by Li,56
organization (TUBB2B, ODF2, AKAP4, KRT1, and CLU); the authors analyzed the proteomic pattern in patients
(2) energy and metabolism (COX6B, GAPDS, PHGPx), subjected to ionizing radiation, such as cosmic radiation,
(3) stress response and turnover (HSPA2, HSPA9), (4) sig- diagnostic X-rays, and radiotherapy for cancer. More pre-
naling and transportation (VDAC2), and (5) antioxidant cisely, cancer patients receiving radiotherapy are often
activity (GSTMu3). reported to have infertility problems,57 implying the need
Among the proteins enriched in sperm tails from nor- for a detailed investigation on the effect of radiation on
mozoospermic donors, TUBB2B and ODF2, which showed male fertility. With the help of proteomic approaches,
decreased expression in asthenozoospermic sperm tails, Li  et  al.56 have identified profound diminution in the
were found to be crucial for sperm tail motility. Tubulin expression pattern of Hsp70-2, phospholipases C, PHGPx,
beta 2B is a component of falagellar microtubule49 and β-tubulin, and GAPDHS. All these proteins are also
outer dense fiber protein 2 forms the component of outer observed to have decreased expression in the sperm tails
dense fibers, which are the accessory structures found sur- of asthenozoospermic patients, suggesting that these pro-
rounding the axenome of sperm tail and are involved in teins might act as potential biomarkers for diagnosis and
regulating elastic rigidity of sperm flagellum.50,51 Reduced therapeutic strategies. Huang et al. have also characterized
expression of these proteins is obvious to impart nega- the proteomic signature in response to exposure of arsenic
tive effects in sperm tail movement in asthenozoospermic in humans.58 In their study they identified that the pro-
patients. Similarly, A- AKAP4 is the abundant struc- teins, voltage-dependent anion-selective channel protein
tural protein in fibrous sheath that anchors cyclic-AMP- 3, heat shock 70 kDa protein 4L, microtubule-associated
(cAMP)-dependent protein kinase A (PKA) to the fibrous protein 1B, glutathione peroxidase 4, and Y-box-binding
sheath, therein increasing phosphorylation activities lead- protein 3 are significantly downregulated, and they
ing to activation of fibrous sheath. Deletion of AKAP4 is reported the involvement of ERK/AKT/NF-κB signaling
shown to inactivate fibrous sheath and successively impair pathway in arsenic-induced male reprotoxicity.58
sperm tail motility.52,53 GAPDS is another important pro- In general, a diverse set of proteomic studies have been
tein that is downregulated in asthenozoospermic patients. carried out recently to assess the protemic signatures of
GAPDS is located along the principal piece of sperm tail infertile and fertile men under diverse conditions. These
and plays a role in ATP production in the distant site of studies are continuously updating several proteins as
flagellum. Gene knockout studies proved that a GAPDS- biomarkers for diagnosis and therapeutic strategies,
deficient mouse is deficient in sperm tail movement.54 and detailed experimental validations are anticipated to
Similarly, the proteins COX6B, PHGPx, VDAC2, and unravel the potential of these biomarkers.
HSPA2 and HSPA9 also can be used as biomarkers for
understanding male infertility. MODELS BASED ON EXOGENOUS COMPOUNDS
In another study, 55 Liu et al. analyzed the proteome TO IDENTIFY POTENTIAL BIOMARKERS
expression pattern in patients affected with obesity. The IN SPERMATOGENESIS
authors found that 105 proteins shown less abundance Although genomics, transcriptomics, and proteomics
while 22 were enriched in obesity-associated asthenozoo­ methods are used to profile an expression pattern of genes,
spermic patients. They further analyzed the functionality miRNA, and proteins in different conditions pertaining to
of these proteins and found them involved in actin orga- male infertility, the resulting number of targets still needs
nization, flagellar assembly, vesicule traffic, signaling, experimental validation, thus delaying their selection as
metabolism, protein degradation, and stress resistance. biomarkers. On other hand, the study of spermatogenesis
224  Computational characterization and integrative analysis of proteins involved in spermatogenesis

events in the presence of foreign substances such as toxi- of BPA on the steady state level of Cx43, and using BPA as a
cants and chemical substances such as contraceptives has model, they show that Cx43 is crucial for maintaining the
proven to be a valid approach for identifying potential tar- integrity of BPA.66 In their studies, treatment of BPA has
gets. Spermatogenesis, the coordinated series of precisely decreased the steady state level of Cx43 and other BTB-
regulated events that takes place in the microenviron- associated proteins such as occludin, ZO-1,N-cadherin,
ment of the testis, is organized into four phases involving β-catenin, and α-catenin. However, after 24 hours of BPA
(1) spermatogoniogenesis, (2) spermatocyte differentiation, removal, these junction proteins, except for Cx43, relocal-
(3) spermiogenesis, and (4) spermiation. These processes ized toward the cell-cell surface, but the level of Cx43 was
take place in the seminiferous epithelium of the testis, maintained at a low level, while knockdown of Cx43 was
where the basal and adluminal compartments are por- shown to affect the relocalization of these proteins. Studies
tioned by the presence of the testis-specific BTB, which show that ERK1/2 signaling induced by BPA could play a
serves as an immunological barrier and is involved in the crucial role in BTB disruption but in the junction reas-
transport of preleptotene spermatocytes into the adlumi- sembly. Other studies also show that BPA can effectively
nal compartment where it further differentiates to form decrease the level of Cx43 and therefore it can affect the
zygotene and diplotene spermatocytes that follow two localization of N-cadherin and ZO1 in BTB and reduce
meiotic divisions to form haploid spermatids. Haploid the sperm count.67 Together, these studies indicate that
spermatids then enter into a spermiogenesis event where Cx43 is the potential cellular target of BPA although it can
they mature into round and elongated spermatids, which downregulate the expression of other BTB proteins.
throughout their maturation process remain attached with In another study, Xio et al. elucidated that BPA can affect
somatic nurse cells called Sertoli cells. The cytoskeleton F-actin organizations and can impair Sertoli cell adhesive
network of Sertoli cells constituted by actin and micro- functions.68 In this study, BPA exposure altered the local-
tubules remains crucial for attachment and transport of ization of actin regulatory proteins such as Arp3, Eps8,
spermatids across the adluminal compartment toward the c-SRC, and annexin-II. Actin-related protein 3 (Arp3)
lumen where the matured elongated spermatids get trans- and epidermal growth factor receptor substrate 8 (Eps8)
ferred into lumen as sperms. are two critical proteins that are expressed differentially
The precise restructuring of a BTB, apical ES, and at BTB near the basal ES and are shown to regulate the
regulation of Sertoli cells, actin- and microtubule-based organization of actin microfilaments. Studies show that
network is essential for successful termination of sper- Eps8 is essential in bundling of actin microfilaments while
matogenesis. Various studies have established that BTB,59 Arp3 induces the nucleation of branched actin filaments.69
apical ES,60 actin- and microtubule-based network,61,62 and Changes from bundled to branched architecture is pivotal
Sertoli cells63 serve as the potent targets of diverse repro- for restructuring the BTB. At the given concentrations
ductive toxicants and contraceptives. Extensive studies BPA has altered the localization of these proteins and has
using these exogenous compounds as models has identi- resulted in the conversion of bundled actin filaments to
fied various potent biomarkers that can be used for diag- branched structures. Further, this structural change is
nosis of male infertility as well as in the design of potent associated with impairing the association of Jam-ZO1
nonhormonal male contraceptives. and N-cadherin-β-catenin complexes with actin bundles,
which results in BTB disruption at undesirable stages of
BPA-BASED MODEL FOR IDENTIFICATION OF CRUCIAL the epithelial cycle.
DRUG TARGETS BPA is also shown to induce apoptosis in Sertoli cells
In a study model based on BPA, researchers have identi- through varied mechanisms. Qian et al. has shown the
fied several crucial proteins that can be used as potential involvement of CaM-CaMKII-ERK1/2 in mediating the
biomarkers. BPA, bisphenol A, [2,2-bis(4-hydroxyphenol) Sertoli cell apoptosis.70 Here, the authors show that BPA
propane] (BPA), is a ubiquitously occurring endocrine dis- increased the expression of CaM and subsequently acti-
ruptor, and it is characterized as a Category 3 reproductive vated the phosphorylation of CamKII and on the other
toxicant by the European Chemicals Bureau. (A  Class 3 hand BPA affected the Ca2+–ATPase activity and increased
reproductive toxicant causes irregularities affecting human the Ca2+ concentration in Sertoli cells. In general, CaM
fertility or results in developmental defects as evidenced by is shown to mediate the Ca2+ mediated mitochondrial
animal studies.)64 damage, where its activation leads to its association with
The studies of Li et al. show that BPA disrupts BTB by CaMKII, which drives the opening of the mitochondrial
declining the steady state level of N-cadherin, occludin, permeability transition pores.71 The opening of the mito-
and connexin 43 (Cx43).65 N-Cadherin forms the ecto­ chondrial permeability transition pores substantially
plasmic specialization protein found at apical and basal increases the permeability in the inner mitochondrial
ES, while occludin is a part of the occluding-ZO-1 adhe- membrane leading to mitochondrial depolarization, ATP
sion complex found at tight junctions and Cx43 is a part depletion, mitochondrial swelling, and subsequent release
of the gap junction proteins. A decrease in the steady state of cytochrome C to cytosol. Together with an increase in
level of these proteins substantially affects the BTB integ- cytosolic cytochrome C, BPA also increased the expres-
rity that in turn affects the normal progression of sper- sion of caspases and downregulated expression of Bcl-2
matogenesis. In a similar study, Li et al. discussed the effect antiapoptotic proteins leading to Sertoli cell apoptosis.
Male contraceptive-based model  225

BPA is also shown to decrease the serum testosterone nucleation complex Arp2/3. 86 By downregulating the
level in rats exposed to BPA from postnatal days 21 to 35, expression of proteins adjudin typically affects the apical
where a decrease in the testosterone level, accompanied ES restructuring, leading to the release of immature sper-
with upregulation of Fas and FasL, activated Fas/FasL sig- matids into the lumen. Adjudin also affects the dynamics
naling and upregualted the cytochrome C and Bax pro- of the Sertoli cell microtubule network by downregulating
teins leading to Sertoli cells apoptosis.72 the expression of microtubule affinity regulating kinase 4
In another study, D’Cruz et al. showed that BPA (MARK4).87 MARK4 is a typical member of the MARK
decreased the activity of antioxidant enzymes such as family that shows direct binding with microtubules.88,89
superoxide dismutase, catalase, as well as it affected the Adjudin treatment is shown to delocalize MARK4 from
activities of glycolytic enzymes such as hexokinase, phos- the concave side of the spermatid head, affecting sperm
phofructokinase, insulin receptor subtype-2 (IRS-2), and polarity and resulting in premature release of immature
glucose transporter 8 (GLUT-8) and increased the level spermatid into the lumen.87 The adjudin model has also
of insulin. In doing so, BPA effectively induced oxidative highlighted the function of the Fascin 1 protein.90 The
stress, impaired glucose homeostasis and insulin signal- Fascin protein (Fascin 1-3) is involved in binding with
ing, and affected steriodogenesis in rat testis.73 In a simi- and bundling actin filaments by cross-linking acting fila-
lar study, D’Cruz et al. reported that exposure of BPA also ments to form tight bundles.91 Fascin 1 is highly expressed
decreased the level of IRS-1 and GLUT-2 and impaired in Sertoli cells and in germ cells. Its localization has been
the activities of enzymes involved in steroidogenesis such observed up to stage VIII near the basal ES at the BTB, in
as 17β-hydroxysteroid dehydrogenase (17β-HSD) and stages VII to late-stage VIII in apical ES, and is restricted
3-β-hydroxysteroid dehydrogenase (3β-HSD), and down- to stage 19 spermatids of the seminiferous epithelium.
regulated steroidogenic acute regulatory protein (StAR) Adjudin-based models highlight that Fascin 1 is crucial
protein and affected testosterone production.74 for conferring polarity to the sperm head of spermatids
Together these studies identify connexin Cx43, occludin, adhered to apical ES and its targeted disruption affects
ZO-1,N-cadherin, β-catenin, Arp3, Eps8, CaM-CaMKII- sperm polarity and results in premature sperm release into
ERK1/2 signaling proteins, Fas and FasL, superoxide the lumen.90
dismuatase, catalase, hexokinase, phosphofructokinase, Although adjudin treatment provides useful insights
IRS-2, GLUT-8, 17β-HSD, 3β-HSD, and StAR as the key into the underlying mechanisms of BTB and apical ES
targets for diagnosis of male infertility and for develop- assembly/disassembly, the efficacy of adjudin remains very
ment of male contraceptives. low due its limited availability.59,92 Studies using an adju-
din model were therefore designed to identify factors that
CADMIUM-BASED MODEL SYSTEMS affect bioavailability and results show that P-glycoprotein
(a member of the multidrug resistance gene MDR1),93
Cadmium is a well-studied heavy metal that is shown
multidrug resistance-related protein 1(MRP1), a member
to impart reproductive toxicity. Studies elucidate that
of the multidrug resistance-related protein family,93–95
cadmium can effectively affect BTB functions in differ-
and breast cancer related protein ([BCRP], ATP-binding
ent ways and can act on the Sertoli cell cytoskeleton.68,75
cassette, subfamily G, member 2)96 are three crucial drug
Cadmium is shown to perturb tight junction permeability
transporters located in the BTB that are involved in drug
by selectively targeting occludin and it is shown to acti-
efflux across the BTB. Adjudin-based studies show that a
vate JNK and p38 MAPK pathways.75–77 It is also shown to
steady state level of P-glycoprotein was increased after the
downregulate the expression of p-FAK-Tyr576 and p-FAK-
exposure of adjudin, which returns to their normal level
Tyr397, and therefore cadmium destabilizes apical ES
following the elimination of adjudin.93 Results highlight
integrity as well as the functions of BTB.78
that P-glycoprotein is actively involved in elimination of
adjudin, and that coimmunoprecipitation revealed that
MALE CONTRACEPTIVE-BASED MODEL P-glycoprotein associates with occludin, Claudin-11, and
Adjudin 1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide JAM-A of the BTB and aids in further regulation of BTB
is a well-studied nonhormonal male contraceptive designed integrity therein to prevent entry of adjudin into the adlu-
and patented by the C. Yan Cheng lab.79 In their experi- minal compartment to prevent the contraceptive actions
ments, scientists from this lab used this compound to of adjudin.93 In another study by Qian et al., the authors
identify various protein targets that can be exploited for showed that BCRP is expressed by both Sertoli cells and
the use of potent nonhormonal male contraceptives.80,81 germ cells although it is not located in BTB but is observed
Their studies highlight that adjudin more prominently in apical ES. Expression of BCRP was observed in Sertoli
exerts its effects in the proteins localized in the api- step 18–19 spermatid interface in apical ES and is limited
cal ectoplasmic specialization (ES).82–84 Apical ES is the to stages VI to early VIII tubules, implying that matur-
testis-­specific cell junction constituted by adherens junc- ing spermatids utilize the potential of BCRP to safeguard
tions, focal contact, and tight junction proteins.85 Adjudin themselves from the harmful effects of drugs.96
is observed to disrupt F-actin organization at apical ES, Adjudin models also highlight the function of three
downregulate the expression of actin bundling/barbed members of the Src family kinases (c-Src, c-yes, and FAK)
end capping protein Eps8, palladin, and barbed  end in spermatogenesis.97–99 The protein c-Src is expressed in
226  Computational characterization and integrative analysis of proteins involved in spermatogenesis

both Sertoli cells and germ cells, and is upregualted at api- phosphorylate occludin and are shown to regulate the bio-
cal ES, conferring its role in restructuring apical ES for the availability of adjudin, understanding how these kinases
release of mature spermatozoa into the lumen.100 Adjudin interact with both occludin and with adjudin gains func-
models have confirmed this function of c-Src, particularly tional importance. Results of their study highlighted
of its phosphorylated form p-c-Src-Tyr419.101–103 Protein the presence of important interactions that mediate the
c-Src is shown to physically associate with N-cadherin/β- association of c-Src, c-Yes, and FAK with occludin and
catenin, β1-integrin, which is a component of the α6β1- also of adjudin with c-Src, c-Yes, and FAK. These results
integrin-lamnin-α3β3γ3 complex.104 Adjudin treatment also showed that adjudin can bind with the hinge region
is shown to enhance the phsophoryaltion activity of c-Src between the two lobes of kinase in a similar way as with
that is in turn shown to phosphorylate the cellular targets reported drugs and can alter the activity of these pro-
of c-Src, leading to dismantling of apical ES components teins, thus explaining how this association can regulate its
favoring release of immature spermatids into the lumen.104 bioavailability.
Activity of c-Src is also observed at the BTB, where it colo- In another study, Su et al. detailed how adjudin binds
calizes with N-cadherin/β-catenin, and also binds and with P-glycoprotein.111 P-glycoprotein is potentially
phosphorylates occludin, hampering the binding of occlu- involved in altering the bioavailability and hence in this
din with Zoa-1 that is shown to affect the tight junction study, by utilizing homology modeling and docking stud-
(TJ) barrier in the BTB.105 ies, the authors predicted the structure for inward facing
As well, c-Yes, another member of the SRC family of conformation of P-glycoprotein and analyzed the binding
kinase, is also a crucial target of adjudin that shows abun- association of adjudin. The results suggest designing com-
dant expression in the BTB.106 It is shown to associate with pounds with better structural components that can help
N-cadherin and occludin and is also shown to be involved them traverse across the BTB for targeting proteins in the
in restructuring of the BTB.106–108 Similarly, FAK is also adluminal compartment.
involved in phosphorylation, a component of the BTB, and Similar to P-glycoprotein, BCRP has also been shown
apical ES. Specifically, studies show that p-FAK-Tyr407, to affect bioavailability of adjudin, such that under-
-Tyr397, and -Tyr576 are highly expressed in the BTB and standing how adjudin binds with BCRP can also help in
also in apical ES,109 but are predominantly expressed in the designing potent contraceptives. In view of this, Qian et
BTB. In the BTB they are involved in disrupting the Sertoli al. and Cheng et al., in independent studies, predicted the
cell TJ permeability by regulating the phosphorylation inward and outward facing conformations of BCRP as
status of occludin, while in apical ES it is involved in the well as tetrameric arrangement of BCRP.112,113 BCRP, like
restructuring of F-actin filaments, thus implying that FAK is P-glycoprotein, has N-terminal DNA binding domain fol-
also involved in regulating the disassembly of both the BTB lowed by a transmembrane region at its C-terminal NH2-
and apical ES.98 Results indicate that the phosphorylation NBD-TM-COOH. BCRP adopts a characteristic V-shaped
function of these proteins are also involved in deciding the conformation at its inward-facing conformation and
bioavailablity of adjudin, and therefore understanding their adopts an X-shaped conformation at its outward confor-
function is crucial for developing better contraceptives. mation. The switch between the conformations helps in
Together, the results of these models highlight the poten- traversing or repelling out the molecules away from the
tial of several proteins in the BTB, apical ES, and Sertoli BTB barrier. The results show that adjudin forms strong
cells as crucial drug targets whose deregulation can help hydrogen bonds, pi interactions, electrostatic interactions,
impaired spermatogenesis and can also be used to design and van der Waals interactions with residues of BCRP.
drugs for developing nonhormonal male contraceptives. Interestingly, it switches between the interaction with cat-
alytic R465 and R482 in accordance with inward and out-
ROLE OF BIOINFORMATICS IN UNDERSTANDING ward conformations, and in both cases, 3-carbohydrazide
THE STRUCTURE AND FUNCTION OF BIOMARKERS moiety of adjudin helps in anchoring adjudin with BCRP,
IDENTIFIED USING OMICS APPROACHES and thus a suitable replacement of this moiety can help
With the identification of potential protein biomarkers, it adjudin to escape these interactions for traversing into the
becomes essential to understand their structure and inter- adluminal compartment.
action mechanism with their cellular partners in order to In addition to the studies aimed at improving efficacy of
design strategies to inhibit its biological function. Besides adjudin, other studies were also conducted to identify new
targeted inhibition, understanding the structural mecha- promising leads that can be used as male contraceptives.
nism underlying the functional activities carried out by In a study conducted by Jenardhanan et al., the authors
the selected proteins also becomes essential to explain analyzed the structure of MARK4,114 which is shown to be
the observed biological phenotypes that result upon their important in conferring polarity to maturing spermatids
inhibition in in vitro and in vivo studies. that are attached with apical ES and targeted inhibition of
In the study conducted by Xiao et al, the authors have MARK4 is shown to impair spermatogenesis, resulting in
attempted to predict the structural basis of the molecu- premature release of immature spermatid into the lumen.
lar association of occludin with c-Src, c-Yes, and FAK.110 In this study, the authors predicted the three-dimensional
Since these Src kinases are observed to bind with and conformation for the kinase-UBA construct of MARK4
Conclusion and future perspectives  227

and identified MARK4-specific inhibitors.114 MARK4 structural mechanism behind the biological function of
belongs to the serine/threonine kinases and is the mam- selected biomarkers.
malian homolog of partition-defective 1 protein. MARK4 See Table 18.1 for more details.
has four important regions: N-terminal kinase domain,
autoregulatory UBA domain, disordered spacer region,
and a terminal kinase-associated domain. As a means of CONCLUSION AND FUTURE PERSPECTIVES
identifying novel inhibitors for MARK4, the authors have With an increasing population and declining male infertil-
predicted the structure for the kinase–UBA construct ity rate, the need for designing potent techniques to achieve
in an inactive catalytic conformation and revealed that efficient diagnosis and treatment strategies also increases.
MARK4 has unique structural features such as DFG-in- Although the severity of the problems increase the poten-
αC-helix-out inactive conformation that differentiates tial of diagnosis and treatment, strategies still suffer
it from other ser/thr kinases. The presence of a unique from various unknown factors. The potency of advanced
DFG-in-αC-helix-out mode necessitates the design of genomic, proteomic, and computational techniques should
MARK-specific inhibitors. In doing this, the authors uti- be integrated for translating the understanding of funda-
lized the pharmocophoric feature of 9-oxo derivatives that mental mechanisms of spermatogenesis toward identify-
are shown to be MARK-specific and used them to screen ing targets that mark the presence of unusual conditions
a library of kinase inhibitors. The results identified 10 such as male infertility. Further, these identified targets
potent leads that on molecular dynamics simulation study should be evaluated for their potency to act as drug targets
maintained their binding association and showed steady for designing effective treatment strategies. This chapter
pulling of N- and C-terminal lobes of kinase domain, discussed the application of genomics, proteomics, and
thereby arresting the catalytic conformation from phos- bioinformatics methods in translating the basic under-
phorylating its substrates. The identified leads function standing of spermatogenesis toward biomarker and drug
as potent ATP competitors such that with unique DFG-in target identification. Various studies in these areas should
conformation they can be used to inhibit both active and be designed in the future by integrating these techniques,
inactive states of MARK4, thus favoring their selection for and several targets should be identified in the near future
the design of next generation nonhormonal contracep- so that diagnosis and treatment strategies can be rein-
tives. There are few studies that illustrate how structural vented for overcoming the problems of male infertility and
bioinformatics can be used to understand the underlying increasing population growth.

Table 18.1  Details of biomarkers and drug targets discussed.


Protein
GO NCBI ID
S.no Target identified Localization class GeneID uniprot PDB Id
Genomics Study
1. Septin 4 (SEPT4) Sperm Sperm Mitochondria 5414 O43236 Not available
organization
2. Zinc metallopeptidase Testis Metalloendopeptidase 10269 O75844 2YPT, 4AW6
(ZMPSTE24)
3. Ca+/calmodulin protein Testis Calmodulin-dependent 814 Q16566 2W4O
kinase IV (CaMKIV) protein kinase activity
4. Calspermin (Camk4) Testis Calmodulin-dependent 25050 P13234 Not available
protein kinase activity
5. A-kinase-anchoring protein Sperm flagellum Flagellated sperm motility 8852 Q5JQC9 Not available
(AKAP4) Sperm principal
Piece
6. Ubiquilin 3 (UBQLN3) Testis Cell cycle progression 50613 Q9H347 1YQB
in spermatogenesis
7. Calpain 11 (C APN11) Acrosome Cytoskeletal remodeling 11131 Q9UMQ6 Not available
and signal transduction
8. Gametogenetin (GGN) Sperm Spermatogenesis 199720 Q86UU5 Not available
9. Sperm acrosome-associated 4 Acrosome Not defined 171169 Q8TDM5 Not available
(SPACA4)
10. Spermatogenesis-associated 3 Spermatocyte Not defined 130560 Q8NHX4 Not available
(SPATA3)
(Continued)
228  Computational characterization and integrative analysis of proteins involved in spermatogenesis

Table 18.1 (Continued)  Details of biomarkers and drug targets discussed.


Protein
GO NCBI ID
S.no Target identified Localization class GeneID uniprot PDB Id
11. Family with sequence similarity Testis Not defined 84691 Q96KD3 Not available
71, member F1 (FAM71F1)

Proteomics Study
12. Doublesex- and mab-3-related Spermatagonia Male sex determination 1761 Q9Y5R6 4YJ0
transcription factor 1 (DMRT1) and differentiation
13. P antigen family member 1 Testis Uncharacterized 8712 O75459 Not available
(PAGE1)
14. Transition protein 1 (TNP1) Spermatid Sperm motility, spermatid 7141 P09430 Not available
developemt
15. Sperm mitochondria- Sperm motility 4184 P49901 Not available
associated cysteine-rich
protein (SMCP)
16. Actin-like protein 7B (ACTL7B) Testis-cytoskeleton 10880 Q9Y614 Not available
17. Defensin, beta 119 (DEFB119) Sertoli cell, testis Uncharacterized 245932 Q8N690 Not available
18. A kinase anchor protein 4 Sertoli cell, sperm Sperm motility 8852 Q5JQC9 Not available
(AKAP4)
19. Glyceraldehyde-3-phosphate Sperm Energy production in sperm 26330 O14556 3H9E
dehydrogenase (GAPDHS)
20. Tubulin beta 2B (TUBB2B) Sperm tail Constituent of microtubules 347733 Q9BVA1 Not available
21. Outer dense fiber protein Sperm tail Sperm tail outer dense fibers 4957 Q5BJF6 Not available
2,ODF2 constituent
22. Cytochrome c oxidase subunit Sperm tail Cytochrome C oxidase activity 1340 P14854 Not available
6B (COX6B)
23. Phospholipid hydroperoxide Sperm tail Glutathione peroxidase activity 2879 P36969 2GS3
glutathione
peroxidasemitochondrial
(PHGPx)
24. Voltage-dependent Sperm tail Mitochondrial membrane 7417 P45880 Not available
anionselective channel activity
protein 2 (VDAC2)
25. Heat shock-related 70-kDa Sperm tail Spermatogenesis, meiosis 3306 P54652 5FPN
protein 2 (HSPA2)
26. Sperm protein associated with Sperm tail Spermatid development 728695 Q9NS25 Not available
the nucleus on the X
chromosome B (SPANXB)
27. Keratin, type II cytoskeletal 1 Sperm tail Regulation of kinase activity 3848 P04264 4ZRY
(KRT1) of its partners like protein
kinase A
28. Isoaspartyl peptidase/ Sperm tail L-asparaginase and beta- 80150 Q7L266 4O0C
Lasparaginase (ASRGL1) aspartyl peptidase activity
29. Clusterin (CLU) Sperm tail Chaperone binding 1191 P10909 Not available

Exogenous Compound Based Models


30. N-cadherin Testis Assembly and disassembly 1000 P19022 Not available
of BTB and apical ES
31. β-catenin Testis 1499 P35222 4DJS
32. Occludin Testis 100506658 Q16625 1WPA,
1XAW,3G7C
33. Connexin (Cx43) Testis 30236 P17302 2LL2
(Continued)
References 229

Table 18.1 (Continued)  Details of biomarkers and drug targets discussed.


Protein
GO NCBI ID
S.no Target identified Localization class GeneID uniprot PDB Id
34. Zona occludens protein 1 Testis 7082 Q07157 4Q29
(ZO-1)
35. Actin-related protein 3 (Arp)3 Testis 10096 P61158 Not available
36. Epidermal growth factor Testis 2059 Q12929 2E8M
receptor substrate 8 (Eps8)
37. P-glycoprotein Testis Drug Transporters at BTB for 5243 P08183 Not available
38. Breast cancer-related protein Testis effecting protection to 9429 Q9UNQ0 Not available
(BCRP) developing spermatids form
drugs and other exogenous
compounds
39. Fascin 1 Testis Regulates ectoplasmic 6624 Q16658 1DFC
specialization
40. c-SRC Testis Kinases located at cell 6714 P12931 2HSH
41. c-Yes Testis junctions for modulating 7525 P07947 2HDA
42. Focal adhesion kinase (FAK) Testis assembly and disassembly 5747 Q05397 1MP8, 1K04
of BTB and apical ES
43. Microtubule affinity regulating Testis Conferring polarity to 57787 Q96L34 5ES1
kinase (MARK4) elongating spermatids
at apical ES
Maintains dynamics of
microtubules

REFERENCES 8. Castillo J, Estanyol JM, Ballesca JL, Oliva R. Human


1. Zegers-Hochschild F, Adamson GD, de Mouzon sperm chromatin epigenetic potential: Genomics,
J et  al. International Committee for Monitoring proteomics, and male infertility. Asian J Androl.
Assisted Reproductive Technology (ICMART) and 2015;17(4):601–609. doi:10.4103/1008-682X.153302.
the World Health Organization (WHO) revised 9. Jenardhanan P, Panneerselvam M, Mathur PP. Effect
glossary of ART terminology, 2009. Fertil Steril. of environmental contaminants on spermatogenesis.
2009;92(5):​1520–1524. doi:10.1016/j.fertnstert​. 2009​ Semin Cell Dev Biol. 2016; 59:126–140. DOI:10.1016/j​
.09.009. .semcdb.2016.03.024.
2. Skakkebaek NE, Rajpert-De Meyts E, Buck Louis 10. Manfo FP, Nantia EA, Mathur PP. Effect of environ-
GM et al. Male reproductive disorders and fertility mental contaminants on mammalian testis. Curr
trends: Influences of environment and genetic sus- Mol Pharmacol. 2014;7(2):119–135.
ceptibility. Physiol Rev. 2016;96(1):55–97. doi:10.1152​ 11. Mruk DD, Cheng CY. Environmental contaminants:
/­physrev.00017.2015. Is male reproductive health at risk? Spermatogenesis.
3. Stephen EH, Chandra A. Updated projections of 2011;1(4):283–290. doi:10.4161/spmg.1.4.18328.
infertility in the United States: 1995–2025. Fertil 12. Kovac JR, Pastuszak AW, Lamb DJ. The use of
Steril. 1998;70(1):30–34. genomics, proteomics, and metabolomics in iden-
4. Thonneau P, Marchand S, Tallec A. et al. Incidence tifying biomarkers of male infertility. Fertil Steril.
and main causes of infertility in a resident popula- 2013;​99(4):998–1007. doi:10.1016/j.fertnstert.2013.01​
tion (1,850,000) of three French regions (1988–1989). .​111.
Hum Reprod. 1991;6(6):811–816. 13. Luk AC, Chan WY, Rennert OM, Lee TL. Long non-
5. Sharlip ID, Jarow JP, Belker AM et al. Best prac- coding RNAs in spermatogenesis: Insights from
tice policies for male infertility. Fertil Steril. recent high-throughput transcriptome studies.
2002;77(5):873–882. Reproduction. 2014;147(5):R131–R141. doi:10.1530​
6. Turek PJ. Practical approaches to the diagnosis and /REP-13-0594.
management of male infertility. Nat Clin Pract Urol. 14. Upadhyay RD, Balasinor NH, Kumar AV, Sachdeva
2005;2(5):226–238. doi:10.1038/ncpuro0166. G, Parte P, Dumasia K. Proteomics in reproductive
7. Neto FT, Bach PV, Najari BB, Li PS, Goldstein M. biology: Beacon for unraveling the molecular com-
Genetics of male infertility. Curr Urol Rep. 2016;​ plexities. Biochim Biophys Acta. 2013;1834(1):8–15.
17(10):70. doi:10.1007/s11934-016-0627-x. doi:10.1016/j.bbapap.2012.10.004.
230  Computational characterization and integrative analysis of proteins involved in spermatogenesis

15. Huang DW, Sherman BT, Lempicki RA. Systematic 29. Wang H, Zhou Z, Xu M et al. A spermatogenesis-
and integrative analysis of large gene lists using related gene expression profile in human sperma-
DAVID bioinformatics resources. Nat Protoc. 2009;​ tozoa and its potential clinical applications. J Mol
4(1):44–57. doi:10.1038/nprot.2008.211. Med (Berl). 2004;82(5):317–324. doi:10.1007/s00109​
16. Huang DW, Sherman BT, Lempicki RA. Bioinfor­ -004-0526-3.
matics enrichment tools: Paths toward the compre- 30. Dear TN, Boehm T. Diverse mRNA expression patterns
hensive functional analysis of large gene lists. Nucleic of the mouse calpain genes Capn5, Capn6 and Capn11
Acids Res. 2009;37(1):1–13. doi:10.1093/nar/gkn923. during development. Mech Dev. 1999;​89(1–2):201–209.
17. The Gene Ontology Consortium. Gene Ontology 31. Li L, Liu G, Fu JJ. et al. Molecular cloning and
Consortium: Going forward. Nucleic Acids Res. characterization of a novel transcript variant of
2015;43:D1049–D1056. doi:10.1093/nar/gku1179. Mtsarg1 gene. Mol Biol Rep. 2009;36(5):1023–1032.
18. Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of doi:10.1007/s11033-008-9276-6.
genes and genomes. Nucleic Acids Res. 2000;28(1):​ 32. Zhao Q, Zhou Y, Cao Z, Zhu H, Huang P, Lu B.
27–30. Germ-cell specific protein gametogenetin protein
19. Szklarczyk D, Franceschini A, Wyder S et al. STRING 2 (GGN2), expression in the testis, and associa-
v10: Protein-protein interaction networks, integrated tion with intracellular membrane. Mol Reprod Dev.
over the tree of life. Nucleic Acids Res. 2015;43:D447– 2005;72(1):31–39. doi:10.1002/mrd.20313.
D452. doi:10.1093/nar/gku1003. 33. Abu-Halima M, Hammadeh M, Schmitt J et al.
20. Sali A, Blundell TL. Comparative protein model- Altered microRNA expression profiles of human
ling by satisfaction of spatial restraints. J Mol Biol. spermatozoa in patients with different spermato-
1993;234(3):779–815. doi:10.1006/jmbi.1993.1626. genic impairments. Fertil Steril. 2013;99(5):1249–
21. Raman S, Vernon R, Thompson J et al. Structure 1255. doi:10.1016/j.fertnstert.2012.11.054.
prediction for CASP8 with all-atom refinement 34. Abu-Halima M, Backes C, Leidinger P et al.
using Rosetta. Proteins. 2009;77(Suppl 9):89–99. doi:​ MicroRNA expression profiles in human testicular
10.1002/prot.22540. tissues of infertile men with different histopathologic
22. van Zundert GC, Rodrigues JP, Trellet M et al. The patterns. Fertil Steril. 2014;101(1):78–86. doi:10.1016/j​
HADDOCK2.2 web server: User-friendly integra- .fertnstert.2013.09.009.
tive modeling of biomolecular complexes. J Mol 35. Yu Z, Raabe T, and Hecht NB. MicroRNA Mirn122a
Biol. 2016;428(4):720–725. doi:10.1016/j.jmb.2015.09​ reduces expression of the posttranscriptionally
.014. regulated germ cell transition protein 2 (Tnp2)
23. Halgren TA, Murphy RB, Friesner RA et al. Glide: messenger RNA (mRNA) by mRNA cleavage. Biol
A new approach for rapid, accurate docking and Reprod. 2005;73(3):427–433. doi:10.1095/biolreprod​
scoring. 2. Enrichment factors in database screen- .105.040998.
ing. J Med Chem. 2004;47(7):1750–1759. doi:10.1021​ 36. Hayashi T, Kageyama Y, Ishizaka K, Xia G, Kihara
/­jm030644s. K, Oshima H. Requirement of Notch 1 and its ligand
24. Pronk S, Pall S, Schulz R et al. GROMACS 4.5: A jagged 2 expressions for spermatogenesis in rat and
high-throughput and highly parallel open source human testes. J Androl. 2001;22(6):999–1011.
molecular simulation toolkit. Bioinformatics. 2013:​ 37. Zhuang X, Li Z, Lin H et al. Integrated miRNA and
29(7):845–854. doi:10.1093/bioinformatics/btt055. mRNA expression profiling to identify mRNA tar-
25. Lin YH, Lin YM, Teng YN, Hsieh TY, Lin YS, Kuo gets of dysregulated miRNAs in non-obstructive
PL. Identification of ten novel genes involved in azoospermia. Sci Rep. 2015;5:7922. doi:10.1038​
human spermatogenesis by microarray analysis of /­srep07922.
testicular tissue. Fertil Steril. 2006;86(6):1650–1658. 38. Shang PW, Baarends M, Hoogerbrugge J et al.
doi:10.1016/j.fertnstert.2006.04.039. Functional transformation of the chromatoid body
26. Feig C, Kirchhoff C, Ivell R, Naether O, Schulze W, in mouse spermatids requires testis-specific serine/
Spiess AN. A new paradigm for profiling testicular threonine kinases. J Cell Sci. 2010;123(Pt 3):331–9
gene expression during normal and disturbed human doi:10.1242/jcs.059949.
spermatogenesis. Mol Hum Reprod. 2007;13(1):33– 39. Spiridonov NA, Wong L, Zerfas P.M et al.
43. doi:10.1093/molehr/gal097. Identification and characterization of SSTK, a serine/
27. Liu F, Jin S, Li N, Liu X, Wang H, Li J. Comparative threonine protein kinase essential for male fertility.
and functional analysis of testis-specific genes. Biol Mol Cell Biol. 2005;25(10):4250–4261. DOI:10.1128​
Pharm Bull. 2011;34(1):28–35. /­MCB.25.10.4250–4261.2005.
28. Malcher A, Rozwadowska N, Stokowy T et al. 40. Yuan S, Qin W, Riordan CR, McSwiggin H, Zheng
Potential biomarkers of nonobstructive azoosper- H, Yan W. Ubqln3, a testis-specific gene, is dispens-
mia identified in microarray gene expression analy- able for embryonic development and spermatogen-
sis. Fertil Steril. 2013;100(6):1686–1694. doi:10.1016/j​ esis in mice. Mol Reprod Dev. 2015;82(4):266–267.
.fertnstert.2013.07.1999. DOI:10.1002/mrd.22475.
References 231

41. Guo X, Zhao C, Wang F et al. Investigation of human 53. Miki K, Willis WD, Brown PR, Goulding EH,
testis protein heterogeneity using 2-dimensional­elec- Fulcher KD, Eddy EM. Targeted disruption of the
trophoresis. J Androl. 2010;31(4):419–429. doi:10.2164​ Akap4 gene causes defects in sperm flagellum and
/­jandrol.109.007534. motility. Dev Biol. 2002;248(2):331–342.
42. von Kopylow K, Kirchhoff C, Jezek D et al. Screening 54. Miki K, Qu W, Goulding EH et al. Glyceraldehyde
for biomarkers of spermatogonia within the human 3-phosphate dehydrogenase-S, a sperm-specific gly-
testis: A whole genome approach. Hum Reprod. colytic enzyme, is required for sperm motility and
2010;25(5):1104–1112. DOI:10.1093/humrep/deq053. male fertility. Proc Natl Acad Sci U S A. 2004;101(47):​
43. Djureinovic D, Fagerberg L, Hallstrom B et al. The 16501–16506. DOI:10.1073/pnas.0407708101.
human testis-specific proteome defined by tran- 55. Liu Y, Guo Y, Song N et al. Proteomic pattern changes
scriptomics and antibody-based profiling. Mol Hum associated with obesity-induced asthenozoosper-
Reprod. 2014;20(6):476–488. DOI:10.1093/molehr​ mia. Andrology. 2015;3(2):247–259. DOI:10.1111/andr​
/­gau018. .289.
44. Matson CK, Murphy MW, Griswold MD, Yoshida S, 56. Li HY, Zhang H. Proteome analysis for profiling
Bardwell VJ, Zarkower D. The mammalian double- infertility markers in male mouse sperm after carbon
sex homolog DMRT1 is a transcriptional gatekeeper ion radiation. Toxicology. 2013;306:85–92. DOI:10​
that controls the mitosis versus meiosis decision .1016/j.tox.2013.02.008.
in male germ cells. Dev Cell. 2010;19(4):612–624. 57. Wang B, Murakami M, Eguchi-Kasai K et al. Effects
DOI:10.1016/j.devcel.2010.09.010. of prenatal irradiation with an accelerated heavy-
45. Li J, Liu F, Liu X et al. Mapping of the human tes- ion beam on postnatal development in rats: I.
ticular proteome and its relationship with that of the Neurophysiological alterations. Radiat Res. 2005;164​
epididymis and spermatozoa. Mol Cell Proteomics. (4 Pt 2):561–566.
2011;10(3):M110 004630. DOI:10.1074/mcp.M110​ 58. Huang Q, Luo L, Alamdar A et al. Integrated pro-
.004630. teomics and metabolomics analysis of rat testis: Mech­
46. Liu X, Liu F. Indepth mapping of human testicu- anism of arsenic-induced male reproductive toxicity.
lar and epididymal proteins and their functional Sci Rep. 2016;6:32518. DOI:10.1038/srep32518.
association with spermatozoa. Mol Med Rep. 2015;​ 59. Cheng CY, Mruk DD. The blood-testis barrier and its
12(1):173–179. DOI:10.3892/mmr.2015.3435. implications for male contraception. Pharmacol Rev.
47. Yamakawa K, Yoshida K, Nishikawa H, Kato T, 2012;64(1):16–64. DOI:10.1124/pr.110.002790.
Iwamoto T. Comparative analysis of interindividual 60. Mok KW, Lie PP, Mruk DD et al. The apical ectoplas-
variations in the seminal plasma proteome of fertile mic specialization-blood-testis barrier functional
men with identification of potential markers for azo- axis is a novel target for male contraception. Adv Exp
ospermia in infertile patients. J Androl. 2007;28(6):​ Med Biol. 2012;763:334–355.
858–865. DOI:10.2164/jandrol.107.002824. 61. Boekelheide K, Neely MD, Sioussat TM. The Sertoli
48. Hashemitabar M, Sabbagh S, Orazizadeh M, cell cytoskeleton: A target for toxicant-induced
Ghadiri A, Bahmanzadeh M. A proteomic analysis germ cell loss. Toxicol Appl Pharmacol. 1989;101(3):​
on human sperm tail: Comparison between normo- 373–389.
zoospermia and asthenozoospermia. J Assist Reprod 62. Lee NP, Cheng CY. Ectoplasmic specialization, a
Genet. 2015;32(6):853–863. DOI:10.1007/s10815-015​ testis-specific cell-cell actin-based adherens junction
-0465-7. type: Is this a potential target for male contraceptive
49. Fackenthal JD, Turner FR, Raff EC. Tissue-specific development? Hum Reprod Update. 2004;10(4):349–
microtubule functions in Drosophila spermato- 369. DOI:10.1093/humupd/dmh026.
genesis require the beta 2-tubulin isotype-specific 63. Gao Y, Mruk DD, Cheng CY. Sertoli cells are the
carboxy terminus. Dev Biol. 1993;158(1):213–27. target of environmental toxicants in the testis—A
DOI:10.1006/dbio.1993.1180. mechanistic and therapeutic insight. Expert Opin
50. Cao W, Gerton GL, Moss SB. Proteomic profiling of Ther Targets. 2015;19(8):1073–1090. DOI:10.1517/147
accessory structures from the mouse sperm flagel- 28222.2015.1039513.
lum. Mol Cell Proteomics. 2006;5(5):801–810. DOI:10​ 64. Manfo FP, Jubendradass R, Nantia EA, Moundipa PF,
.1074/mcp.M500322-MCP200. Mathur PP. Adverse effects of bisphenol A on male
51. Petersen C, Fuzesi L, Hoyer-Fender S. Outer dense reproductive function. Rev Environ Contam Toxicol.
fibre proteins from human sperm tail: Molecular 2014;228:57–82. DOI:10.1007/978-3-319-01619-1_3.
cloning and expression analyses of two cDNA tran- 65. Li MW, Mruk DD, Lee WM, Cheng CY. Disruption
scripts encoding proteins of approximately 70 kDa. of the blood-testis barrier integrity by bisphenol A
Mol Hum Reprod. 1999;5(7):627–635. in vitro: Is this a suitable model for studying blood-
52. Eddy EM, Toshimori K, O’Brien DA. Fibrous sheath testis barrier dynamics? Int J Biochem Cell Biol.
of mammalian spermatozoa. Microsc Res Tech. 2009;41(11):2302–2314. DOI:10.1016/j.biocel.2009.05​
2003;61(1):103–115. DOI:10.1002/jemt.10320. .016.
232  Computational characterization and integrative analysis of proteins involved in spermatogenesis

66. Li MW, Mruk DD, Lee WM, Cheng CY. Connexin 43 77. Lui WY, Lee WM, Cheng CY. Transforming growth
is critical to maintain the homeostasis of the blood-­ factor beta3 regulates the dynamics of Sertoli cell
testis barrier via its effects on tight junction reassembly. tight junctions via the p38 mitogen-activated protein
Proc Natl Acad Sci U S A. 2010;107(42):17998–8003. kinase pathway. Biol Reprod. 2003;68(5):1597–1612.
DOI:10.1073/pnas.1007047107. DOI:10.1095/biolreprod.102.011387.
67. Salian S, Doshi T, Vanage G. Neonatal exposure of 78. Siu ER, Wong EW, Mruk DD et al. An occludin-
male rats to bisphenol A impairs fertility and expres- focal adhesion kinase protein complex at the blood-
sion of Sertoli cell junctional proteins in the testis. testis barrier: A study using the cadmium model.
Toxicology. 2009;265(1–2):56–67. DOI:10.1016/j.tox​ Endocrinology. 2009;150(7):3336–3344. DOI:10.1210​
.2009.09.012. /en​.2008-1741.
68. Xiao X, Mruk DD, Tang EI et al. Environmental 79. Cheng CY, Silvestrini B, Grima J et al. Two new
toxicants perturb human Sertoli cell adhesive func- male contraceptives exert their effects by depleting
tion via changes in F-actin organization mediated by germ cells prematurely from the testis. Biol Reprod.
actin regulatory proteins. Hum Reprod. 2014;29(6):​ 2001;65(2):449–461.
1279–1291. DOI:10.1093/humrep/deu011. 80. Cheng CY, Mo M, Grima J et al. Indazole carboxylic
69. Cheng CY, Mruk DD. Regulation of spermiogenesis, acids in male contraception. Contraception. 2002;
spermiation and blood-testis barrier dynamics: Novel 65(4):265–268.
insights from studies on Eps8 and Arp3. Biochem J. 81. Grima J, Silvestrini B, Cheng CY. Reversible inhibition
2011;435(3):553–562. DOI:10.1042/BJ20102121. of spermatogenesis in rats using a new male contracep-
70. Qian W, Zhu J, Mao C et al. Involvement of CaM- tive, 1-(2,4-dichlorobenzyl)-indazole-3-​carbohydrazide­.
CaMKII-ERK in bisphenol A-induced Sertoli cell Biol Reprod. 2001;64(5):1500–1508.
apoptosis. Toxicology. 2014;324:27–34. DOI:10.1016/j​ 82. Mruk DD, Cheng CY. Cell-cell interactions at the
.tox.2014.06.001. ectoplasmic specialization in the testis. Trends
71. Toledo FD, Perez LM, Basiglio CL, Ochoa JE, Endocrinol Metab. 2004;15(9):439–447. DOI:10.1016​
Sanchez Pozzi EJ, Roma MG. The Ca(2)(+)-​ /j​.tem​.2004.09.009.
calmodulin­-Ca(2)(+)/calmodulin-dependent protein 83. Wong EW, Mruk DD, Lee WM, Cheng CY. Par3/
kinase II signaling pathway is involved in oxidative Par6 polarity complex coordinates apical ectoplas-
stress-induced mitochondrial permeability tran- mic specialization and blood-testis barrier restruc-
sition and apoptosis in isolated rat hepatocytes. turing during spermatogenesis. Proc Natl Acad Sci
Arch Toxicol. 2014;88(9):1695–1709. DOI:10.1007​ U S A. 2008;105(28):9657–9662. DOI:10.1073/pnas​
/­s00204-014-1219-5. .0801527105.
72. Wang Q, Zhao XF, Ji YL et al. Mitochondrial 84. Mruk DD, Cheng CY. Testin and actin are key
signaling pathway is also involved in bisphenol molecular targets of adjudin, an anti-spermatogenic
A induced germ cell apoptosis in testes. Toxicol agent, in the testis. Spermatogenesis. 2011;1(2):137–
Lett. 2010;199(2):129–135. DOI:10.1016/j.toxlet.2010​ 146. DOI:10.4161/spmg.1.2.16449.
.08​.014. 85. Wan HT, Mruk DD, Wong CK, Cheng CY. The apical
73. D’Cruz SC, Jubendradass R, Mathur PP. Bisphenol ES-BTB-BM functional axis is an emerging target for
A induces oxidative stress and decreases levels of toxicant-induced infertility. Trends Mol Med. 2013;​
insulin receptor substrate 2 and glucose trans- 19(7):396–405. DOI:10.1016/j.molmed.2013.03.006.
porter 8 in rat testis. Reprod Sci. 2012;19(2):163–172. 86. Cheng CY. Toxicants target cell junctions in the
DOI:10.1177/1933719111415547. testis: Insights from the indazole-carboxylic acid
74. D’Cruz SC, Jubendradass R, Jayakanthan M, Rani model. Spermatogenesis. 2014;4(2):e981485. DOI:10​
SJ, Mathur PP. Bisphenol A impairs insulin signaling .4161/21565562.2014.981485.
and glucose homeostasis and decreases steroidogen- 87. Tang EI, Xiao X, Mruk DD et al. Microtubule
esis in rat testis: An in vivo and in silico study. Food ­affinity-regulating kinase 4 (MARK4) is a compo-
Chem Toxicol. 2012;50(3–4):1124–133. DOI:10.1016/j​ nent of the ectoplasmic specialization in the rat tes-
.fct.2011.11.041. tis. Spermatogenesis. 2012;2(2):117–126. DOI:10.4161​
75. Lui WY, Wong CH, Mruk DD, Cheng CY. TGF-beta3 /­spmg​.20724.
regulates the blood-testis barrier dynamics via the 88. Marx A, Nugoor C, Panneerselvam S, Mandelkow E.
p38 mitogen activated protein (MAP) kinase path- Structure and function of polarity-inducing kinase
way: An in vivo study. Endocrinology. 2003;144(4):​ family MARK/Par-1 within the branch of AMPK/
1139–1142. DOI:10.1210/en.2002-0211. Snf1-related kinases. FASEB J. 2010;24(6):1637–1648.
76. Wong CH, Mruk DD, Siu MK, Cheng CY. Blood- DOI:10.1096/fj.09-148064.
testis barrier dynamics are regulated by {alpha}2- 89. Matenia D, Mandelkow EM. The tau of MARK: A
macroglobulin via the c-Jun N-terminal protein polarized view of the cytoskeleton. Trends Biochem
kinase pathway. Endocrinology. 2005;146(4):1893– Sci. 2009;34(7):332–342. DOI:10.1016/j.tibs.2009.03​
1908. DOI:10.1210/en.2004-1464. .008.
References 233

90. Gungor-Ordueri NE, Celik-Ozenci C, Cheng CY. 104. Lee NP and Cheng CY. Protein kinases and adherens
Fascin 1 is an actin filament-bundling protein that junction dynamics in the seminiferous epithelium
regulates ectoplasmic specialization dynamics in of the rat testis. J Cell Physiol. 2005;202(2):344–360.
the rat testis. Am J Physiol Endocrinol Metab. 2014;​ DOI:10.1002/jcp.20119.
307(9):E738–E753. DOI:10.1152/ajpendo.00113.2014. 105. Elias BC, Suzuki T, Seth A et al. Phosphorylation of
91. Edwards RA, Bryan J. Fascins, a family of actin bun- Tyr-398 and Tyr-402 in occludin prevents its interac-
dling proteins. Cell Motil Cytoskeleton. 1995;32(1):1–9. tion with ZO-1 and destabilizes its assembly at the
DOI:10.1002/cm.970320102. tight junctions. J Biol Chem. 2009;284(3):1559–1569.
92. Cheng CY, Mruk D, Silvestrini B et al. AF-2364 DOI:10.1074/jbc.M804783200.
[1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide] 106. Xiao X, Mruk DD, Lee WM, Cheng CY. c-Yes
is a potential male contraceptive: A review of recent regulates cell adhesion at the blood-testis barrier
data. Contraception. 2005;72(4):251–261. DOI:10.1016/j​ and the apical ectoplasmic specialization in the
.contraception.2005.03.008. seminiferous epithelium of rat testes. Int J Biochem
93. Su L, Cheng CY, Mruk DD. Drug transporter, Cell Biol. 2011;43(4):651–665. DOI:10.1016/j.biocel​
P-glycoprotein (MDR1), is an integrated component .2011.01​.008.
of the mammalian blood-testis barrier. Int J Biochem 107. Chen YH, Lu Q, Goodenough DA, Jeansonne B.
Cell Biol. 2009;41(12):2578–2587. DOI:10.1016/j.biocel​ Nonreceptor tyrosine kinase c-Yes interacts with
.2009.08.015. occludin during tight junction formation in canine
94. Su L, Mruk DD, Lui WY, Lee WM, Cheng CY. kidney epithelial cells. Mol Biol Cell. 2002;13(4):1227–
P-glycoprotein regulates blood-testis barrier dynam- 1237. DOI:10.1091/mbc.01-08-0423.
ics via its effects on the occludin/zonula occludens 1 108. Su L, Cheng CY, Mruk D.D. Adjudin-mediated
(ZO-1) protein complex mediated by focal adhesion Sertoli-germ cell junction disassembly affects Sertoli
kinase (FAK). Proc Natl Acad Sci U S A. 2011;108(49):​ cell barrier function in vitro and in vivo. Int J Biochem
19623–19628. DOI:10.1073/pnas.1111414108. Cell Biol. 2010;42(11):1864–1875. DOI:10.1016/j.biocel​
95. Flens MJ, Zaman GJ, van der Valk P et al. Tissue dis- .2010.08.004.
tribution of the multidrug resistance protein. Am J 109. Lie PP, Mruk DD, Mok KW, Su L, Lee WM, Cheng
Pathol. 1996;148(4):1237–1247. CY. Focal adhesion kinase-Tyr407 and -Tyr397
96. Qian X, Mruk DD, Wong EW, Cheng CY. Breast exhibit antagonistic effects on blood-testis barrier
cancer resistance protein regulates apical ectoplas- dynamics in the rat. Proc Natl Acad Sci U S A. 2012;​
mic specialization dynamics stage specifically in 109(31):12562–12567. DOI:10.1073/pnas.1202316109.
the rat testis. Am J Physiol Endocrinol Metab. 2013;​ 110. Xiao X, Mruk DD, Jenardhanan P et al. Nonreceptor
304(7):E757–E769 DOI:10.1152/ajpendo.00645.2012. protein kinases c-Src, c-Yes, and FAK are biomark-
97. Wan HT, Mruk DD, Tang EI et al. Role of non- ers for male contraceptive research. In Lee NP,
receptor protein tyrosine kinases in spermatid trans- Cheng CY, JM Luk, eds. New Advances on Disease
port during spermatogenesis. Semin Cell Dev Biol. Biomarkers and Molecular Targets in Biomedicine.
2014;30:65–74. DOI:10.1016/j.semcdb.2014.04.013. Totowa, NJ: Humana Press; 2013:1–25. DOI:10.1007​
98. Gungor-Ordueri NE, Mruk DD, Wan HT et al. New /978-1-62703-456-2_1.
insights into FAK function and regulation during sper- 111. Su L., Jenardhanan P., Mruk DD et al. Role of
matogenesis. Histol Histopathol. 2014;​29(8):​977–989. P-glycoprotein at the blood-testis barrier on adjudin
99. Xiao X, Mruk DD Cheng FL, Cheng C.Y. C-Src and distribution in the testis: A revisit of recent data. Adv
c-Yes are two unlikely partners of spermatogenesis Exp Med Biol. 2012;763:318–333.
and their roles in blood-testis barrier dynamics. Adv 112. Qian X, Cheng YH, Jenardhanan P et al. Adjudin
Exp Med Biol. 2012;763:295–317. disrupts spermatogenesis by targeting drug trans-
100. Wine RN and Chapin RE. Adhesion and signaling porters: Lesson from the breast cancer resistance
proteins spatiotemporally associated with spermia- protein (BCRP). Spermatogenesis. 2013;3(2):e24993.
tion in the rat. J Androl. 1999;20(2):198–213. DOI:10.4161/spmg.24993.
101. Mruk DD, Wong CH, Silvestrini B, Cheng CY. A 113. Cheng YH, Jenardhanan P, Mathur PP, Qian X,
male contraceptive targeting germ cell adhesion. Nat Xia W, Silvestrini B, Cheng CY. Interaction of oligo-
Med. 2006;12(11):1323–1328. DOI:10.1038/nm1420. meric breast cancer resistant protein (BCRP) with
102. Mok KW, Mruk DD, Lie PP, Lui WY, Cheng CY. adjudin: A  male contraceptive with anti-cancer
Adjudin, a potential male contraceptive, exerts its activity. Curr Mol Pharmacol. 2014;7(2):147–153.
effects locally in the seminiferous epithelium of 114. Jenardhanan P, Mannu J, Mathur PP. The structural
mammalian testes. Reproduction. 2011;141(5):571– analysis of MARK4 and the exploration of specific
580. DOI:​10.1530/REP-10-0464. inhibitors for the MARK family: A computational
103. Cheng CY, Mruk DD. New frontiers in nonhormonal approach to obstruct the role of MARK4 in prostate
male contraception. Contraception. 2010;82(5):476– cancer progression. Mol Biosyst. 2014;10(7):1845–
482. DOI:10.1016/j.contraception.2010.03.017. 1868. DOI:10.1039/c3mb70591a.
Effects of chemical exposures on testis
cell-cell interactions and endocrine
19
function
RACHEL C. KNIGHT, JENNIFER R. PANIZZI, and BENSON T. AKINGBEMI

INTRODUCTION EXPRESSION OF STEROID HORMONE RECEPTORS


Increased exposure to environmental endocrine dis- IN TESTICULAR CELLS
rupting chemicals (EDCs) and possible association with The testis consists of a tubular compartment of seminif-
negative reproductive effects in the population continue erous tubules containing Sertoli cells and germ cells at
to raise public health concerns. An apparent decline in different stages of development.7 An interstitial compart-
sperm production capacity in men, a higher incidence of ment between seminiferous tubules includes connective
urogenital anomalies such as cryptorchidism and hypo- tissue (i.e., collagen fibers, fibroblasts, mesenchymal cells,
spadias in newborns, testicular cancer in young adults, as and immune cells), blood vessels, lymphatics, nerves, and
well as the obesity epidemic in the population1 apparently Leydig cells (Figure 19.1). Leydig cells are the major source
correspond to a marked increase in the use of industrial of androgens, including testosterone (T) and dihydrotes-
chemicals in the last 40 years.2,3 The general population is tosterone (DHT).7 Cloning of the ESR1 (formerly ERα) and
exposed to EDCs present in food, air, water, and soil while impairment of spermatogenesis in ESR1 knockout mice
those with occupations in the agricultural, petrochemi- indicated a physiological role for estrogen in the testis.8 A
cal, and construction industry are subject to even greater subsequently cloned estrogen receptor subtype was desig-
exposures.4 Therefore, it is imperative that further stud- nated ESR2.9 In rats, ESR1 was expressed in Sertoli cells,
ies be conducted to determine the precise effects chemical peritubular myoid cells, as well as in rat fetal and adult
toxicants have on human reproductive and overall health. Leydig cells, while ESR2 is found in rat germ cells10–12 and
was localized to adult mouse Leydig cells.13,14 In human
CLASSES OF ENDOCRINE DISRUPTING CHEMICALS testes, ESR2 expression began at 16 weeks of gestation,15
EDCs are classified based on their applications, chem- and ESR1 and ESR2 were both expressed in Sertoli cells,
istry, and/or hormonal activity (Table 19.1). Chemicals spermatocytes, and elongating spermatids in the adult.16,17
are considered estrogenic if they bind to estrogen recep- Only ESR2 was present in human Leydig cells.18 The aro-
tors (ESRs) and mimic activity of the natural estrogen matase enzyme, which catalyzes conversion of androgens
17β-estradiol (E2). Compounds that prevent E2 binding into estrogens, was found predominantly in prepubertal
of the ESR and/or suppress E2 biosynthesis are consid- rat Sertoli cells and germ cells while expression appeared
ered antiestrogenic. Chemicals with ESR agonist activ- in Leydig cells after 21 days of age.19 Notably, estrogen bio-
ity (called xenoestrogens) include pesticides (e.g., 1, 1, synthesis and aromatase activity occur mostly in adipose
1-trichloro-2, 2-bis [p-chlorophenyl] ethane [DDT], tissue in male subjects because the testis synthesizes only
methoxychlor, dimethoate, and chlordecone) and indus- about 25% of E2 in circulation.20
trial pollutants [e.g., polychlorinated biphenols (PCBs)],
alkylphenols, and bisphenol A (BPA). While PCBs are
GENERAL MECHANISMS OF ENDOCRINE DISRUPTION
present in industrial applications,5 BPA is used in the
manufacture of food packaging products and dental seal- Estrogenic chemicals
ants while alkylphenols are used in the manufacture of Steroid hormones regulate target cell activity by activation
industrial detergents. Also contributing to EDC levels in of their cognate receptors (ESR1, ESR2, ARs) and bind-
the environment are pharmaceuticals in human and ani- ing of estrogen and androgen response elements (EREs
mal waste (e.g., the oral contraceptive ethinyl estradiol) and AREs) in promoters of target genes.21,22 Estrogens
and dietary estrogens (e.g., soy isoflavones).5 Additionally, may also induce rapid nongenomic responses through
other compounds interfere with and/or suppress andro- plasma membrane receptors (mESR1 and mESR2) and
gen receptor- (AR-) mediated activity acting as antian- cause Ca++ release and changes in intra- and extracellular
drogens, such as the fungicide vinclozolin, the persistent signaling.23,24 A seven-transmembrane estrogen receptor,
DDT metabolite p, p-chlorophenyl, ethylene (p-p’-DDE), GPR30, is present in the brain, placenta, ovary, testis, and
and phthalate esters used in the manufacture of plastics.6 prostate.25 ESR and GPR30 agonists induced activation of

234
General mechanisms of endocrine disruption  235

Table 19.1  Classes of endocrine disrupting chemicals.


Substances Category Mechanism of action
Bisphenol A (BPA) Plasticizer Estrogen receptor agonist
Phthalates Plasticizer Androgen receptor antagonist
Dichlorodiphenyl-trichloroethane (DDT) Pesticide Estrogen receptor agonist
Polychlorinated biphenyls (PCBs) Industrial by-product Thyroid hormone receptor antagonist
Diethylstilbestrol Synthetic, non-steroidal estrogen Estrogen receptor antagonist
Alkylphenol Surfactant Estrogen receptor agonist
Vinclozolin Fungicide Androgen receptor antagonist
Soy isoflavones Dietary estrogens Estrogen receptor agonists
Dioxins Environmental toxins Estrogen receptor agonists

Elongated spermatids
Round spermatids

Secondary spermatocytes
Sertoli
cell
Sertoli Primary spermatocyte
cell

Basal lamina Spermatogonium

Fibroblast
Peritubular myoid
cells

Capillary Capillary

Leydig cells

Figure 19.1  Cellular composition of seminiferous tubules in testis: Supporting Sertoli cells and germ cells at different stages of
development (spermatogonia, primary and secondary spermatocytes, and round and elongated spermatids); and, interstitial con-
nective tissue containing fibroblasts and Leydig cells (not drawn to scale).

secondary messengers and signaling cascades mediated by hepatic mono-oxygenase activity, thereby promoting
tyrosine kinase receptors and protein kinases (e.g., insulin androgen degradation and disruption of AR-mediated
growth factor- [IGF-] insulin receptor [IR], protein kinase activity.32 Interestingly, BPA decreased E2 secretion while
B [Akt], mitogen-activated protein kinases [MAPK], pro- DEHP increased aromatase expression and E2 biosynthe-
tein kinase A [PKA], and protein kinase C [PKC]).26,27 sis in rodent testis, which increased serum E2 concentra-
Interestingly, BPA exhibited high binding affinities for tions (Figure 19.2).33,34 Therefore, the estrogenic and/or
GPR30 in ESR-negative cells,28 implying that EDC action antiandrogenic activity of EDCs possibly varies with the
may be mediated by multiple transcription factors. tissue and stage of development, and dosage and duration
of chemical exposure.
Antiandrogenic chemicals
Chemicals may block AR binding and activation by endog- Antithyroid agents
enous androgen. For example, the fungicide vinclozolin The thyroid hormone receptor (TR) belongs to the nuclear
has been shown to bind DNA and inhibit dihydrotestos- family of transcription factors.35 Thus, EDC-induced
terone- (DHT-) induced transcriptional activation of the disruption of thyroid function may result from altered
AR.29 Similarly, the phthalate esters dibutyl phthalate TR expression, changes in triiodothyronine (T3) and
(DBP) and di(2-ethylhexyl) phthalate (DEHP) inhibited tetraiodothyronine (T4) concentrations, disruption of
AR transcriptional activity and disrupted sexual differen- coactivator recruitment, and/or interference with bind-
tiation in male rats.30,31 DDT-related compounds inhibited ing of TR-ligand complexes to thyroid hormone response
236  Effects of chemical exposures on testis cell-cell interactions and endocrine function

0.12

Basal leydig cell E2 production


* 1.2 *
*
Serum E2 levels (ng/mL) 1.1 1.0

(ng/106 cells • 3h)

Pituitary LHβ subunit protein/ACTB


Pituitary LHβ subunit protein/ACTB
0.08 0.8 1.0
0.9 0.8
0.8
0.7 0.6
0.04 0.4 0.6
0.5 *
0.4
0.4
0.00 0.0 0.3
(a) (b) 0.2
0.2
2.4 0.8 0.1
*
production (ng/106 cells • 3 h)

0.0
LH-stimulated leydig cell E2

* 0.0 25 0 50
Aromatase mRNA levels 0
1.8 (aromatase/S16) 0.6 (a) Dose of BPA (µg/kg bw) (b) Dose of DEHP (µg/kg bw)

*
1.2 0.4

Pituitary FSHβ subunit protein/ACTB


1.0

Pituitary FSHβ subunit protein/ACTB


0.8
0.6 0.2
0.8
0.6
0.0 0 0.6
0 mg/kg/day DEHP Aromatase 0.4
290 bp
10 mg/kg/day DEHP 0.4 *
S16 *
100 mg/kg/day DEHP 150 bp 0.2
0 10 100 0.2
Dose of DEHP (mg/kg/day)
(c) (d)
0.0 0.0
0 25 0 50
(c) Dose of BPA (µg/kg bw) (d) Dose of DEHP (µg/kg bw)
Figure 19.2  Serum E2 concentrations (a) were associated
with increased Leydig cell E2 production (b and c) and increased
aromatase gene expression (d) after DEHP treatment of rats Figure 19.3  Male rats were exposed to BPA and DEHP by
from 21 to 48 days of age (n = 10). Leydig cells were incubated maternal gavage from gestational day 12 to 21. Gonadotropin
in culture media for 3 h, and E2 was measured in aliquots of the subunit protein was analyzed in pituitary glands from 90-day-old
spent media by RIA and normalized to nanograms per 106 cells. male rats (n = 6 animals/group): LHβ (a and b) and FSHβ (c and d).
S16, rat ribosomal protein S16; *, p < 0.01 compared to control. Protein measurements were done by Western blotting proce-
Data are presented as mean +/– SEM. (See further Proc. Natl. dures using antisera specific to rat LHβ and FSHβ and the appro-
Acad. Sci. U S A. 2004; 103:775-780.) priate secondary antibodies. Assays of samples were repeated
at least four times and protein levels normalized to beta-actin
elements (TREs).21,36–38 For example, organohalogen (ACTB). *, p < 0.05 vs control. (See further Endocrinology. 2015;156
chemicals decreased thyroid hormone biosynthesis,39,55 (12):4672–4685.)
increased biliary excretion of T4 by induction of uridine
5’-diphospho (UDP)-glucuronyl transferase activity,40 Men with inactivating mutations of the aromatase and
and prevented T4 binding to the TR.41,42 BPA phthalates, ESR1 gene had elevated serum FSH and LH concentra-
and DDT induced T3 binding to the retinoid X receptor/ tions.47,48 Exposure of newborn male rats to environmen-
thyroid receptor complex and regulated SF-1 and steroido- tal estrogens results in delayed and sustained high levels
genic acute regulatory protein (StAR) gene expression in of prolactin,49 a potent inhibitor of hypothalamic GnRH
mouse Leydig cells.43 As well, serum phthalate, BPA, and production50 Additionally, gestational exposures of male
cadmium concentrations were inversely related to thyroid rats to BPA and DEHP decreased pituitary LH protein
hormone levels in male subjects.44 expression in adulthood51 (Figure 19.3). Therefore, neuro-
endocrine pathways regulating the reproductive axis are
Endocrine disrupting chemical (EDC) affection targets for chemical agents.
of neuroendocrine control of testicular function
Hypothalamic pathways regulate gonadotropin-releasing EDC TARGETING OF TESTICULAR CELLS
hormone (GnRH) secretion and control pituitary luteiniz- Chemical exposure effects are presumably exerted in mul-
ing hormone (LH) and follicle-stimulating hormone (FSH) tiple testicular cells with implications for exocrine and
secretion and release while gonadal steroids and other fac- endocrine function. The primary androgen T stimulates
tors acting through feedback mechanisms regulate GnRH sexual differentiation in the prenatal period, promotes
and gonadotropin secretion. Estrogenic activity was asso- development of male secondary sex characteristics, sup-
ciated with regulation of hypothalamic GnRH pulses ports hormonal imprinting of the liver, prostate, and hypo-
and GnRH receptor gene expression in gonadotropes.45,46 thalamus, and maintains the male phenotype and fertility.6
EDC targeting of testicular cells  237

Sertoli cells phosphorylation of steroid hormone receptor coactiva-


Somatic Sertoli cells produce several factors, including tors, ERK1/2, Akt, and promoted survival and prolifera-
androgen-binding protein (ABP), sulfated glycoproteins tion of PGCs.63 Other agents such as cadmium and BPA
(SGP) 1/2, transferrin, and inhibin B, which support germ caused germ cell depletion in the seminiferous epithelium
cell development. Exposures to perfluorooctanesulfonic acting via mitogen-activated protein kinases (MAPKs).64
acid (PFOS) decreased inhibin B expression in testes of adult Recent studies have demonstrated that exposures to the
mice and disrupted feedback regulation of pituitary FSH EDC vinclozolin disrupted epigenetic pathways leading to
secretion.52 Exposure to bisphenol AF (an analog of BPA) transgenerational effects by modifying germ cell differen-
increased serum gonadotropin concentrations associated tiation through dysregulation of microRNAs (mmu-miR-
with decreased testicular inhibin B mRNA in male rats, and 23b and mmu-miR-21) and disruption of the Lin28/let-7/
serum BPA concentrations were inversely related to inhibin Blimp1 pathway in germ cells.65 Interestingly, inactivating
B concentrations in infertile male subjects.53,54 As well, peri- mutations of the ESR1 gene impaired sperm viability and
natal exposures to dioxin decreased serum inhibin B and fertility66 and ESR1 gene deficiency was associated with
elevated FSH levels in male subjects.55 Prenatal exposures decreased fertility in transgenic mice.8
to BPA activated Raf1 and extracellular regulated kinase
Leydig cells
(ERK)1/2 in rat Sertoli cells56 and disrupted junctional bar-
riers in vivo and in vitro.57,58 Furthermore, BPA exposures Fetal Leydig cells arise from interstitial mesenchymal stem
were associated with redistribution of junctional proteins cells (MSCs) and disappear soon after birth with a small
such as occludin, zonula occludens 1, N-cadherin, β-catenin, remnant of cells carried over into the postnatal period.
or connexin 43 in the rat and human testes, which dis- Thus, it is conceivable that chemical exposure effects
rupted intercellular communication.57,59–62 Chemical dis- imprinted on MSCs early in development persist in adult
ruption of the blood-testis barrier (BTB) and ectoplasmic tissues and/or are passed in the germ line to subsequent
specializations (ES) in Sertoli cells likely renders the testes generations.67,68 The LH signaling pathway is the primary
more vulnerable to chemical exposure effects. regulator of Leydig cells, which enzymatically convert
the cholesterol substrate into androgen metabolites and
Germ cells androgen (Figure 19.4). Chemical-induced inhibition of
Germ cells begin differentiation as primordial germ cells T secretion may be receptor-mediated or occur by tran-
(PGCs) in the embryo. E2 and ESR agonists induced scriptional regulation of steroidogenic enzyme activity.

LH
Chol Gs Gi
esters Adenylate
cyclase

Cholesterol cAMP

Steroidogenic
PKA enzyme gene
transcription

StAR
DNA
PBR
mRNA

SER Nucleus

Mitochondrion

Testosterone

Figure 19.4  The LH-stimulated signaling pathway is the primary regulator of Leydig cells. LH binding of its cognate receptors causes
cyclic AMP production and protein kinase A (PKA) activation and recruitment of cholesterol as substrate for androgen biosynthesis.
Cholesterol (CHOL) is moved into mitochondria by transport proteins (PBR and StAR) for enzymatic cleavage into pregnenolone
which then crosses into the SER for further enzymatic action by other enzymes (3β-hydroxysteroid hydrogenase, cytochrome
P45017α-hydroxylase, and 17β-hydroxysteroid dehydrogenase). Finally, T diffuses into the seminiferous tubules or enter into blood
capillaries for circulation to peripheral tissues. LH, luteinizing hormone; PBR, peripheral benzodiazepine receptor; StAR, steroido-
genic acute regulatory protein; SER, smooth endoplasmic reticulum.
238  Effects of chemical exposures on testis cell-cell interactions and endocrine function

LH
BPA
Octylphenol
LH Dimethoate
receptor
N
T
StAR SER
17β-HSD
AR
ER P45017α Octylphenol
CHO 3β-HSD

PREG
M P450scc
HPTE
Dimethoate
Ligand

Figure 19.5  Endocrine disruptor (ED)-induced inhibition of Leydig cell testosterone biosynthesis. Both ER (ESRs) and ARs are
present in Leydig cells. Several steps in the androgen biosynthetic pathway are inhibited by EDs, which decrease steroidogenic
enzyme activity. HPTE (2,2-bis-[p-hydroxyphenyl]-trichloroethane) and dimethoate both reduced cholesterol side-chain cleavage
(P450scc) activity while octyphenol decreases P45017α activity. Gene transcription for the protein that causes movement of choles-
terol into mitochondria for side-chain cleavage, steroidogenic acute regulatory protein (StAR), was inhibited by dimethoate. Some
agents such as octylphenol interfered with luteinizing hormone (LH) binding of its receptor. AR, androgen receptor; CHO, choles-
terol; ER, estrogen receptors; M, mitochondria; N, nucleus; PREG, pregnenolone; SER, smooth endoplasmic reticulum; T, testosterone.
(See further Ann Med. 2001;33(6):391–403.)

For example,  2, 2-bis-(p-hydroxyphenyl)-trichloroethane act as autocrine or paracrine regulators (Table 19.2). It is


(HPTE) and dimethoate both decreased cholesterol side- likely that EDC effects on individual testicular cells alter
chain cleavage (P450scc) enzyme activity while octylphenol the patterns of cell-cell interactions necessary to optimize
decreased P45017α activity. Other chemicals interfered with testicular function.
LH binding to receptors in Leydig cells (e.g., octylphenol)
(Figure 19.5). Inactivation of the ESR gene in Leydig cells FSH
affected androgen biosynthesis,8 and males with complete Receptors for the gonadotropin FSH are present only in
androgen insensitivity syndrome due to a mutation in the Sertoli cells70,71 while inhibin B, produced by Sertoli cells,
AR gene exhibited female external and internal genita- provide feedback to the hypothalamus and pituitary gland
lia.69 Thus, decreased Leydig cell androgen secretion and to regulate FSH secretion. FSH and androgen appear to
disruption of AR-mediated activity both adversely affect be the primary factors controlling spermatogenesis in
reproductive development and function. the rodent.72–74 Inhibin B secretion appears to vary with
changing roles of Sertoli cells in the immature and adult
EDC DISRUPTION OF PARACRINE RELATIONSHIPS testes,75 and its levels are negatively correlated with those
BETWEEN TESTICULAR CELLS of FSH.76 Serum FSH concentrations were increased after
Testicular cells express receptors for pituitary gonadotro- treatment of male rats with aromatase inhibitors and in
pins FSH, LH, and steroid hormones and are therefore reg- ESR1- and aromatase-deficient male subjects.77 Prenatal
ulated by gonadotropins and steroid hormones. Testicular exposures of male rats to BPA and DEHP were associ-
cells also secrete growth factors and other cytokines that ated with increased LHβ and FSHβ protein expression in

Table 19.2  Paracrine regulators of testicular cells.


Hormone/Growth factor Source Target
AMH Sertoli cells Leydig cells, germ cells
Androgen Leydig cells Sertoli cells, peritubular myoid cells
Estrogen Sertoli cells, Leydig cells Germ cells
IGF-1 Sertoli cells Multiple tissues
Stem cell factor Sertoli cells Leydig cells, germ cells
Transforming growth factors (α and β) Sertoli cells Leydig cells, germ cells
Conclusion 239

the prepubertal period.51 Thus, EDC disruption of steroid GnRH neurons to control GnRH secretion.85 Deletion
hormone feedback regulation of the hypothalamus and of ARs from Sertoli cells caused meiotic arrest in germ
pituitary gland likely affects FSH secretion and germ cell cells,86,87 indicating that proliferation and maturation
development. of Sertoli cells is androgen-dependent. In the testis, T  is
bound to Sertoli cell-specific ABP, which provides high
Anti-Müllerian hormone (AMH) androgen concentrations necessary for spermatogenesis.88
AMH, a product of Sertoli cells, which is required to induce Thus, EDCs that target Sertoli cells, ABP expression, and/
mesonephric cell migration during mammalian testis for- or androgen secretion likely disrupt spermatogenesis.89
mation in the embryo, has receptors (AMHR2) in germ Moreover, maintenance of cell adhesions at the BTB
cells and Leydig cells.78,79 In the postnatal period, AMH (Sertoli-Sertoli junctions) and ES (Sertoli-spermatid junc-
concentrations were negatively correlated with testicular tions) is androgen-dependent.61,90–94 Exposure of male rats
and epididymal weight.80 Transgenic mice with deletion to BPA, CdCl2, and PFOS95 downregulated Cx43 expres-
of AMH or AMHR2 showed decreased testicular ste- sion in Sertoli cells and was associated with oxidative
roidogenic capacity while AMH overexpression increased stress-induced damage to junctional complexes and germ
serum T concentrations.81,82 In immature animals and cell apoptosis.52,93,96–98 Thus, chemical-induced changes in
in tfm mice, FSH administration increased AMH levels, androgen secretion may compromise BTB and ES integ-
suggesting FSH regulates AMH secretion.83 Interestingly, rity and thereby disrupt germ cell development.
prenatal exposure of male rats to BPA and DEHP was asso-
ciated with increased serum AMH levels in adulthood51 Estrogen
(Figure 19.6). As well, AMH expression was downregu- Similar to androgen, estrogen is critical to maintenance of
lated by development of meiotic germ cells while serum junctional complexes between testicular cells and orderly
AMH concentrations correlated with increased FSH and progression of spermatogenesis.99,100 Aro­ matization of
T levels and decreased inhibin B concentrations in male androgens to estrogens occurs in testicular cells that
subjects after administration of chemical toxicants (cis- express the cytochrome P450 aromatase enzyme.55,101
platin and busulfan).80,84 AMH may be a good candidate However, overexpression of aromatase and excessive E2
for development as a biomarker for impaired testicular exposure increased germ cell apoptosis and impaired
development and/or damage as reflected in serum levels spermatogenesis.102,103 Importantly, exposure of rodents
and due to lack of diurnal variations and relative ease of to several estrogenic compounds (e.g., BPA, dioxin, cad-
measurement. mium, and phthalates) interfered with estrogen homeo-
stasis in the testicular microenvironment and disrupted
LH and androgen germ cell development.104
Pituitary LH is the primary regulator of Leydig cells,
and circulating androgens act on ARs in hypothalamic Growth factors
Somatotropin or the growth hormone (GH), acting in
AMH concert with IGF-1, serves as a paracrine regulator of
ACTB testicular cells. While GH is produced by the pituitary
* 2.4 * gland, IGF-1 is secreted by Sertoli cells and Leydig cells
1.4
in response to FSH and LH stimulation in order to sup-
2.0
Serum AMH protein/ACTB
Serum AMH protein/ACTB

1.2 port both Sertoli cell maturation and testicular steroido-


1.0 1.6 genesis in an autocrine and paracrine manner.105 Exposure
0.8 of rats to dioxin decreased serum IGF-1 concentrations106
1.2
while PFOS inhibited IGF-1 secretion and downregulated
0.6
0.8 IGF-1R expression in the liver and testis.107 These observa-
0.4 tions imply that growth factor receptors (e.g., epidermal
0.2 0.4 growth factor receptors and IGF1-Rs) may act in concert
0.0 0.0
with other transcription factors to mediate EDC activity
0 25 0 50 in the testis.
(a) Dose of BPA (µg/kg bw) (b) Dose of DEHP (µg/kg bw)
CONCLUSION
Figure 19.6  Male rats were exposed to BPA and DEHP by Chemical exposures occurring in the fetal period are
maternal gavage from gestational day 12 to 21. Adult male linked to altered sexual differentiation that impact
rats at 90 days of age were processed to obtain serum (n = 10 gonadotropin subunit expression in the pituitary gland
animals per group) to measure AMH protein (a and b). Protein and gonadal secretion of AMH and steroid hormones
analysis was achieved using Western blotting procedures and (Figure 19.7). Because developing organisms are more
antisera specific to rat AMH. Assays per sample were repeated sensitive to EDCs, children and adolescents are especially
at least four times and protein levels were normalized to ACTB. vulnerable segments of the population. EDCs are widely
*, p < 0.05 vs control. (See further Endocrinology. 2015;156(12):​ distributed in the environment but only a few studies
4672–4685.) of mixed chemical exposure effects have been reported.
240  Effects of chemical exposures on testis cell-cell interactions and endocrine function

Germ cells

testosterone

Leydig cells
AMH
IGF-1
Stem Cell Factor
Transforming Growth Factors
(α & β) 17β-estradiol

testosterone

Sertoli cells

Figure 19.7  Paracrine regulation of testicular cells. Sertoli cells and Leydig cells produce factors which self-regulate (autocrine
regulation) or pass to other cells as paracrine regulators.

For example, exposures of male rats to a phthalate mix- 7. Senger PL. Pathways to Pregnancy and Parturition,
ture caused greater effects than those due to individual 2nd Ed. Redmond, Oregon: Current Conceptions,
chemicals.108 Thus, studies of single individual chemicals Inc.; 1997:373.
may mask biological effects that are present in the body. 8. Eddy EM, Washburn TF, Bunch DO et al. Targeted
Nevertheless, identification of molecular mechanisms disruption of the estrogen receptor gene in male
related to EDC effects in the male reproductive tract mice causes alteration of spermatogenesis and infer-
will identify signaling molecules and pathways mediat- tility. Endocrinology. 1996;137(11):4796–4805.
ing chemical exposure effects.109,110 Studies addressing 9. Mahato D, Goulding EH, Korach KS, Eddy
identified research gaps will generate data to support risk EM. Spermatogenic cells do not require  estro-
assessment of the population. gen receptor-alpha for development or function.
Endocrinology. 2000;141(3):1273–1276.
ACKNOWLEDGMENTS 10. van Pelt AM, de Rooij DG, van der Burg B, van der
This work was supported in part by funding from the Saag PT, Gustafsson JA, Kuiper GG. Ontogeny
Animal Health and Disease Research grant award from of estrogen receptor-beta expression in rat testis.
the College of Veterinary Medicine at Auburn University, Endocrinology. 1999;140(1):478–483.
and NIH grant ES 15886 (to BTA). 11. Kuiper GG, Gustafsson JA. The novel estrogen
receptor-beta subtype: Potential role in the cell- and
REFERENCES promoter-specific actions of estrogens and anti-
1. Boisen KA, Main KM, Rajpert-De Meyts E, estrogens. FEBS Lett. 1997;410(1):87–90.
Skakkebaek NE. Are male reproductive disorders a 12. Saunders PT, Maguire SM, Gaughan J, Millar MR.
common entity? The testicular dysgenesis syndrome. Expression of oestrogen receptor beta (ER beta) in
Ann N Y Acad Sci. 2001;948:90–99. multiple rat tissues visualised by immunohisto-
2. Pfaehler A, Nanjappa MK, Coleman ES et al. chemistry. J Endocrinol. 1997;154(3):R13–R16.
Regulation of adiponectin secretion by soy isofla- 13. Rosenfeld CS, Ganjam VK, Taylor JA et al.
vones has implication for endocrine function of the Transcription and translation of estrogen receptor-
testis. Toxicol Lett. 2012;209(1):78–85. beta in the male reproductive tract of estrogen
3. Heindel JJ. Endocrine disruptors and the obesity epi- receptor-alpha knock-out and wild-type mice. Endo­
demic. Toxicol Sci. 2003;76(2):247–249. crinology. 1998;139(6):2982–2987.
4. Brophy JT, Keith MM, Watterson A et al. Breast 14. Saunders PT, Fisher JS, Sharpe RM, Millar MR.
cancer risk in relation to occupations with expo- Expression of oestrogen receptor beta (ER beta) occurs
sure to carcinogens and endocrine disruptors: A in multiple cell types, including some germ cells, in
Canadian case-control study. Environ Health–Glob. the rat testis. J Endocrinol. 1998;156(3):​R13–R17.
2012;​11:87. 15. Brandenberger AW, Tee MK, Lee JY, Chao V, Jaffe
5. Kurzer MS, Xu X. Dietary phytoestrogens. Annu Rev RB. Tissue distribution of estrogen receptors alpha
Nutr. 1997;17:353–381. (ER-alpha) and beta (ER-beta) mRNA in the midg-
6. Akingbemi BT, Hardy MP. Oestrogenic and anti- estational human fetus. J Clin Endocrinol Metabol.
androgenic chemicals in the environment: Effects 1997;82(10):3509–3512.
on male reproductive health. Ann Med. 2001;33(6):​ 16. Pentikainen V, Erkkila K, Suomalainen L, Parvinen
391–403. M, Dunkel L. Estradiol acts as a germ cell survival
References 241

factor in the human testis in vitro. J Clin Endocrinol is mediated by antiandrogenic metabolites. Toxicol
Metabol. 2000;85(5):2057–2067. Appl Pharmacol. 1994;126(2):276–285.
17. Taylor AH, Al-Azzawi F. Immunolocalisation of 31. Gray LE Jr, Ostby J, Monosson E, Kelce WR.
oestrogen receptor beta in human tissues. J Mol Environmental antiandrogens: Low doses of the
Endocrinol. 2000;24(1):145–155. fungicide vinclozolin alter sexual differentiation of
18. Enmark E, Pelto-Huikko M, Grandien K et al. the male rat. Toxicol Ind Health. 1999;15(1–2):48–64.
Human estrogen receptor beta-gene structure, chro- 32. Bulger WH, Muccitelli RM, Kupfer D. Studies on
mosomal localization, and expression pattern. J Clin the in vivo and in vitro estrogenic activities of
Endocrinol Metabol. 1997;82(12):4258–4265. methoxychlor and its metabolites. Role of hepatic
19. Bourguiba S, Lambard S, Carreau S. Steroids con- mono-oxygenase in methoxychlor activation. Biochem
trol the aromatase gene expression in purified germ Pharmacol. 1978;27(20):2417–2423.
cells from the adult male rat. J Mol Endocrinol. 33. Akingbemi BT, Sottas CM, Koulova AI, Klinefelter
2003;31(1):83–94. GR, Hardy MP. Inhibition of testicular steroidogene-
20. Levine AC, Kirschenbaum A, Gabrilove JL. The role sis by the xenoestrogen bisphenol A is associated with
of sex steroids in the pathogenesis and maintenance reduced pituitary luteinizing hormone secretion and
of benign prostatic hyperplasia. Mt Sinai J Med. decreased steroidogenic enzyme gene expression in
1997;64(1):20–25. rat Leydig cells. Endocrinology. 2004;145(2):592–603.
21. Hofmann PJ, Schomburg L, Kohrle J. Interference 34. Akingbemi BT, Ge R, Klinefelter GR, Zirkin BR,
of endocrine disruptors with thyroid hormone Hardy MP. Phthalate-induced Leydig cell hyperplasia
receptor-dependent transactivation. Toxicol Sci. is associated with multiple endocrine disturbances.
2009;110(1):125–137. Proc Natl Acad Sci U S A. 2004;101(3):775–780.
22. Levin ER. Integration of the extranuclear and 35. Yen PM. Physiological and molecular basis of thyroid
nuclear actions of estrogen. Mol Endocrinol. 2005;​ hormone action. Physiol Rev. 2001;81(3):1097–1142.
19(8):1951–1959. 36. Jugan M-L, Levi Y, Blondeau J-P. Endocrine dis-
23. Nadal A, Ropero AB, Laribi O, Maillet M, Fuentes ruptors and thyroid hormone physiology. Biochem
E, Soria B. Nongenomic actions of estrogens and Pharmacol. 2010:939–947.
xenoestrogens by binding at a plasma membrane 37. Crofton KM, Craft ES, Hedge et al. JM. Thyroid-
receptor unrelated to estrogen receptor a and estro- hormone-disrupting chemicals: Evidence for dose-
gen receptor b. Proc Natl Acad Sci U S A. 2000;​ dependent additivity or synergism. Environ Health
97(21):11603–11608. Perspect. 2005;113(11):1549–1554.
24. Kuiper GG, Carlsson BO, Grandien KAJ et al. 38. Kodavanti PRS, Curras-Collazo MC. Neuroendo­
Comparison of the ligand binding specificity and crine actions of organohalogens: Thyroid hormones,
transcript tissue distribution of estrogen receptor arginine vasopressin, and neuroplasticity. Front
alpha and beta. Endocrinology. 1997;139:863–870. Neuroendocrinol. 2010;31(4):479–496.
25. Thomas P, Dong J. Binding and activation of the 39. Collins Jr WT, Capen CC. Fine structural lesions and
seven-transmembrane estrogen receptor GPR30 by hormonal alterations in thyroid glands of perinatal
environmental estrogens: A potential novel mecha- rats exposed in utero and by the milk to polychlori-
nism of endocrine disruption. J Steroid Biochem Mol nated biphenyls. Am J Pathol. 1980;99(1):125–141.
Biol. 2006;102:175–179. 40. van Birgelen APJM, Smit EA, Kampen IM et al.
26. Carreau S, Bouraima-Lelong H, Delalande C. Estrogens Subchronic effects of 2,3,7,8-TCDD or PCBs on thy-
in male germ cells. Spermatogenesis. 2011;​1(2):90–94. roid hormone metabolism: Use in risk assessment.
27. Wang L, Hao J, Hu J et al. Protective effects of ginsen- Eur J Pharmacol. 1995;293(1):77–85.
osides against Bisphenol A-induced cytotoxicity in 41. Chauhan KR, Kodavanti PRS, McKinney JD. Assess­
15P-1 Sertoli cells via extracellular signal-regulated ing the role of ortho-substitution on polychlorinated
kinase 1/2 signalling and antioxidant mechanisms. ­biphenyl binding to transthyretin, a thyroxine transport
Basic Clin Pharmacol Toxicol. 2012;111(1):42–49. ­protein. Toxicol Appl Pharmacol. 2000;162(1):10–21.
28. Camacho L, Basavarajappa MS, Chang CW et al. Effects 42. Lans MC, Klasson-Wehler E, Willemsen M, Meussen
of oral exposure to bisphenol A on gene expression E, Safe S, Brouwer A. Structure-dependent, com-
and global genomic DNA methylation in the prostate, petitive interaction of hydroxy-polychlorobiphenyls,
female mammary gland, and uterus of NCTR Sprague- -dibenzo-p-dioxins and -dibenzofurans with human
Dawley rats. Food Chem Toxicol. 2015;81:92–103. transthyretin. Chem Biol Interact. 1993;88(1):7–21.
29. Merlet J, Racine C, Moreau E, Moreno SG, Habert R. 43. Manna PR, Roy P, Clark BJ, Stocco DM, Huhtaniemi
Male fetal germ cells are targets for androgens that IT. Interaction of thyroid hormone and steroidogenic
physiologically inhibit their proliferation. Proc Natl acute regulatory (StAR) protein in the regulation of
Acad Sci U S A. 2007;104(9):3615–3620. murine Leydig cell steroidogenesis. J Steroid Biochem
30. Kelce WR, Monosson E, Gamcsik MP, Laws SC, Mol Biol. 2001;76(1–5):167–177.
Gray LE Jr. Environmental hormone disruptors: 44. Meeker JD, Ferguson KK. Relationship between
Evidence that vinclozolin developmental toxicity urinary phthalate and bisphenol A concentrations
242  Effects of chemical exposures on testis cell-cell interactions and endocrine function

and serum thyroid measures in U.S. adults and 58. Izumi Y, Yamaguchi K, Ishikawa T et al. Molecular
adolescents from the National Health and Nutrition changes induced by bisphenol-A in rat Sertoli cell
Examination Survey (NHANES) 2007–2008. Environ culture. Syst Biol Reprod Med. 2011;57(5):228–232.
Health Perspect. 2011;119(10):1396–1402. 59. Li MW, Mruk DD, Lee WM, Cheng CY. Disruption
45. Shupnik MA. Gonadotropin gene modulation by of the blood-testis barrier integrity by bisphenol A
steroids and gonadotropin-releasing hormone. Biol in vitro: Is this a suitable model for studying blood-
Reprod. 1996;54(2):279–286. testis barrier dynamics? Int J Biochem Cell Biol.
46. Gregg DW, Schwall RH, Nett TM. Regulation of 2009;41(11):2302–2314.
gonadotropin secretion and number of gonadotropin- 60. Xiao X, Mruk DD, Tang EI et al. Environmental
releasing hormone receptors by inhibin, activin-A, toxicants perturb human Sertoli cell adhesive func-
and estradiol. Biol Reprod. 1991;44(4):725–732. tion via changes in F-actin organization medi-
47. Bagatell CJ, Dahl KD, Bremner WJ. The direct pitu- ated by actin regulatory proteins. Hum Reprod.
itary effect of testosterone to inhibit gonadotropin 2014;29(6):1279–1291.
secretion in men is partially mediated by aromatiza- 61. Cheng CY, Wong EW, Lie PP et al. Environmental
tion to estradiol. J Androl. 1994;15(1):15–21. toxicants and male reproductive function. Sperma­
48. Carani C, Qin K, Simoni M et al. Effect of testos- togenesis. 2011;1(1):2–13.
terone and estradiol in a man with aromatase defi- 62. Li MW, Mruk DD, Lee WM, Cheng CY. Connexin
ciency. N Engl J Med. 1997;337(2):91–95. 43 and plakophilin-2 as a protein complex that regu-
49. Khurana S1 RS, Ben-Jonathan N. Exposure of new- lates blood-testis barrier dynamics. Proc Natl Acad
born male and female rats to environmental estro- Sci U S A. 2009;106(25):10213–10218.
gens: Delayed and sustained hyperprolactinemia 63. Brieno-Enriquez MA, Garcia-Lopez J, Cardenas DB
and alterations in estrogen receptor expression. et al. Exposure to endocrine disruptor induces trans-
Endocrinology. 2000;141(12):4512–4517. generational epigenetic deregulation of microRNAs in
50. Marques P, Skorupskaite K, Rozario KS, Anderson primordial germ cells. PloS One. 2015;10(4):​e0124296.
RA, George JT. Physiology of Gnrh and gonadotro- 64. Urriola-Muñoz P, Lagos-Cabré R, Moreno, RD. A
pin secretion. In: De Groot LJ, Chrousos G, Dungan mechanism of male germ cell apoptosis induced by
K et al., eds. Endotext. South Dartmouth, MA: bisphenol-A and nonylphenol involving ADAM17
MDText.com, Inc.; 2000. and p38 MAPK activation. PLoS One. 2014;9(12):​
51. Abdel-Maksoud FM, Leasor KR, Butzen K, Braden e113793 9.
TD, Akingbemi BT. Prenatal exposures of male rats to 65. Brieno-Enriquez MA, Larriba E, Del Mazo J.
the environmental chemicals bisphenol A and Di(2- Endocrine disrupters, microRNAs, and primor-
ethylhexyl) phthalate impact the sexual differentia- dial germ cells: A dangerous cocktail. Fertil Steril.
tion process. Endocrinology. 2015;156(12):​4672–4683. 2016;106(4):871–879.
52. Wan HT, Mruk DD, Wong CK, Cheng CY. Targeting 66. Smith EP, Boyd J, Frank GR et al. Estrogen resistance
testis-specific proteins to inhibit spermatogenesis: caused by a mutation in the estrogen-receptor gene
Lesson from endocrine disrupting chemicals. Expert in a man. N Engl J Med. 1994;331(16):1056–1061.
Opin Ther Targets. 2013;17(7):839–855. 67. Benton L, Shan LX, Hardy MP. Differentiation
53. Meeker JD, Godfrey-Bailey L, Hauser R. Relationships of adult Leydig cells. J Steroid Biochem Mol Biol.
between serum hormone levels and semen qual- 1995;53(1–6):61–68.
ity among men from an infertility clinic. J Androl. 68. Nanjappa MK, Simon L, Akingbemi BT. The indus-
2007;28(3):397–406. trial chemical bisphenol A (BPA) interferes with
54. Meeker JD, Calafat AM, Hauser R. Urinary bisphe- proliferative activity and development of steroido-
nol A concentrations in relation to serum thyroid and genic capacity in rat Leydig cells. Biol Reprod.
reproductive hormone levels in men from an infertil- 2012;86(5):135, 1–12.
ity clinic. Environ Sci Technol. 2010;44(4):​1458–1463. 69. Patterson MN, Hughes IA, Gottlieb B, Pinsky L. The
55. Mocarelli P, Gerthoux PM, Needham LL et al. androgen receptor gene mutations database. Nucleic
Perinatal exposure to low doses of dioxin can perma- Acids Res. 1994;22(17):3560–3562.
nently impair human semen quality. Environ Health 70. Griswold MD, Heckert L, Linder C. The molecular
Perspect. 2011;119(5):713–718. biology of the FSH receptor. J Steroid Biochem molec-
56. Thuillier R, Manku G, Wang Y, Culty M. Changes in ular Biol. 1995;53(1–6):215–218.
MAPK pathway in neonatal and adult testis follow- 71. Menon KM, Menon B. Structure, function and regu-
ing fetal estrogen exposure and effects on rat testicu- lation of gonadotropin receptors – a perspective. Mol
lar cells. Microsc Res Tech. 2009;72(11):773–786. Cell Endocrinol. 2012;356(1–2):88–97.
57. Li MW, Mruk DD, Lee WM, Cheng CY. Connexin 72. Jorgensen N, Liu F, Andersson AM et al. Serum
43 is critical to maintain the homeostasis of the inhibin-b in fertile men is strongly correlated with
blood-testis barrier via its effects on tight junction low but not high sperm counts: A coordinated study
reassembly. Proc Natl Acad Sci U S A. 2010;107(42):​ of 1,797 European and US men. Fertil Steril. 2010;​
17998–18003. 94(6):2128–2134.
References 243

73. McNeilly AS. Diagnostic applications for inhibin 88. Grover A, Smith CE, Gregory M, Cyr DG, Sairam
and activins. Mol Cell Endocrinol. 2012;359(1–2):​ MR, Hermo L. Effects of FSH receptor deletion on
121–125. epididymal tubules and sperm morphology, num-
74. Chang H, Brown CW, Matzuk MM. Genetic analysis bers, and motility. Mol Reprod Dev. 2005;72(2):​
of the mammalian transforming growth factor-beta 135–144.
superfamily. Endocr Rev. 2002;23(6):787–823. 89. Xi W, Wan HT, Zhao YG, Wong MH, Giesy JP, Wong
75. Meachem SJ, Nieschlag E, Simoni M. Inhibin B in CK. Effects of perinatal exposure to bisphenol A
male reproduction: Pathophysiology and clinical rel- and di(2-ethylhexyl)-phthalate on gonadal devel-
evance. Eur J Endocrinol. 2001;145(5):561–571. opment of male mice. Environ Sci Pollut Res Int.
76. O’Connor AE, De Kretser DM. Inhibins in normal 2011;19(7):2515–2527.
male physiology. Semin Reprod Med. 2004;22(3):​ 90. Cheng J, Watkins SC, Walker WH. Testosterone
177–185. activates mitogen-activated protein kinase via Src
77. Szymanski L, Bakker J. Aromatase knockout mice show kinase and the epidermal growth factor recep-
normal steroid-induced activation of gonadotrophin- tor in Sertoli cells. Endocrinology. 2007;148(5):​
releasing hormone neurones and luteinising hormone 2066–2074.
surges with a reduced population of kisspeptin neu- 91. Wang RS, Yeh S, Tzeng CR, Chang C. Androgen
rones in the rostral hypothalamus. J Neuroendocrinol. receptor roles in spermatogenesis and fertility:
2012;24(9):1222–1233. Lessons from testicular cell-specific androgen recep-
78. Ross AJ, Tilman C, Yao H, MacLaughlin D, Capel B. tor knockout mice. Endocr Rev. 2009;30(2):119–132.
AMH induces mesonephric cell migration in XX 92. Shupe J, Cheng J, Puri P, Kostereva N, Walker WH.
gonads. Mol Cell Endocrinol. 2003;211(1–2):1–7. Regulation of Sertoli-germ cell adhesion and sperm
79. Nakamura S, Watakabe I, Nishimura T et al. release by FSH and nonclassical testosterone signal-
Hyperproliferation of mitotically active germ cells ing. Mol Endocrinol. 2011;25(2):238–252.
due to defective anti-Mullerian hormone signal- 93. Siu ER, Mruk DD, Porto CS, Cheng CY. Cadmium-
ing mediates sex reversal in medaka. Development. induced testicular injury. Toxicol Appl Pharmacol.
2012;139(13):2283–2287. 2009;238(3):240–249.
80. Levi M, Hasky N, Stemmer SM, Shalgi R, Ben- 94. O’Donnell L, Stanton PG, Bartles JR, Robertson DM.
Aharon I. Anti-Mullerian hormone is a marker for Sertoli cell ectoplasmic specializations in the semi-
chemotherapy-induced testicular toxicity. Endo­ niferous epithelium of the testosterone-suppressed
crinology. 2015;156(10):3818–3827. adult rat. Biol Reprod. 2000;63(1):99–108.
81. Behringer RR, Finegold MJ, Cate RL. Mullerian- 95. Gao Y, Mruk DD, Cheng CY. Sertoli cells are the
inhibiting substance function during mammalian target of environmental toxicants in the testis—A
sexual development. Cell. 1994;79(3):415–425. mechanistic and therapeutic insight. Expert Opin
82. Fynn-Thompson E, Cheng H, Teixeira J. Inhibition Ther Targets. 2015;19(8):1073–1090.
of steroidogenesis in Leydig cells by Mullerian- 96. Li MW, Mruk DD, Cheng CY. Gap junctions
inhibiting substance. Mol Cell Endocrinol. 2003;211​ and blood-tissue barriers. Adv Exp Med Biol.
(1–2):99–104. 2012;763:260–280.
83. Al-Attar L, Noel K, Dutertre M et al. Hormonal 97. Mruk DD, Cheng CY. Environmental contaminants:
and cellular regulation of Sertoli cell anti-Mullerian Is male reproductive health at risk? Spermatogenesis.
hormone production in the postnatal mouse. J Clin 2011;1(4):283–290.
Invest. 1997;100(6):1335–1343. 98. Pointis G, Gilleron J, Carette D, Segretain D.
84. Matuszczak E, Hermanowicz A, Komarowska M, Testicular connexin 43, a precocious molecular tar-
Debek W. Serum AMH in physiology and pathology get for the effect of environmental toxicants on male
of male gonads. Int J Endocrinol. 2013;2013. fertility. Spermatogenesis. 2011;1(4):303–317.
85. Shakil T, Hoque AN, Husain M, Belsham DD. 99. O’Donnell L, Meachem SJ, Stanton PG et al. Endocrine
Differential regulation of gonadotropin-releasing Regulation of Spermatogenesis. 3 ed. Neill JD, editor.
hormone secretion and gene expression by andro- Amsterdam: Elsevier; 2006.
gen: Membrane versus nuclear receptor activation. 100. O’Donnell L, Robertson KM, Jones ME, Simpson ER.
Mol Endocrinol. 2002;16(11):2592–2602. Estrogen and spermatogenesis. Endocr Rev. 2001;22(3):​
86. De Gendt K, Swinnen JV, Saunders PT et al. A Sertoli 289–318.
cell-selective knockout of the androgen receptor 101. Carreau S, Delalande C, Galeraud-Denis I. Mammalian
causes spermatogenic arrest in meiosis. Proc Natl sperm quality and aromatase expression. Microsc Res
Acad Sci U S A. 2004;101(5):1327–1332. Techn. 2009;72(8):552–557.
87. Chang C, Chen YT, Yeh SD et al. Infertility with 102. Assinder S, Davis R, Fenwick M, Glover A. Adult-
defective spermatogenesis and hypotestosteronemia only exposure of male rats to a diet of high phy-
in male mice lacking the androgen receptor in toestrogen content increases apoptosis of meiotic
Sertoli cells. Proc Natl Acad Sci U S A. 2004;101(18):​ and post-meiotic germ cells. Reproduction. 2007;​
6876–6881. 133(1):11–19.
244  Effects of chemical exposures on testis cell-cell interactions and endocrine function

103. Li X, Rahman N. Impact of androgen/estrogen ratio: 107. Wan HT, Zhao YG, Wong MH et al. Testicular sig-
Lessons learned from the aromatase over-expression naling is the potential target of perfluorooctane-
mice. Gen Comp Endocrinol. 2008;159(1):1–9. sulfonate-mediated subfertility in male mice. Biol
104. Yeung BH, Wan HT, Law AY, Wong CK. Endocrine Reprod. 2011;84(5):1016–1023.
disrupting chemicals: Multiple effects on testicular 108. Howdeshell KL, Wilson VS, Furr J et al. A mixture of
signaling and spermatogenesis. Spermatogenesis. five phthalate esters inhibits fetal testicular testosterone
2011;1(3):231–239. production in the Sprague-Dawley rat in a cumulative,
105. Chandrashekar V, Zaczek D, Bartke A. The conse- dose-additive manner. Toxicol Sci. 2008;105(1):153–165.
quences of altered somatotropic system on reproduc- 109. vom Saal FS, Akingbemi BT, Belcher SM et al. Flawed
tion. Biol Reprod. 2004;71(1):17–27. experimental design reveals the need for guidelines
106. Croutch CR, Lebofsky M, Schramm KW, Terranova requiring appropriate positive controls in endocrine
PF, Rozman KK. 2,3,7,8-Tetrachlorodibenzo-p- disruption research. Toxicol Sci. 2010;115(2):612–613;
dioxin (TCDD) and 1,2,3,4,7,8-hexachlorodibenzo- author reply 4–20.
p-dioxin (HxCDD) alter body weight by decreasing 110. Martina CA, Weiss B, Swan SH. Lifestyle behaviors
insulin-like growth factor I (IGF-I) signaling. Toxicol associated with exposures to endocrine disruptors.
Sci. 2005;85(1):560–571. Neurotoxicology. 2012;33(6):1427–1433.
Environmental toxicants on Leydig
cell function
20
LEPING YE, XIAOHENG LI, XIAOMIN CHEN, QINGQUAN LIAN, and REN-SHAN GE

INTRODUCTION organophosphates, azole compounds, 1, 2-dibromo-3-​


Leydig cells reside in the interstitium of the testis and make chloropropane, heavy metals, and plant toxicants have
androgens and peptide hormones for the male reproduc- been observed to disrupt Leydig cell functions. These
tive system. In rodents, there are two different populations compounds may target Leydig cells, acting on multiple
of Leydig cells: fetal Leydig cells and adult Leydig cells. levels to disrupt the normal physiological functions of
The primary function of Leydig cells is to make androgens Leydig cells via blocking their development, downregulat-
(mainly testosterone and dihydrotestosterone). During the ing the expression levels of the genes for the biosynthesis
fetal period, testosterone and its more potent 5α-reduced of hormones (androgen and insulin-like 3), and/or directly
metabolite dihydrotestosterone are very important for ini- inhibiting androgen biosynthetic and metabolizing
tiating the development of the male reproductive tract and enzyme activities, and/or inducing Leydig cell apoptosis.
for inducing testis descent.1 The disruption of the andro- In this chapter, we discuss some of these environmental
gen biosynthesis in fetal Leydig cells causes various types toxicants on Leydig cells.
of malformity, including male-to-female sex reversal,
cryptorchidism, and hypospadias. During the postnatal ANDROGEN BIOSYNTHESIS
period, androgens are critical for initiating pubertal devel- In adult Leydig cells, at least four enzymes are involved
opment in male adolescents, maintaining the accessory in testosterone synthesis. Biosynthesis starts with the
sex organs,2 stimulating spermatogenesis,3 and promot- substrate cholesterol. The first enzyme is P450 cyto­chrome
ing sexual behavior in adults.4 Although adult Leydig cells cholesterol side chain cleavage enzyme (CYP11A1). This
only account for about 1%–5% of all testicular cell types, enzyme resides in the inner membrane of the mitochon-
they make up about 95% of the total amount of testoster- dria of the Leydig cells.13 The enzyme catalyzes choles-
one in the body.5 terol into pregnenolone. Pregnenolone diffuses from the
Besides the production of androgens, Leydig cells also mitochondria into outside cellular milieu, which are full
secrete insulin-like 3, a peptide hormone. Insulin-like 3, of smooth endoplasmic reticulum. Three steroidogenic
secreted by rodent fetal Leydig cells, is critical for the ini- enzymes reside in the smooth endoplasmic reticulum: 3β-​
tial testis descent.6,7 Insulin-like 3 (encoded by Insl3) pro- hydroxysteroid dehydrogenase (3β-HSD), cytochrome
duced by adult Leydig cells plays a role in spermatogenesis.8 P450 17α-hydroxylase/17,20-lyase (CYP17A1), and 17β-​
Interestingly, primates, including humans, have a third hydroxysteroid dehydrogenase 3 (17β-HSD3).14 Preg­
generation of Leydig cells, called neonatal Leydig cells, nenolone is finally converted into testosterone by the
which exist in the testis for a very short time from birth sequential reactions of these enzymes. Because there are
to 6 months.9 However, their functions are still not clear, differences in the affinity of the steroidogenic enzymes
and therefore, we only discuss fetal and adult Leydig cells between human and rodents, the catalytic reactions go
in this chapter. down different pathways (Δ4 or Δ5 pathway) with dif-
Many environmental chemicals can affect the develop- ferent steroid intermediates (Figure 20.1). Rodents
ment of both generations of Leydig cells and their func- predominantly take the Δ4 pathway: pregnenolone →
tions. They can disrupt the production of androgens and progesterone → 17α-hydroxyprogesterone → andro-
insulin-like 3. Because there are increased uses of indus- stenedione → testosterone.14 Humans mainly take the Δ5
trial compounds (some of which are potential Leydig cell pathway: pregnenolone → 17α-hydroxypregnenolone  →
toxicants) in modern society, increasing incidences of dehydroepiandrosterone → androstenedione → testoster-
cryptorchidism, hypospadias, testicular cancers, and male one.14 However, fetal Leydig cells in mice lack 17β -HSD3,
infertility over the past 40 years has been noted.10,11 The which is expressed in Sertoli cells. Therefore, fetal Leydig
association of many environmental toxicants with the mal- cells make androstenedione, which is transferred to
formity of male reproductive tracts, testicular cancer, and Sertoli cells where testosterone is synthesized by 17β-
male infertility has been reported.12 These environmental HSD3.15 The Leydig cell precursors in the adult Leydig cell
chemicals can disrupt the normal male reproductive sys- population also contain a significantly high level of type
tem in the experimental animal (mainly rat) models. 1 5α-reductase, and thus 5α-reduced androgen is made in
Several classes of compounds, including perfluoroalkyl these Leydig cells.16
substances, phthalates, bisphenols, benzophenones, organo- When testosterone is produced, it is metabolized
chlorines, carbamates, polycyclic aromatic hydrocarbon, to dihydrotestosterone, a more potent androgen than

245
246  Environmental toxicants on Leydig cell function

testosterone, in Leydig cells or peripheral tissues by sev- catalyzed by this enzyme occurs in the inner membrane
eral types of 5α-reductase (SRD5A1, 2 and 3). Of three of the mitochondria. Two hydroxyl groups are added
isoforms of 5α-reductase, SRD5A2 has a high affinity for to cholesterol (at C20 and C22) first. Then, the cleavage
testosterone and is particularly important for the genera- between the added hydroxyl groups is followed, leading
tion of dihydrotestosterone.17 Dihydrotestosterone is criti- to the formation of pregnenolone.14 CYP11A1 is the first
cal for the development of male reproductive tracts during enzyme for the formation of all steroids and it expresses
the fetal period.18,19 in adrenals, gonads (including Leydig cells), and placen-
tas. Thus, the disruption of CYP11A1 not only impairs
ENZYMES FOR TESTOSTERONE BIOSYNTHESIS Leydig cell functions but also the functions of other
AND METABOLISM endocrine systems.
CYP11A1 3β-HSD
CYP11A1 is encoded by human CYP11A1 or rat Cyp11a1.
20 21 There are several isoforms of 3β-HSD, each encoded by
Thus far, no additional isoforms have been found for this a different gene. In humans, there are two isoforms of
enzyme. 3β-HSD (3β-HSD1 and 3β-HSD2). Human 3β-HSD1 and
CYP11A1 is a complex enzyme, requiring NADPH as 3β-HSD2 are encoded by human HSD3B1 and HSD3B2
the cofactor to provide the electrons (Figure 20.2). The (review in Simard et al.24), respectively. Human HSD3B1
electrons are transferred from NADPH to CYP11A1 via and HSD3B2 genes have 81.9% identity. Human HSD3B1
two electron transfer proteins: adrenodoxin reductase is primarily expressed in the placenta. Human HSD3B2 is
and adrenodoxin.22,23 All three proteins constitute the predominantly expressed in adrenals and gonads (includ-
cholesterol side-chain cleavage complex.23 The complex ing Leydig cells). Therefore, in this chapter, we discuss
catalyzes three sequential reactions using three mol- human HSD3B2 and its encoded protein 3β-HSD2. In
ecules of cofactor NADPH for electron transfers via the rats, there are four isoforms of 3β-HSD.14 Each isoform
mitochondrial electron transfer system.14 The reaction is encoded by a distinct gene.14 In rat Leydig cells, the

LH/hCG
HDL
SCARB1 LHCGR

Cholesterol
Triclosan
cAMP cAMP
StAR Chlorpyrifos

Cholesterol MXC
CYP11A1 Lindane
Etomidate
Pregnenolone Gossypol

Pregnenolone

3β-HSD

Progesterone Androstenedione Testosterone


Tributyltin
CYP17A1 17β-HSD3 SRD5A
Triphenyltin
Genistein
Dihydrotestosterone

PFASs Bisphenol A PFASs


Phthalates Prochloraz Phthalates
Bisphenol A Triphenyltin Bisphenol A
MXC Tributyltin Etomidate
Etomidate DBCP Triphenyltin
Prochloraz Gossypol Gossypol
Tributyltin MXC
Gossypol Genistein

Figure 20.1  The steroidogenic pathway in rodent Leydig cells and the possible inhibitory sites of the environmental toxicants.
Hormones: LH, luteinizing hormone; hCG, human choriogonadotropin. Receptor: LHCHR, luteinizing hormone/choriogonado-
tropin receptor. Signaling: cAMP, cyclic adenosine monophosphate. Cholesterol transporters: SCARB1, scavenger receptor class B
member 1; StAR, steroidogenic acute regulatory protein; Enzymes: CYP11A1, P450 cholesterol side chain cleavage enzyme; 3β-HSD,
3β-hydroxysteroid dehydrogenase; CYP17A1, P450 17α-hydroxylase/17.20-lyase; 17β-HSD3, 17β-hydroxysteroid dehydrogenase 3;
SRD5A1, 5α-reductase.
Cell signaling for testosterone biosynthesis in Leydig cells  247

predominant isoform is 3β-HSD1, which is encoded by


Hsd3b1.25
H
Human 3β-HSD2 primarily catalyzes the conversion
of dehydroepiandrosterone to androstenedione, while
H
rat 3β-HSD1 predominantly catalyzes pregnenolone to
progesterone in the Leydig cells. Both require the cofac-
H H
tor NAD+. The catalysis by 3β-HSD undergoes two steps
(Figure 20.1). The first step of the enzyme is to catalyze (a) HO
dehydrogenation and is the rate-limiting. The second step
is to isomerize steroid molecule. 3β-HSD is critical for the O
biosynthesis of all biologically active steroids in adrenals, H
gonads (including Leydig cells), and placentas.26 Thus, the
disruption of 3β-HSD not only impairs Leydig cell func- H
tions but also the functions of other endocrine systems.
Patients having the HSD3B2 mutation27,28 cannot make H H
testosterone, thus leading to male-to-female sex reversal HO
(b)
or a partial feminization.

CYP17A1 Figure 20.2  (a) Chemical structures of cholesterol and


CYP17A1 is encoded by CYP17A1 (human)29 or Cyp17a1 (b) pregnenolone.
(rat).30 Thus far, no additional CYP17A1 isoforms have
been identified.
CYP17A1 is a P450 cytochrome enzyme. It is located HSD17B3 causes various phenotypes, including pseudo-
in the smooth endoplasmic reticulum. CYP17A1 cataly- hermaphroditism, with very low circulating testosterone
sis depends on cytochrome P450 oxidoreductase, cyto- in males.37,38
chrome b5, and phosphorylation.31 Cytochrome b5 acts
SRD5A
to facilitate CYP17A1 17, 20-lyase. Human CYP17A1 is
phosphorylated on serine and threonine residues by a Three distinct isoforms of SRD5A in both humans and
cAMP-dependent protein kinase and the phosphorylated rats have been characterized. Human genes encode type
CYP17A1 has increased 17,20-lyase activity.31 Human 1 (SRD5A1),39 2 (SRD5A2),40 and 3 (SRD5A3).41 Rat genes
CYP17A1 requires cytochrome P450 oxidoreductase for encode the isoforms of SRD5A with a similar designation
the transfer of electrons. Both human and rat CYP17A1 (Srd5a1, Srd5a2, and Srd5a3).
enzymes catalyze two steps of reaction: 17α-hydroxylation SRD5A catalyzes the activation of testosterone into a
and 20-lyation. Each reaction requires cofactor NADPH.14 more potent androgen, dihydrotestosterone (Figure 20.1).42–44
Human CYP17A1 primarily catalyzes pregnenolone to These enzymes use NADPH as a cofactor to transfer elec-
17-hydroxyprogenolone and then to dehydroepiandros- trons.16,45 The reaction by SRD5A involves an irreversible
terone (Figure 20.1).32 However, rat CYP17A1 primarily breakage of the double bond between Δ4, 5  carbons with
catalyzes progesterone into 17-hydroxyprogesterone and the facilitation of NADPH.
then to androstenedione.33 No clear mutation of human SRD5A1 has been found to
A mutation of human CYP17A1 alters the steroidogen- be associated with human diseases. The human SRD5A2
esis in the males, leading to defective masculinization that mutation is associated with male pseudohermaphrodit-
can range from partial to complete pseudohermaphrodit- ism.46 Human SRD5A3 encodes a protein that not only
ism and breast enlargement.34,35 catalyzes testosterone into dihydrotestosterone but also
converts polyprenol to dolichol.41 The human SRD5A3
17β-HSD3 mutation causes a congenital glycosylation disorder but
There are 14 different 17β-hydroxysteroid dehydrogenase does not affect reproduction.44
isoforms.25 Different isoforms of 17β-HSD are encoded by
different genes. The only isoform involved in testosterone CELL SIGNALING FOR TESTOSTERONE BIOSYNTHESIS
production in Leydig cells is 17β-hydroxysteroid dehydro- IN LEYDIG CELLS
genase isoform 3 (17β-HSD3), which is encoded by human Steroidogenesis in Leydig cells is regulated by lutein-
HSD17B336 or rat Hsd17b3.16 izing hormone (LH), which is secreted by the pituitary.
17β-HSD3 is located in the smooth endoplasmic reticu- LH reaches Leydig cells via circulation and it binds to
lum in Leydig cells. It catalyzes the final step of androgen the LH receptor (LHCGR, encoded by human LHCGR
biosynthesis, converting androstenedione into testos- or rat Lhcgr) on the surface of Leydig cells. This receptor
terone (Figure 20.1).16 17β-HSD3 requires NADPH as its is mainly coupled to adenylate cyclase pathways.47 After
cofactor for the electron transfer. the LHCGR is bound by LH, the receptor causes a rapid
The production of testosterone is considered as the increase of intracellular cAMP level. The increased cAMP
end-product in adult Leydig cells. The mutation of human level induces a series of downstream signaling to increase
248  Environmental toxicants on Leydig cell function

the expression levels of cholesterol-transporting proteins biosynthesis-related genes and insulin-like 3, or directly
and steroidogenic enzymes (Figure 20.1). inhibit testosterone biosynthetic and metabolizing enzyme
One cholesterol-transporting protein is scavenger recep- activities (Figure 20.1). Disrupting one or more steps in
tor class B member 1 (SCARB1, also known as SR-BI) Leydig cell development and steroidogenesis can poten-
(Figure 20.1). SCARB1 is encoded by the human SCARB1 tially cause lower testosterone and/or insulin-like 3 levels.64
gene48 or rat Scarb1 gene.49 In rat testis, SCARB1 is primarily Table 20.1 lists environmental toxicants that block Leydig
expressed by Leydig cells.50 SCARB1 is a receptor for high- cell development and/or downregulate the expression lev-
density lipoprotein, and is a 509-amino acid protein with els of testosterone biosynthesis- and metabolism-related
two cytoplasmic C- and N-terminal domains separated by genes and insulin-3, and/or induce Leydig cell apopto-
a large extracellular domain. SCARB1 activity depends on sis. Table 20.2 lists environmental toxicants that directly
lipoprotein binding to its extracellular domain and subse- inhibit testosterone-biosynthetic enzyme activities. Table
quent lipid exchange at the plasma membrane to transport 20.3 lists the toxicants that directly inhibit steroid metabo-
cholesterol from the extracellular location into the cytosol.51 lizing enzyme activities. The spectrum of environmental
The second cholesterol-transporting protein is the ste- toxicants on Leydig cells has been expanded to many cat-
roidogenic acute regulatory protein (StAR) (Figure 20.1). egories of compounds, which include industrial materials
StAR is encoded by human STAR and rat Star.52 StAR (perfluoroalkyl substances [PFASs], phthalates, bisphe-
mobilizes cytosolic cholesterol into the inner membrane nols, benzophenones, organochlorines, carbamates, poly-
of the mitochondria. The mechanism by which StAR helps cyclic aromatic hydrocarbon, organophosphates, azole
cholesterol movement remains unclear but possible mech- compounds, 1,2-dibromo-3-chloropropane [DBCP]), heavy
anisms are as a cholesterol-transporting shuttle53 or by metals, and plant toxicants.
binding to cholesterol.54 Star is the rate-limiting step for
androgen production in Leydig cells, and thus the protein PFASs
is critical for androgen production. PFASs are widely used for processing industrial and con-
Adrenals and Leydig cells share many common sumer products as coatings of textiles, paper, and uphol-
enzymes, including CYP11A1 and 3β-HSD. These two stery and as reaction additives in various industrial
compartments may interact with each other. The exces- processes.65–67 The molecules of PFASs contain carbon and
sive glucocorticoids secreted by adrenal cortex can inhibit fluorine atoms. Some PFASs also contain oxygen, hydro-
testosterone production in Leydig cells by downregulat- gen, or sulfur atoms. PFASs differ from one another in
ing Lhcgr, Scarb1, Star, Cyp11a1, and other steroidogenic the chain length (the number of carbon atoms) and other
enzymes55–57 or by inducing the apoptosis of Leydig cells.58 elements besides carbon and fluorine atoms. For example,
Interestingly, Leydig cells express two glucocorticoid perfluorododecanoic acid (PFDoA, C12) contains 12 car-
metabolizing enzymes,59–61 11β-hydroxysteroid dehy- bon atoms, perfluorononanoic acid (PFNA, C9) contains
drogenase isoform 1 (11β-HSD1) and 2 (11β-HSD2). 11β- nine carbon atoms, perfluorooctanoic acid (PFOA, C8)
HSD1 is an oxidoreductase and uses NADP+/NADPH as contains eight carbon atoms, perfluorooctane sulfonate
cofactors and primarily acts as an oxidase in adult Leydig (PFOS, C8+S1) contains eight carbon atoms and a sulfur
cells, and 11β-HSD2 is a unidirectional NAD+-dependent atom, perfluorohexane sulfonate (PFHxS, C6+S1) contains
oxidase. Both protect Leydig cell function by inactivating 6 carbon atoms and a sulfur atom, and perfluorobutane
excessive glucocorticoids in Leydig cells.59–61 Thus, inhibi- sulfonate (PFBS, C4+S1) contains 4 carbon atoms and a
tion of both these enzymes in Leydig cells could disrupt sulfur atom. PFASs, especially PFOA, PFOS, and PFHxS,
the Leydig cell function. are widespread around the world in part because of their
unique properties in the environment.68
LEYDIG CELL DEVELOPMENT When absorbed into the human body, they are
In both humans and rats, Leydig cells develop from extremely persistent because their elimination half-lives
stem Leydig cells.5 The first cells that have biomarkers in are over 4 years in humans.69 PFAS levels in the blood of
the Leydig cell lineage are the progenitor Leydig cells.62 humans are related to their exposure levels and duration.
Progenitor Leydig cells are spindle-shaped cells and express The blood levels of PFOS, PFOA, and PFHxS in the general
steroidogenic enzymes, including CYP11A1, CYP17A1, and population in the United States in 2006 were 14.7, 3.4, and
3β-HSD.63 Stem and progenitor Leydig cells have high pro- 1.5 ng/mL, respectively.70 Because of their high levels and
liferative capacity and the increased numbers of these cells potential toxicities, PFOS and PFOA were no longer man-
are critical for the population of fetal Leydig cells and adult ufactured in the United States after 2015. However, their
Leydig cells. They eventually develop into mature types of production in other countries, such as in China, is signifi-
Leydig cells (fetal Leydig cells and adult Leydig cells) with cantly increased. As a result, the levels of PFOS and PFOA
full capacity to produce androgens and insulin-like 3.5 in the cord blood were 2.48 and 6.96 ng/mL, respectively,
in residents of Shanghai in 2012.71
ENVIRONMENTAL TOXICANTS TARGETING Evidence shows that PFASs interfere with the Leydig cell
LEYDIG CELLS functions. A survey of professional workers in a branch of
Environmental toxicants can block Leydig cell develop- the 3M Company in Cottage Groove, Minnesota, which
ment, downregulate the expression levels of testosterone manufactures PFOA, was conducted, and the study found
Environmental toxicants targeting Leydig cells  249

that workers with higher serum PFOA levels had lower tes- PFASs may have multiple mechanisms to lower the bio-
tosterone levels.72,73 A study of 225 Taiwanese 13–15-year- synthesis of testosterone and insulin-like 3 by (1) reducing
old adolescents from 2009 to 2010 demonstrated that Leydig cell numbers, (2) downregulating the expression
higher levels of PFASs were associated with lower testos- levels of Leydig cell steroidogenesis-related genes, and
terone levels in males.74 A cross-sectional study of 247 men (3) inhibiting androgen-biosynthesizing enzyme activities.
in Denmark from 2008 to 2009 showed that there was a Exposure to 5- and 10-mg/kg body weight (BW)/day
negative association between serum PFOS levels and tes- PFDoA to adult rats for 14 days lowered serum testos-
tosterone concentrations.75 Animal studies also showed terone levels and downregulated Scarb1, Star, Cyp11a1,
that rats and mice had lower testosterone secretions after Hsd3b1, and Cyp17a1.77 Low doses of PFDoA (0.2 and
exposure to PFOS or PFOA.76–78 0.5 mg/kg BW/day) for long-term (110 days) treatment also

Table 20.1  Toxicants that inhibit Leydig cell development, downregulating the expressions of testosterone
biosynthesis-related genes and insulin-like 3, and inducing cell apoptosis.
Action Sites Compounds Use
Leydig cell development Perfluorooctane sulfonate Surfactant
Dipropyl phthalate Plasticizer
Dibutyl phthalate Plasticizer
Dipentyl phthalate Plasticizer
Diisoheptyl phthalate Plasticizer
Bis(2-butoxyethyl) phthalate Plasticizer
Dicyclohexyl phthalate Plasticizer
Diethylhexyl phthalate Plasticizer
Di-n-octyl phthalate Plasticizer
Diisononyl phthalate Plasticizer
Methoxychlor/hydroxychlor Insecticide
Dimethoate Insecticide
Dichlorvos Insecticide
Downregulating the expressions of Perfluorooctane sulfonate Surfactant
testosterone biosynthesis-related genes Perfluorooctane acid Surfactant
and insulin-like 3 Perfluorododecanoic acid Surfactant
Dipropyl phthalate Plasticizer
Dibutyl phthalate Plasticizer
Dipentyl phthalate Plasticizer
Diisoheptyl phthalate Plasticizer
Bis(2-butoxyethyl) phthalate Plasticizer
Dicyclohexyl phthalate Plasticizer
Diethylhexyl phthalate Plasticizer
Di-n-octyl phthalate Plasticizer
Di-isononyl phthalate Plasticizer
Bisphenol A (bisphenol AF) Plasticizer
TCDD Industrial waste
Aroclor 1254 Industrial waste
PCB126 Industrial waste
Methoxychlor/hydroxychlor Insecticide
Zearalenone (zearalenol) Mycotoxin
Aflatoxin B1 Mycotoxin
T2-toxin Mycotoxin
Citrinin Mycotoxin
Causing Leydig cell apoptosis Heavy metals Industrial waste
Zearalenone (zearalenol) Mycotoxin
Deoxynivalenol Mycotoxin
Aflatoxin B1 Mycotoxin
cAMP signaling Triclosan Fungicide
Chlorpyrifos Insecticide
250  Environmental toxicants on Leydig cell function

Table 20.2  Toxicants that directly inhibit testosterone biosynthetic enzyme activities.
Enzyme Compounds Use Mode of action
CYP11A1 Methoxychlor/hydroxychlor Insecticide Noncompetitive
Lindane Insecticide Unknown
Etomidate Drug Competitive
Gossypol Plant toxin Unknown
3β-HSD Perfluorooctane sulfonate Surfactant Competitive
Perfluorooctane acid Surfactant Competitive
Dipropyl phthalate Plasticizer Competitive
Dibutyl phthalate Plasticizer Competitive
Dipentyl phthalate Plasticizer Competitive
Bis(2-butoxyethyl) phthalate Plasticizer Competitive
Dicyclohexyl phthalate Plasticizer Competitive
Bisphenol A Plasticize Competitive
Methoxychlor/hydroxychlor Insecticide Noncompetitive
Etomidate Drug Competitive
Triphenyltin Biocide Unknown
Tributyltin Biocide Unknown
Gossypol Plant constituent Competitive
Genistein Plant ingredient Competitive
CYP17A1 Bisphenol A Plasticizer Competitive
Prochloraz Fungicide Unknown
1,2-Dibromo-3-chloropropane Insecticide Unknown
Triphenyltin Biocide Unknown
Tributyltin Biocide Unknown
Gossypol Plant constituent Unknown
17β-HSD3 Perfluorooctane sulfonate Surfactant Noncompetitive
Perfluorooctane acid Surfactant Noncompetitive
Bis(2-butoxyethyl) phthalate Plasticizer Competitive
Dicyclohexyl phthalate Plasticizer Competitive
Bisphenol A Plasticizer Competitive
Benzophenones UV blocker Unknown
Methoxychlor/hydroxychlor Insecticide Noncompetitive
Triphenyltin Biocide Unknown
Tributyltin Biocide Unknown
Gossypol Plant constituent Competitive

reduced serum testosterone levels and downregulated Star testicular 3β-HSD activity at ≤250 μM.68 In contrast,
and Cyp11a1 expressions.79 PFDoA in vitro downregulated PFOS and PFOA competitively inhibited human testicular
Star expression at its promoter region in mouse mLTC-1 17β-HSD3 with IC50 values of 6.0 and 127.6 μM, respec-
Leydig cells and primary rat Leydig cells.80 Exposure to tively.68 PFOA inhibited rat Leydig cell 17β-HSD3 with an
3- and 5-mg/kg BW/day PFNA to adult rats for 14 days IC50 value of 17 μM.83 The inhibition of 3β-HSD and 17β-
significantly lowered serum testosterone levels.81 Exposure HSD3 activities clearly caused the reduction of testoster-
to 5- or 20-mg/kg/day PFOS to prenatal rats significantly one secretion by rat Leydig cells.83 PFOS and PFOA also
lowered fetal Leydig cell numbers and downregulated the competitively inhibited human 11β-HSD1 with IC50 values
expression levels of Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, of 7.56 and 37.61 μM and rat 11β-HSD1 with IC50 values
and Cyp17a1.82 Exposure to 1-, 5-, or 10-mg/kg PFOS to of 3.45 and 45.31 μM, respectively.84 PFOS competitively
adult mice for 7, 14, or 21 days dose-dependently down- inhibited human and rat 11β-HSD2 with IC50 values of 48
regulated the expression levels of Star, Cyp11a1, Cyp17a1, and 293 nM, respectively.85 The potencies to inhibit human
and Hsd3b1.78 and rat 11β-HSD1 and 11β-HSD2 are PFOS > PFOA >
PFOS and PFOA competitively inhibited rat testicu- PFHxS > PFBS.84,85 The inhibition of both 11β-HSD1 and
lar 3β-HSD activity with IC50 values of 1.3 and 53.2 μM, 11β-HSD2 in Leydig cells could cause the inhibition of
respectively.68 Surprisingly, PFASs did not inhibit human Leydig cell steroidogenesis by excessive glucocorticoids.
Environmental toxicants targeting Leydig cells  251

Table 20.3  Toxicants that directly inhibit steroid metabolizing enzyme activities.
Enzyme Compounds Use Mode of action
SRD5A1 Tributyltin Biocide Competitive
Genistein Food ingredient Unknown
SRD5A2 Triphenyltin Biocide Noncompetitive
Tributyltin Biocide Noncompetitive
Genistein Food ingredient Unknown
11β-HSD1 Perfluorooctane sulfonate Surfactant Competitive
Perfluorooctane acid Surfactant Competitive
Dipropyl phthalate Plasticizer Competitive
Dibutyl phthalate Plasticizer Competitive
Dipentyl phthalate Plasticizer Competitive
Bis(2-butoxyethyl) phthalate Plasticizer Competitive
Dicyclohexyl phthalate Plasticizer Competitive
Bisphenol A Plasticize Competitive
Methoxychlor/hydroxychlor Insecticide Competitive
Ziram Fungicide Competitive
11β-HSD2 Perfluorooctane sulfonate Surfactant Competitive
Perfluorooctane acid Surfactant Competitive
Dipropyl phthalate Plasticizer Competitive
Dibutyl phthalate Plasticizer Competitive
Dicyclohexyl phthalate Plasticizer Competitive
Mono-(2-ethylhexyl) phthalate Plasticizer Competitive
Bisphenol A Plasticizer Unknown
Methoxychlor/Hydroxychlor Insecticide Competitive
Ziram Fungicide Noncompetitive
Thiram Fungicide Noncompetitive

Phthalates Worldwide, approximately 8.4 million tons of plasticiz-


Phthalates are synthetic compounds that are esters cre- ers are consumed every year, of which 88% are phthal-
ated by joining a phthalic anhydride with two alcohols ates (data from 2005).88 Phthalates are added to polymers
that range from methanol (C1) up to tridecyl alcohol without chemically binding to the polymers and easily
(C13). Phthalates can have either a straight chain or a leak out into the environment. The leached products in
branching chain. They are widely used as plasticizers the environment are significant because phthalates usu-
and solvents in a variety of consumer products.86 Low- ally make up to 40% of the volume of the plastics.89 The
molecular-weight phthalates (those with 1–6 carbons most abundantly used phthalates are DEHP, DBP, DEP,
in the alcohol moiety), including dimethyl (DMP, C1), and DINP.90
diethyl (DEP, C2), dipropyl (DPrP, C3), dibutyl (DBP, C4), When absorbed into the human body, phthalate dies-
diisobutyl (DIBP, C4), dipentyl (DPeP, C5), butyl benzyl ters are rapidly converted into monoester metabolites; for
(BBzP, C4/C6), dihexyl (DHP, C6), bis(2-butoxyethyl) example, DEHP into mono-ethylhexyl phthalate (MEHP)
(BBOP, C6+O), and dicyclohexyl (DCHP, C6) phthalate, and DBP into mono-butyl phthalate (MBP).91 Some mono-
are dominantly used in personal care products (such as ester metabolites are believed to be more potent than their
perfumes, lotions, and other cosmetics) or as solvents for parent compounds in their toxicity. It was found that
cellulose acetate, and in making lacquers and varnishes.87 MEHP was 10 times more potent than DEHP to disturb
High-molecular-weight phthalates (those with 7–13 car- rat testis function92 and that MBP was 1000 times more
bons in the alcohol moiety), including diisoheptyl (DIHP, potent than DBP to inhibit androgen production and
C7), di(2-ethylhexyl) (DEHP, C8), di-n-octyl (DNOP, C8), downregulate steroidogenic gene (Cyp11a1 and Hsd3b1)
diisononyl (DINP, C9), diisodecyl (DIDP, C10), diundecyl expression.93
(DIUP, C11), and ditridecyl (DTDP, C13), are primarily A human can be exposed to phthalates via various
used as plasticizers in the manufacture of flexible vinyl sources such as indoor air, dust ingestion, and food.94 For
that is used in consumer products such as flooring and example, DEHP is absorbed via food contamination.94
wall coverings, food containers and covers, and medical Low-molecular-weight phthalates such as DEP, DBP, and
devices.87 BBzP may be dermally absorbed.95 Human blood and fat
tissue samples have significant levels of phthalates. DEHP,
252  Environmental toxicants on Leydig cell function

DEP, and DBP were detected at levels of 5.71 mg/L in respectively.127 High-molecular-weight phthalates or some
serum, of 0.30 mg/L in semen, and of 0.72 mg/kg in fat low-molecular-weight phthalates (DMP and DEP) did
samples.96 not inhibit these enzyme activities at ≤1mM.127 Modes
Evidence shows that phthalates interfere with Leydig of action of these phthalates to inhibit 3β-HSD and 17β-
cell functions. Epidemiological studies claim that expo- HSD3 are competitive against steroid substrates.127 Some
sure to phthalates may be associated with male reproduc- phthalates also directly inhibited human 11β-HSD2 activ-
tive malformity in male babies.97–99 A survey of 483 men ities. DPrP, DBP, and DCHP competitively inhibited rat
showed that MEHP was inversely associated with serum 11β-HSD2 with IC50 values of 85.59, 13.69, and 32.64 μM,
testosterone levels.100 A study for serum levels of insulin- respectively. Diphthalates with 1–2 and 7–10 carbons in
like 3 (the Leydig cell biomarker) and testosterone and 14 the alcohol moiety did not inhibit 11β-HSD2 at 1 mM.128
phthalate metabolites in 1066 men showed that insulin- MEHP competitively inhibited human 11β-HSD2 with
like 3 levels were negatively associated with MEHP and IC50 values of 110.8 and 121.8 μM, while DEHP did not.128
that MBP was negatively associated with total testosterone
and free testosterone levels.101 Animal studies showed that Bisphenols
rats and mice had lower testosterone secretions after expo- Bisphenols are a group of compounds with two hydroxy-
sure to phthalates.102 phenyl groups. The most abundantly produced bisphenol
Phthalates may have multiple mechanisms to lower is bisphenol A (BPA). BPA is used to make certain plas-
the biosynthesis of testosterone and insulin-like 3 by tics and epoxy resins. A human can be exposed to BPA
(1)  disrupting Leydig cell development, (2) downregu- via various sources such as indoor air, dust ingestion, and
lating Leydig cell steroidogenesis-related gene and Insl3 food.129 Human urine samples had significant levels of
expressions, and (3) inhibiting the androgen-biosynthetic BPA with 95% detection.130–132 Men had higher BPA levels
enzyme activities. (1.49 ng/mL) than women (0.64 ng/mL), possibly because
Phthalates have been classified as antiandrogens. of androgen-related metabolism of BPA.133
However, phthalates do not block androgen receptors.90 Concerns about BPA have arisen because BPA binds to
Many in vivo studies using animal models indeed showed estrogen receptors.134,135 Evidence shows that bisphenols
that phthalates were antiandrogens, inducing androgen- (specifically BPA) interfere with the Leydig cell functions.
deficient reproductive malformations, lowering testoster- A study with 560 adult men in China showed the inverse
one levels, and causing infertility.103–108 association of serum BPA levels and testosterone levels.136
Phthalates induced abnormal Leydig cell aggrega- A National Health and Nutrition Examination Survey
tion in the fetal rat testis. Studies on DBP,109–111 DCHP,112 (NHANES) 2011–2012 survey of serum testosterone levels
DEHP,113,114 and DINP115 showed that all these phthalates and urinary BPA levels in adolescents showed the inverse
induced abnormal Leydig cell aggregation in the fetal rat association between these two indicators in boys.137
testes. The induction of fetal Leydig cell aggregation by BPA may have multiple mechanisms to lower the testos-
phthalates was independent of their inhibition of andro- terone biosynthesis by (1) blocking Leydig cell development
gen.110 Phthalate-induced fetal Leydig cell aggregation via as an androgen receptor antagonist, (2)  downregu-
may contribute to the focal dysgenesis in the testis, which lating Leydig cell steroidogenesis-related gene expres-
causes reduced fertility.116 sions, and (3) inhibiting androgen-biosynthetic enzyme
Phthalates downregulate the expression levels of activities.
steroidogenesis-related genes and Insl3 in fetal Leydig BPA is androgen receptor antagonist because it binds to
cells. Studies on DBP,117 BBzP,118 DPeP,119,120 DCHP,112 the human androgen receptor, blocking dihydrotestosterone-
DEHP,113,114,118,121 and DINP115 showed that some of mediated androgen receptor transcription activity134,135
the expression levels of these genes, including Insl3, with a similar potency as the androgen receptor antago-
Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, and Cyp17a1, were nist flutamide.134,135 Androgens have been shown to criti-
downregulated. cal for Leydig cell development, as shown by the evidence
Studies showed that DEHP and DBP inhibited Leydig of lower expression levels of CYP17A1 and 17β-HSD3 in
cell differentiation in the rat Leydig cell regeneration the androgen receptor insensitive or knockout rats and
model,113,122,123 although low doses of these phthalates may mice.138–140
increase the proliferation of Leydig cell precursors124–126 and Exposure to BPA caused a lower testosterone produc-
the commitment of stem cells into the Leydig cell lineage.123 tion in rodents.141,142 Male rats exposed to BPA (20, 100,
Studies showed that some phthalates directly inhibited and 200 mg/kg BW/day, subcutaneously) from the pre-
testosterone biosynthetic enzyme activities. Some low- pubertal to the adult period for 6 weeks had a reduced
molecular-weight phthalates, including DPrP, DBP, DPP, plasma and testicular testosterone levels and lower expres-
BBOP, and DCHP significantly inhibited 3β-HSD activi- sions of steroidogenic enzymes.143 Male rats exposed to
ties with IC50 values of 123.0, 24.1, 25.5, 50.3, and 25.5 μM BPA (2.4  μg/kg/day, gavage) from the prepubertal to the
for human enzyme, and 62.7, 30.3, 33.8, 82.6, and 24.7 μM pubertal period for 14 days had a reduced serum testos-
for rat enzyme, respectively.127 BBOP and DCHP potently terone level and the treatment of BPA to rat adult Leydig
inhibited human (IC50 values of 23.3 and 8.2  μM) and cells significantly inhibited testosterone production and
rat (IC50 values of 30.24 and 9.1 μM) 17β-HSD3 activity, Cyp17a1 expression.141 Adult male rats exposed to BPA
Environmental toxicants targeting Leydig cells  253

(400 or 800 μmol/kg BW/day) for 14 days had significantly Organochlorines


lower testosterone production and Cyp17a1 expression.144 Polychlorinated biphenyls (PCBs), pentachlorophenol
Adult mice intraperitoneally exposed to BPA (0.5, 50, and (PCP), and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD),
100 μg/kg BW/day) for 60 days had increased oxidative dichlorodiphenyltrichloroethane (DTT), and methoxy-
stress and lower testosterone production and downregu- chlor are organochlorines. PCBs are widely used as dielec-
lated Star levels.145 tric and coolant fluids in electrical apparatus, carbonless
BPA directly inhibits testosterone biosynthetic enzyme copy paper, and in heat transfer fluids. There are over 200
activities. Although BPA did not directly inhibit CYP11A1 PCB congeners. PCP is an organochlorine compound used
activity,146 it inhibited three other testosterone biosyn- as a pesticide and a disinfectant. TCDD is formed as a side
thetic enzyme activities.147 BPA inhibited human and rat product in organic synthesis and burning of organic mate-
testicular 3β-HSD with IC50 values of 7.9 and 26.5 μM, and rials. These chemicals may have a common mechanism of
human and rat CYP17A1 activities with IC50 values of 18.9 actions via aryl hydrocarbon receptor (AHR).
and 64.6 μM, as well as human and rat 17β-HSD3 with IC50 Exposure to TCDD (1–25 μg/kg BW/day) decreased
values of about 100 μM, respectively.147 BPA is a competi- serum testosterone levels and 3β-HSD and 17β-HSD
tive inhibitor for both 3β-HSD147 and CYP17A1148 against activities in rats.156 Further study showed that TCDD dis-
steroid substrates, possibly because of its similar chemi- rupted cholesterol-transporting protein (possibly StAR)
cal structure to that of steroid substrates. BPA also inhib- and CYP11A1.157 Rats exposured to TCDD (100 ng/kg BW/
ited human and rat 11β-HSD1 activities with IC50 values day) for 15 days downregulated StAR and steroidogenic
of 14.81 and 19.39 μM, respectively.149 BPA weakly inhib- enzyme expressions.158 Exposure to TCDD (0.3–1 μg/kg
ited human and rat 11β-HSD2 activity with IC50 values of BW/day) to pregnant dams also inhibited testicular tes-
about 100 μM.149 tosterone production.159 In vitro treatment of TCDD sig-
Besides BPA, its analogues (bisphenol S, bisphenol AF, nificantly downregulated Cyp11a1 transcript and reduced
bisphenol F, and tetrabromobisphenol) that were recently testosterone output in rat adult Leydig cells.160 Using Ahr
used as alternatives had similar actions to BPA. Human knockout mice, it was demonstrated that TCDD down-
fetal testes exposed to 10-nM BPA, bisphenol S, or bisphe- regulated Lhcgr and Cyp11a1 expressions via the AHR
nol AF had lower basal testosterone secretion.150 Adult pathway.161
male rats exposed to different doses of bisphenol S (1–50 Exposure to Aroclor 1254 (a mixture of PCB, 1, 2, and
μg/kg BW/day) had significantly higher testicular reactive 5 mg/kg BW/day) to lactating rats for 21 days downregu-
oxygen species (ROS) and lower testosterone levels.151 Male lated the expressions of Srd5a1 and androgen receptor.162
rats exposed to bisphenol AF (50 and 200 mg/kg BW/day) Aroclor 1254 (10–100 nM) in vitro significantly inhibited
for 14 days had lower testosterone production and Star basal and LH-stimulated testosterone production and
expression levels.152 BPA, bisphenol F, and tetrabromobi- CYP11A1 and 3β-HSD activities163 and downregulated
sphenol antagonized human androgen receptors with IC50 Star, Cyp11a1, Hsd3b1, Cyp17a1, and Hsd17b3 mRNA lev-
values of 39, 20, and 982 nM, respectively.153 els in isolated rat adult Leydig cells.164
Benzophenones Rats exposed to PCB126 or PCB169 (3 or 30 μg/kg BW/
day) from fetal days 7 to 21 reduced testosterone levels
Benzophenones are a family of compounds to protect in male pups at 21 days postpartum.165 PCB126 inhib-
from ultraviolet (UV) light. They are used in personal care ited the conversion of progesterone and androstenedione
products such as lip balm and nail polish as well as in inks, into testosterone, suggesting that 3β-HSD and 17β-HSD3
imaging, and clear coatings in the printing industry. These were inhibited.166 PCB126 also inhibited Cyp11a1 expres-
chemicals structurally differ in the positions and num- sion in Leydig cells with a mechanism similar to TCDD.167
bers of hydroxy groups and methoxyl groups in phenol. Continuous exposure to Aroclor 1242 (8 and 80 μg/kg
Benzophenone-2 and benzophenone-3 are common ingre- BW/day) to lactating dams reduced androgen production
dients in sunscreen. Benzophenones are widely exposed of adult male offspring.168
because they migrate into food from packaging.154 DDT is an organochlorine used as an insecticide and
Although there are no human epidemiological data, was banned due to its persistent pollution. It has several
in vitro testing showed that many benzophenones have metabolites that also have toxicities to the Leydig cells.
antiandrogenic activities. An in vitro study using human 3-Methylsulfonyl-DDE is a potent Leydig cell toxicant
17β-HSD3 to test the inhibitory potencies of different ben- formed from DDT and is widely present in human blood,
zophenones (benzophenones 1–8 and 12) showed that ben- milk, and fat tissue and can inhibit LH-stimulated testoster-
zophenone 1 was the most potent inhibitor with an IC50 one production. Studies showed that 3-methylsulfonyl-DDE
value of 1 μM while others had IC50 values of 47–111 μM.155 disrupted the mitochondrial proteins of porcine  Leydig
Benzophenone 1 inhibited human 17β-HSD3 more spe- cells.169,170
cifically than other 17β-hydroxysteroid dehydrogenases Methoxychlor is another synthetic organochlorine
(17β-HSD1 and 17β-HSD2) since the IC50 values for 17β- pesticide used as an alternative to DDT. Methoxychlor
HSD1 and 17β-HSD2 are over 20 μM and it did not inhibit has applications for protecting crops, o ­rnamentals,
17β-HSD5.155 Benzophenone 1 also inhibited testosterone livestock, and pets against fleas, mosquitoes, and
production in mouse and rat testes.155
254  Environmental toxicants on Leydig cell function

cockroaches. Methoxychlor was banned in the (0.001–10 μM) resulted in a significantly decreased activity
European Union in 2002 and in the United States in of adenylyl cyclase enzyme and lower testosterone section
2003. However, it is still widely used in other countries in isolated rat Leydig cells, and this effect can be prevented
(see reviews171,172). After being absorbed to mammalian by the treatment of forskolin, indicating that triclosan tar-
bodies, methoxychlor can be metabolized into the toxic gets adenylyl cyclase enzyme.191
metabolite 2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroeth-
ane (hydroxychlor).173 Methoxychlor may have multiple Carbamates
mechanisms to lower the testosterone biosynthesis by Carbamates are used as insecticides and fungicides.
(1) blocking Leydig cell development, (2) downregulat- Maneb is a widely used fungicide in agriculture. Exposure
ing Leydig cell steroidogenesis-related gene expressions, of maneb (1 and 4 mg/kg BW/day, i.p.) for 9–18 days to
and (3) directly inhibiting the androgen-­biosynthetic male rats reduced testosterone production and CYP11A1
enzyme activities. activity in Leydig cells.192 Carbendazim is a metabolite
Exposure to methoxychlor (100 and 200 mg/kg BW/ of benomyl, one of the most widespread environmental
day) to male rats from postnatal day 21 through puberty contaminants. Exposure to carbendazim (25 mg/kg BW/
significantly reduced testosterone production without day) to male rats for 48 days significantly reduced testos-
affecting LH secretion.174 Adult male rats exposed to 100 terone levels and 3β-HSD and 17β-HSD3 activities without
or 500 mg/kg BW/day methoxychlor for 28 days had lower affecting serum LH levels.193 Exposure to carbendazim to
testosterone levels.175 Administration of methoxychlor rats also caused an increase in ROS production and the
(40 and 200 mg/kg BW/day) to adult male rats for 14 days downregulation of Star mRNA levels. The addition of a
also reduced serum testosterone levels.176 Purified rat flavonoid can prevent this, suggesting an ROS-inducing
Leydig cells after treatment with hydroxychlor had a lower mechanism.194
androgen output because Cyp11a1 was downregulated.173 Molinate is a thiocarbamate that is a selective herbi-
Exposure to methoxychlor (10 and 100 mg/kg BW/day) to cide to control weeds. Exposure to molinate (40 mg/kg
adult male rats for 25 days delayed Leydig cell regenera- BW/day) to rats for 7 days inhibited testosterone levels.195
tion.177 Interestingly, prenatal exposure to methoxychlor Ziram is a widely used thiocarbamate fungicide for crops.
(10, 50, or 100 mg/kg/day) to rats increased fetal Leydig Ziram competitively inhibited rat 11βHSD1 with an IC50 of
cell numbers; however, at a high dose methoxychlor 87.07 μM. Ziram noncompetitively inhibited both rat and
directly inhibited testosterone production.178 human 11β-HSD2 with IC50 values of 90.26 and 34.93 μM,
Hydroxychlor in vitro directly inhibited testoster- possibly interacting the cysteine residues of 11β-HSD2.196
one secretion by rat Leydig cells via inhibiting CYP11A1 Thiram is a thiocarbamate fungicide and animal repellent.
activity at ≥100 nM.179 Methoxychlor is an irreversible pig It inhibited human 11β-HSD2 with a similar mechanism
CYP11A1 inhibitor.180 Methoxychlor noncompetitively to ziram.197
inhibited human and rat testicular 3β-HSD with IC50 val-
ues of 53.2 and 46.15 μM, respectively. Hydroxychlor is Polycyclic aromatic hydrocarbon
more potent than methoxychlor to inhibit 3β-HSD with Polycyclic aromatic hydrocarbons (PAHs) are a family of
IC50 values of 8.2 and 13.8 μM.181 Hydroxychlor also inhib- hydrocarbon compounds containing multiple aromatic
ited human and rat 17β-HSD3 with IC50 values of 12.1 and rings. They are produced by incomplete combustion of coal,
32.0 μM and inhibited human and rat CYP17A1 with IC50 gas, and other organic matters. There are over 100  PAH
values of 1.13 and 6.87 μM, respectively.182 Methoxychlor chemicals. One of the PAHs is benzo[a]pyrene (B[a]P), a
and hydroxychlor competitively inhibited human 11β- ubiquitous environmental pollutant. Oral exposure to B[a]
HSD1 with the IC50 values 1.917 and 8.88 μM, respec- P reduced serum and intratesticular fluid testosterone
tively,183 and hydroxychlor inhibited rat 11β-HSD1 with an levels in rats and downregulated the expression levels of
IC50 value of 9.15 μM.183 Hydroxychlor inhibited human Star and Hsd3b1 in Leydig cells.198 7,12-dimethylbenz[a]
and rat 11β-HSD2 activities with IC50 values of 55.57 and anthracene (DMBA) is a PAH metabolite and Leydig cells
12.96 μM.183 are involved in PAH and DMBA metabolism.199 Exposure
Lindane is an organochlorine insecticide that to DMBA to MA-10 Leydig cells and rat primary Leydig
impaired rat reproductive system.184,185 Lindane inhib- cells inhibited cAMP-stimulated steroid production.200
ited LH-stimulated testosterone production by rat Leydig
cells,186,187 suggesting that the compound might affect tes- Organophosphates
ticular steroidogenesis.188 Indeed, lindane inhibited mouse Organophosphates include all the insecticides, herbicides,
CYP11A1 activity.189 and some flame retardants and plasticizers.
Endosulfan is an organochlorine insecticide that is Chlorpyrifos, dimethoate, dichlorvos, and malathion
being phased out globally. It is still used extensively in are organophosphate insecticides. Piperophos is an
India, China, and a few other countries. Endosulfan is a organophosphate herbicide. An in vitro human androgen
potent human androgen receptor antagonist in an in vitro receptor screening assay showed that chlorpyrifos and
screening assay and inhibited testosterone production.190 piperophos are potent androgen receptor antagonists.190 In
Triclosan is an antibacterial and antifungal organochlo- vitro treatment of piperophos and chlorpyrifos decreased
rine used for consumer products. Treatment of triclosan testosterone secretion and downregulated Cyp11a1,
Environmental toxicants targeting Leydig cells  255

Hsd3b1, Hsd17b3, and Star in rat Leydig cells.190 etomidate downregulated the expression levels of Cyp11a1,
Chlorpyrifos also decreased in LH-stimulated stimulated Hsd17b3, and Srd5a1.212 At 1 μM, etomidate inhibited tes-
cAMP production.190 tosterone and progesterone production in isolated rat
In utero and lactational exposure to dimethoate (4, 8, Leydig cells.213
and 16 mg/kg BW) from gestational day 6 to postnatal Rosiglitazone is a thiazolidinedione drug used to treat
day 21 to mice significantly reduced Leydig cell number, diabetes. Exposure to rosiglitazone (5 mg/kg BW/day) for
size, and lowered plasma LH and testosterone levels in 15 days to rats inhibited testosterone production, possibly
the neonates.201 Dimethoate inhibited testosterone bio- via inhibiting CYP11A and 3β-HSD activities.214
synthesis in isolated rat Leydig cells and this effect can Many azole chemicals, such as prochloraz, epoxicon-
be restored by the treatment polyunsaturated fatty acids, azole, tebuconazole, and iprodione are used for fungicides
which increased the mitochondrial cholesterol pool avail- to protect plants in the agriculture field. Prochloraz is
able for testosterone biosynthesis.202 Dimethoate inhibited classified as an antiandrogen. Maternal exposure to pro-
testosterone biosynthesis in Leydig cells may be caused chloraz caused malformation of male reproductive tracts
by downregulating Star expression due to the stimulation in fetal male rats and reduced steroidogenesis in the tes-
of COX-2 because the treatment of rofecoxib (a selective tis.215 Prochloraz decreased serum testosterone levels and
COX-2 inhibitor) prevented the effect of dimethoate.203 delayed puberty in males during pubertal exposure.216
Dimethoate also decreased CYP11A1 activity and choles- Exposure to prochloraz (50 and 150 mg/kg BW/day) to
terol transportation in MA-10 mouse Leydig cells.204 rats from gestational day 7 to postnatal day 16 inhibited
Dichlorvos205,206 and malathion206 treatment disrupted testicular testosterone levels but increased progester-
Leydig cell numbers and lowered testosterone production one levels without affecting Cyp17a1, suggesting a direct
in rats. Exposure to dichlorvos (30 mg/kg/day through inhibition of CYP17A1 in the Leydig cells.217 Indeed, pro-
dermal painting) for 90 days reduced rat Leydig cell chloraz inhibited rat testicular CYP17A1 activity with Ki
numbers.207 around 1 μM.215 Using human adrenal H295R cells, pro-
Tricresyl phosphate is an organophosphate compound chloraz also concentration-dependently inhibited human
that is used as a plasticizer and for diverse other applica- CYP17A1 activity,218 and the inhibition was more selective
tions. Tricresyl phosphate in vivo and in vitro inhibited since it did not inhibit another CYP enzyme CYP11B1,
testosterone production in rat Leydig cells.208 which is required for glucocorticoid biosynthesis.218
Many organophosphates—for example, triphenyl phos- Exposure to epoxiconazole (15 or 50 mg/kg BW/day)
phate and tris-(2-chloroethyl)—phosphate, are also used or tebuconazole (50 or 100 mg/kg BW/day) from gesta-
as flame retardants. Triphenyl phosphate and tris-(2-­ tional day 7 to postnatal day 16 to rats reduced testos-
chloroethyl) phosphate in vitro significantly downregu- terone production in the testis.219 Exposure to iprodione
lated the expressions of Cyp11a1, Cyp17a1, Hsd3b1, and (50, 100, or 200 mg/kg BW/day) from postnatal days 23
Hsd17b3 genes and inhibited testosterone production in to 52 decreased testosterone, 17α-hydroxyprogesterone,
TM3 mouse Leydig cells.209 and androstenedione whereas serum LH was unaffected.
It also reduced ex vivo testosterone and progesterone pro-
Azole compounds duction, suggesting that it affects steroidogenesis through
Azole compounds refer to any of the heterocyclic com- enzyme inhibition of the steroidogenic pathway before
pounds with a five-membered ring of carbon atoms and CYP17A1.220
two to three nitrogen atoms. Azole compounds include
drugs, plant fungicides, and plant growth retardants. DBCP
Many azole compounds such as ketoconazole inhib- DBCP is a pesticide that has been used for over 20 years to
ited cytochrome P450 to exert their effects. They inhibit control plant worms. It was banned in the United States in
cytochrome P450-dependent 14α-demethylase activity 1977 because it induced infertility in male workers.221–223
that catalyzes the conversion of lanosterol to ergosterol, After exposure to DBCP, males may develop oligospermia
an essential component of fungal cell membranes used to and hypogonadism; however, the cause is reversible.224–226
kill fungi.210 Some of these drugs may also have side effects Exposure to DBCP (1–5 mg/kg BW/day) to adult rats for 6
that disrupt Leydig cell functions. Ketoconazole inhibited months significantly reduced intratesticular testosterone
testosterone production in rat Leydig cells by more than levels and LHCGR numbers in adult Leydig cells with the
50% at 0.1 μg/mL. The imidazole antifungal drugs econ- elevation of LH levels indicating direct impairment in the
azole and miconazole are used for skin or vaginal fungal testis.227 Kelce et al.228 demonstrated that DBCP also had a
infections. They decreased cAMP-stimulated StAR pro- direct inhibitory effect on the 17α-hydroxylase activity of
tein expression posttranscriptionally in Leydig cells to CYP17A1 but not the 17,20-lyase activity.228
inhibit testosterone production.211
Etomidate is an intravenous anesthetic agent. In intact Metals
Leydig cells, 30-μM etomidate inhibited androgen syn- Many heavy metals (cadmium, chromium, arsenic, and
thesis in isolated rat Leydig cells. Etomidate competitively organotins) are present in the environment because of
inhibited both CYP11A1 and 3β-HSD1 activities with IC50 industrial wastes or the widespead use of metal-containing
values of 12.62 and 2.75 μM, respectively. In addition, materials.
256  Environmental toxicants on Leydig cell function

Cadmium is used as the stabilizer in the manufacture 0.01, 0.05, and 0.1 μg/mL for 1–7 months caused Leydig
of polyvinyl chloride polymer, phosphate fertilizers, pre- cell death.243
servative pigments, and batteries. Cigarettes are also a Arsenic chemicals are present in the environment and
major source of cadmium. Each cigarette contains 5 mg they may impair Leydig cell function. Exposure of arse-
of cadmium. Cigarette smokers had significant levels of nic (0.15, 0.3, 1.5, and 3.0 mg/kg BW/2 days) for 3 weeks
blood cadmium and the levels of cadmium in blood of reduced LH and testosterone levels and downregulated
smokers was 4–5 times higher than those in nonsmok- Cyp11a1, Hsd3b, and Cyp17a1 mRNA levels.244 Exposure
ers.229 Cadmium has a very long elimination half-life of to sodium meta-arsenite (30–40 mg/L) via drinking water
20–40 years and could lead to accumulation in the human to adult mice for 30, 45, and 60 days induced Leydig cell
body.230 When cadmium was exposed, it strongly accumu- atrophy.245 Exposure to arsenic (3 mg/kg BW/day) for
lated in the interstitial cells in rat testis.231 Cadmium is a 30 days to adult mice led to the reduction of 3β-HSD and
Leydig cell toxicant. Exposure to cadmium (0.015g/L in 17β-HSD3 activities and testosterone levels and decreases
drinking water) for 1, 3, 6, and 12 months to rats reduced in glutathione S-transferase activity and glutathione lev-
Leydig cell numbers and cell size.232 A low dose of cad- els. Coadministration of α-tocopherol and ascorbic acid
mium reduced Leydig cell numbers in mice.233 Cadmium prevented the damage, suggesting that arsenic damages
exposure to rats induced an increase in ROS level and Leydig cells via increasing testicular ROS.246
decreased Star and Hsd3b1, and vitamin C and vitamin Cobalt and copper are also heavy metals that can inhibit
E were able to prevent the impairment.234 Exposure to steroid production in TM3 Leydig cells.242
cadmium (3 mg/kg BW/week, subcutaneous) to rats for Organotins are organic metals. Organotins are widely
4 weeks inhibited serum testosterone levels, and alpha- used as agricultural fungicides, rodent repellents, and anti-
tocopherol was able to prevent it. Cadmium could induce fouling biocides for ships and fishing nets.247 Increasing
ROS in rat Leydig cells to inhibit testosterone production pollution due to organotins in the environment has been
and 3β-HSD activity, and treatment with N-acetylcysteine found. Organotins may be Leydig cell toxicants. Organotins
was capable of preventing the damage.235 These results sug- can directly inhibit many androgen-biosynthetic and
gest a cadmium-mediated ROS-inducing mechanism236,237 metabolizing enzyme activities. Tributyltin inhibited
that could decrease intracellular levels of cAMP and dihy- rat 3β-HSD with a Ki of 2.4 μM.248 Tributyltin inhibited
drolipoamide dehydrogenase activity to lower testoster- pig and rat CYP17A1 with IC50 values of 117 and 50 μM,
one production in Leydig cells.230,238 respectively.249 Tributyltin inhibited pig 17β-HSD3 with
Lead is heavy and often causes the intoxication. Lead the IC50 value of 48 nM.249 Tributyltin inhibited human
environmental contamination includes mining, smelting, SRD5A1 and SRD5A2 with IC50 values of 19.9 and 10.8 μM,
manufacturing and recycling activities, and industrial uses respectively.250 However, the modes of action are different,
(leaded paint and leaded gasoline). In MA-10 cells, treat- being a competitive inhibition for SRD5A1 and irrevers-
ment with lead suppressed the cAMP-induced expression ible for SRD5A2.250 Tributyltin and triphenyltin directly
of StAR and progesterone production.239 Moreover, cad- inhibited pig CYP17A1 with an IC50 value of 117 μM.249
mium, a calcium channel blocker, abolished the inhibitory Triphenyltin directly inhibited pig CYP17A1 with an IC50
effect of lead on MA-10 cell steroid production. This indi- value of 117 μM.249 Triphenyltin inhibited pig 17β-HSD3
cates that lead might act on calcium channel to regulate with an IC50 value of 148 nM.249 Triphenyltin also inhibited
MA-10 cell steroidogenesis.239 human 3β-HSD2, 17β-HSD3, and SRD5A2 with IC50 values
Chromium is a heavy metal that is commonly used in of 4.0, 4.2, and 0.95 μM, respectively.251 Triphenyltin pos-
industrial processes such as leather tanning operations, sibly targeted the critical cysteine residues of SRD5A2 to
metal processing, stainless steel welding, chromate pro- irreversibly inhibit SRD5A2 activity.251
duction, and chrome pigment production. When chro-
mium was exposed, it also strongly accumulated in the Plant toxicants
interstitial cells in rat testis.231 Chromium may also be a Gossypol is a polyphenolic compound isolated from cotton
Leydig cell toxicant. Exposure to hexavalent chromium seeds.252 Gossypol exposure could come from the inges-
(20, 40, and 60 mg/kg BW/day) for 90 days reduced Leydig tion of cotton-seed oils and materials. Gossypol was once
cell numbers, downregulated Hsd3b1, and decreased tes- developed as a male contraceptive,253 but it apparently has
tosterone levels.240 In vitro treatment of chromium to TM3 some severe side effects, including an effect on Leydig cell
Leydig cells inhibited steroid production in these Leydig function. Men with long-term ingestion of crude cotton
cells.241,242 seed oil that contains gossypol had abnormal Leydig cell
Mercury is a heavy metal. Mercury pollution in indus- function.254 Gossypol inhibited hCG-induced testosterone
try is often in an inorganic form. However, organic production in cultured canine Leydig cells.255 Gossypol at
mercury compounds, such as methylmercury, are con- 10–20 μM in vitro also inhibited basal and LH-stimulated
centrated in the food chain because it can be produced testosterone production in mouse Leydig cells.256 Gossypol
inside fish when they are exposed to inorganic mercury. contains enantiomers, (+) gossypol and (-) gossypol, and
Exposure to methylmercury (20 mg/L) via drinking only the (-) gossypol was found to have the antifertility
water to rats for 8 weeks reduced plasma testosterone function.257 However, at ≥21 μM both enantiomers inhib-
levels.243 Oral exposure to mercuric chloride at doses of ited LH-stimulated testosterone production in mouse
References 257

Leydig cells.257 At higher concentrations, gossypol inhib- Aflatoxins are chemicals that are produced by certain
ited cAMP production in Leydig cells.258 Further studies molds. Aflatoxin B1 reduced the responsiveness of rat
showed that gossypol directly inhibited some steroido- Leydig cells to LH/hCG for stimulating testosterone pro-
genic enzymes in Leydig cells. Gossypol inhibited bovine duction.271 Aflatoxin B1 downregulated Star, Hsd3b1, and
CYP11A1 activity at 30 μM.259 Gossypol inhibited human Hsd17b3 mRNA levels, possibly via increasing ERK phos-
and rat Leydig cell 3β-HSD activities with IC50 values of phorylation and suppressing p38 MAPK and JNK activa-
3 and 0.2 μM, respectively.260 Gossypol inhibited human tion in rat Leydig cells.272
and rat 17β-HSD3 in a clear enantiomer-specific manner, T-2 toxin is one of the mycotoxins produced by several
with (−)-gossypol in inhibition of human and rat enzyme fungi.273 T-2 toxin (1–100 nM) inhibited hCG-stimulated
activities (IC50 values of 0.36 and 3.43 μM, respectively) testosterone production and downregulated Star, Cyp11a1,
and (+)-gossypol inhibition of human and rat enzyme and Hsd3b1 mRNA levels in mouse Leydig cells.274 T-2
activities (IC50 values of 1.13 and 10.93 μM, respectively).260 toxin inhibited LH-stimulated testosterone biosynthesis
Gossypol inhibited SRD5A1 more potently than SRD5A2 in primary mouse Leydig cells.275
activity.261 As well, gossypol inhibited the glucocorticoid Citrinin is a mycotoxin that is often found in food.
metabolizing enzyme, human and rat 11β-HSD2, with Citrinin (50 and 100 μM) inhibited hCG-stimulated
IC50 values of about 1 μM.262 testosterone production and downregulated StAR,
Several flavones and isoflavones might also target Leydig CYP11A1, and 3β-HSD1 levels via inducing rat Leydig cell
cells. One of them is genistein. Genistein is a soy isofla- apoptosis.276
vone and a phytoestrogen that is widely distributed in the Enniatin B is a mycotoxin that may be found in con-
human and animal diet. Several studies showed that iso- taminated cereals. Enniatin B (0.1–100 μM) reduced tes-
flavones could inhibit testosterone production in Leydig tosterone production in Leydig cells.277
cells.263 Indeed, genistein competitively and potently Cytochalasins are fungal metabolites that have the abil-
inhibited human and rat Leydig cell 3β-HSD activity with ity to bind to actin filaments. Cytochalasin B (0.1–50 μM)
IC50 values of 90 and 640 nM, respectively.264 Genistein is inhibited LH-, AMP-, and pregnenolone-stimulated tes-
a much more potent inhibitor of SRD5A2 than SRD5A1.261 tosterone production and LH-stimulated cAMP forma-
Considering that there is increasing consumption of soy- tion.278 Cytochalasin B also blocked glucose uptake in rat
based food products and their potential effects on Leydig Leydig cells.279
cells, these findings are greatly relevant to public health.
Many fungi produce toxins. Zearalenone is one of them. SUMMARY AND CONCLUSIONS
Zearalenone is a mycotoxin produced by some Fusarium Testicular Leydig cells are the place for the biosynthesis
and Gibberella species. Studies have pointed to the pos- and secretion of androgens and insulin-like 3, which are
sible toxicity of zearalenone in Leydig cells. Zearalenone critical for developmental and reproductive function in
inhibited LH-stimulated testosterone production in mouse the male. Disruption of testosterone and insulin-like 3
Leydig cells.265 However, it did not affect the expression biosynthesis and androgen metabolic activation by envi-
and the binding activity of LHCGR.265 It also downregu- ronmental toxicants can cause malformity of the male
lated Cyp11a1, Cyp17a, and Hsd17b3 mRNA levels, possibly reproductive tract, sexual dysfunction, infertility, or ste-
via suppressing Nur77 (a transcription factor) expres- rility. Many toxicants are shown to impair Leydig cells via
sion in mouse Leydig cells.265 Zearalenone inhibited cell one or more multiple pathways. Our knowledge on the
growth and increased autophagy in rat Leydig cells.266 It different Leydig cell toxicants for disruption of particular
was found that zearalenone induced Leydig cell apoptosis target molecules involved in steroidogenesis is still lim-
via an endoplasmic reticulum stress-dependent signaling ited, and therefore further studies are warranted to assess
pathway.267 Proteomics analysis showed that zearalenone the effects of environmental toxicants on male fertility.
increased energy production through promoting fatty acid
uptake and beta-oxidation and increased excessive oxida- ACKNOWLEDGMENTS
tive stress, thus possibly leading to lower steroidogenic Supported by NSFC (81102150 to LY, 30871434 and
enzyme expression levels.268 Prevention of ROS did not 31171425 to RSG, 81601266 to XL), Zhejiang Provincial
seem to restore steroidogenesis in MA-10 Leydig cells,269 NSF (LY15H310008 to RSG, LQ16H040005 to XC), and
suggesting that its ROS-inducing effect does not con- Health & Family Planning Commission of Zhejiang
tribute to the inhibition of steroidogenesis. α-Zearalenol Province (11-CX29 and 2013ZDA017 to RSG, 2015103197
(0.01–100 μM), a zearalenone metabolite, also suppressed and 2017KY483 to XL, 2016KYB199 and 2017KY466 to
hCG-induced testosterone production and downregulated XC), Science and Technology Department of Zhejiang
Star, Cyp11a1, and Hsd3b1 in mouse Leydig cells,270 indi- Provincial Government (2012C13016-1 to QL).
cating a similar mechanism to that of zearalenone.
Deoxynivalenol is a type B trichothecene and is a myco- REFERENCES
toxin that occurs predominantly in grains. Deoxynivalenol 1. Huhtaniemi I, Pelliniemi LJ. Fetal Leydig cells:
inhibited Leydig cell steroidogenesis and oxidative stress, Cellular origin, morphology, life span, and spe-
and prevention of oxidative stress also cannot restore its sup- cial functional features. 116. Proc Soc Exp Biol Med.
pression of steroidogenesis in MA-10 mouse Leydig cells.269 1992;201(2):125–140.
258  Environmental toxicants on Leydig cell function

2. Fujii T. Roles of age and androgen in the regula- 18. Samtani R, Bajpai M, Ghosh PK, Saraswathy KN.
tion of sex accessory organs. 106. Adv Sex Horm Res. SRD5A2 gene mutations—A population-based
1977;3:103–137. review. Pediatr Endocrinol Rev. 8(1):34–40.
3. Awoniyi CA, Santulli R, Sprando RL, Ewing LL, 19. Maimoun L, Philibert P, Cammas B et al.
Zirkin BR. Restoration of advanced spermato- Phenotypical, biological, and molecular hetero-
genic cells in the experimentally regressed rat geneity of 5alpha-reductase deficiency: An exten-
testis: Quantitative relationship to testosterone sive international experience of 55 patients. J Clin
concentration within the testis. Endocrinology. Endocrinol Metab. 96(2):296–307.
1989;124(3):1217–1223. 20. Chung BC, Matteson KJ, Voutilainen R, Mohandas
4. Wilson JD. Prospects for research for disorders of the TK, Miller WL. Human cholesterol side-chain
endocrine system. JAMA. 2001;285(5):624–627. cleavage enzyme, P450scc: cDNA cloning, assign-
5. Chen H, Wang Y, Ge R, Zirkin BR. Leydig cell stem ment of the gene to chromosome 15, and expression
cells: Identification, proliferation and differentiation. in the placenta. Proc Natl Acad Sci U S A. 1986;83(23):​
Mol Cell Endocrinol. 2016;445:65–73. 8962–8966.
6. Zimmermann S, Steding G, Emmen JM et al. Targeted 21. Oonk RB, Krasnow JS, Beattie WG, Richards JS.
disruption of the Insl3 gene causes bilateral cryptor- Cyclic AMP-dependent and -independent regulation
chidism. Mol Endocrinol. 1999;13(5):681–691. of cholesterol side chain cleavage cytochrome P-450
7. Adham IM, Emmen JM, Engel W. The role of the tes- (P-450scc) in rat ovarian granulosa cells and corpora
ticular factor INSL3 in establishing the gonadal posi- lutea. cDNA and deduced amino acid sequence of rat
tion. Mol Cell Endocrinol. 2000;160(1–2):11–16. P-450scc. J Biol Chem. 1989;264(36):21934–21942.
8. Anand-Ivell R, Heng K, Hafen B, Setchell B, Ivell R. 22. Hanukoglu I, Gutfinger T, Haniu M, Shively JE.
Dynamics of INSL3 peptide expression in the rodent Isolation of a cDNA for adrenodoxin reductase
testis. Biol Reprod. 2009;81(3):480–487. (ferredoxin-NADP+ reductase). Implications for
­
9. Prince FP. Ultrastructural evidence of mature mitochondrial cytochrome P-450 systems. Eur J
Leydig cells and Leydig cell regression in the neona- Biochem. 1987;169(3):449–455.
tal human testis. Anat Rec. 1990;228(4):405–417. 23. Hanukoglu I, Jefcoate CR. Mitochondrial cyto-
10. Skakkebaek NE, Rajpert-De Meyts E, Main KM. chrome P-450scc. Mechanism of electron transport
Testicular dysgenesis syndrome: An increasingly by adrenodoxin. J Biol Chem. 1980;255(7):3057–3061.
common developmental disorder with environmen- 24. Simard J, Ricketts ML, Gingras S, Soucy P, Feltus
tal aspects. Hum Reprod. 2001;16(5):972–978. FA, Melner MH. Molecular biology of the 3beta-
11. Carlsen E, Swan SH, Petersen JH, Skakkebaek NE. hydroxysteroid dehydrogenase/delta5-delta4 isom-
Longitudinal changes in semen parameters in young erase gene family. Endocr Rev. 2005;26(4):525–582.
Danish men from the Copenhagen area. Hum Reprod. 25. Penning TM. Molecular endocrinology of hydro­
2005;20(4):942–949. xysteroid dehydrogenases. Endocr Rev. 1997;18(3):​
12. Moline JM, Golden AL, Bar-Chama N et al. 281–305.
Exposure to hazardous substances and male repro- 26. Tang PZ, Tsai-Morris CH, Dufau ML. Regulation of
ductive health: A research framework. Environ Health 3beta-hydroxysteroid dehydrogenase in gonadotropin-
Perspect. 2000;108(9):803–813. induced steroidogenic desensitization of Leydig cells.
13. Ge RS, Hardy MP. Regulation of Leydig cells dur- Endocrinology. 1998;139(11):4496–4505.
ing pubertal development. In Payne AH, Hardy MP, 27. Mendonca BB, Bloise W, Arnhold IJ et al. Male pseu-
eds. The Leydig Cell in Health and Disease. Totowa, NJ: dohermaphroditism due to nonsalt-losing 3 beta-
Humana Press; 2007:55–70. hydroxysteroid dehydrogenase deficiency: Gender
14. Payne AH, O’Shaughnessy PJ. Structure, function role change and absence of gynecomastia at puberty.
and regulation of steroidogenic enzymes in the J Steroid Biochem. 1987;28(6):669–675.
Leydig cell. In Payne AH, Hardy MP, and Russell LD, 28. Lutfallah C, Wang W, Mason JI et al. Newly pro-
eds. The Leydig Cell. Vienna, IL: Cache River Press; posed hormonal criteria via genotypic proof for type
1996:259–286. II 3beta-hydroxysteroid dehydrogenase deficiency.
15. Shima Y, Miyabayashi K, Haraguchi S et al. J Clin Endocrinol Metab. 2002;87(6):2611–2622.
Contribution of Leydig and Sertoli cells to testoster- 29. Chung BC, Picado-Leonard J, Haniu M et al.
one production in mouse fetal testes. Mol Endocrinol. Cytochrome P450c17 (steroid 17 alpha-hydroxylase/​
2013;27(1):63–73. 17,20 lyase): Cloning of human adrenal and testis
16. Ge RS, Hardy MP. Variation in the end products of cDNAs indicates the same gene is expressed in both
androgen biosynthesis and metabolism during post- tissues. Proc Natl Acad Sci U S A. 1987;84(2):407–411.
natal differentiation of rat Leydig cells. Endocrinology. 30. Namiki M, Kitamura M, Buczko E, Dufau ML. Rat
1998;139(9):3787–3795. testis P-450(17)alpha cDNA: The deduced amino
17. Eik-Nes KB. Production and secretion of 5a-reduced acid sequence, expression and secondary struc-
testosterone(DHT) by male reproductive organs. tural configuration. Biochem Biophys Res Commun.
J Steroid Biochem. 1975;6(3–4):337–339. 1988;157(2):705–712.
References 259

31. Pandey AV, Miller WL. Regulation of 17,20 lyase activ- 45. Wilson JD, Goldstein JL. Classification of hereditary
ity by cytochrome b5 and by serine phosphorylation disorders of sexual development. Birth Defects Orig
of P450c17. J Biol Chem. 2005;280(14):13265–13271. Artic Ser. 1975;11(4):1–16.
32. Fukami M, Homma K, Hasegawa T, Ogata T. 46. Andersson S, Berman DM, Jenkins EP, Russell
Backdoor pathway for dihydrotestosterone biosyn- DW. Deletion of steroid 5 alpha-reductase 2
thesis: Implications for normal and abnormal human gene in male pseudohermaphroditism. Nature.
sex development. Dev Dyn. 2012;242(4):320–329. 1991;354(6349):159–161.
33. Gilep AA, Sushko TA, Usanov SA. At the crossroads 47. Wuerther JU, Kirstler M, Kratmeier M, Mukhopadhyay
of steroid hormone biosynthesis: The role, substrate AK. LH/hCG-receptor is coupled to both adenylate
specificity and evolutionary development of CYP17. cyclase and protein kinase C pathways in isolated
Biochim Biophys Acta. 1814(1):200–209. mouse Leydig cells. Endocrine. 1995;3:579–584.
34. Geller DH, Auchus RJ, Mendonca BB, Miller WL. 48. Calvo D, Vega MA. Identification, primary struc-
The genetic and functional basis of isolated 17,20- ture, and distribution of CLA-1, a novel mem-
lyase deficiency. Nat Genet. 1997;17(2):201–205. ber of the CD36/LIMPII gene family. J Biol Chem.
35. Jones KL, Freidenberg GR, Buchta R, Derenoncourt 1993;268(25):18929–18935.
A. Male pseudohermaphroditism resulting from 17 49. Mizutani T, Sonoda Y, Minegishi T, Wakabayashi K,
alpha-monooxygenase (P-450C17) deficiency in two Miyamoto K. Cloning, characterization, and cellular
unrelated Guamanians. Am J Dis Child. 1992;146(5):​ distribution of rat scavenger receptor class B type
592–595. I (SRBI) in the ovary. Biochem Biophys Res Commun.
36. Geissler WM, Davis DL, Wu L et al. Male pseudo- 1997;234(2):499–505.
hermaphroditism caused by mutations of testicular 50. Zhang YF, Yuan KM, Liang Y et al. Alterations of
17 beta-hydroxysteroid dehydrogenase 3. Nat Genet. gene profiles in Leydig-cell-regenerating adult rat
1994;7(1):34–39. testis after ethane dimethane sulfonate-treatment.
37. Andersson S, Geissler WM, Wu L et al. Molecular Asian J Androl. 2015;17(2):253–260.
genetics and pathophysiology of 17 beta-hydroxysteroid 51. Rhainds D, Brissette L. The role of scavenger recep-
dehydrogenase 3 deficiency. J Clin Endocrinol Metab. tor class B type I (SR-BI) in lipid trafficking. Defining
1996;81(1):130–136. the rules for lipid traders. Int J Biochem Cell Biol.
38. Rosler A, Silverstein S, Abeliovich D. A (R80Q) 2004;36(1):39–77.
mutation in 17 beta-hydroxysteroid dehydrogenase 52. Lin D, Sugawara T, Strauss JF 3rd et al. Role of ste-
type 3 gene among Arabs of Israel is associated roidogenic acute regulatory protein in adrenal and
with pseudohermaphroditism in males and nor- gonadal  steroidogenesis. Science. 1995;267(5205):​
mal asymptomatic females. J Clin Endocrinol Metab. 1828–1831.
1996;81(5):1827–1831. 53. Bose HS, Whittal RM, Baldwin MA, Miller WL. The
39. Andersson S, Russell DW. Structural and biochemi- active form of the steroidogenic acute regulatory
cal properties of cloned and expressed human and protein, StAR, appears to be a molten globule. Proc
rat steroid 5 alpha-reductases. Proc Natl Acad Sci U S A. Natl Acad Sci U S A. 1999;96(13):7250–7255.
1990;87(10):3640–3644. 54. Roostaee A, Barbar E, Lehoux JG, Lavigne P.
40. Thigpen AE, Davis DL, Gautier T, Imperato- Cholesterol binding is a prerequisite for the activity
McGinley J, Russell DW. Brief report: The molecu- of the steroidogenic acute regulatory protein (StAR).
lar basis of steroid 5 alpha-reductase deficiency Biochem J. 2008;412(3):553–562.
in a large Dominican kindred. N Engl J Med. 55. Xiao YC, Huang YD, Hardy DO, Li XK, Ge RS.
1992;327(17):1216–1219. Glucocorticoid suppresses steroidogenesis in rat pro-
41. Uemura M, Tamura K, Chung S et al. Novel 5 alpha- genitor Leydig cells. J Androl. 2010;31(4):365–371.
steroid reductase (SRD5A3, type-3) is overexpressed 56. Lin H, Yuan KM, Zhou HY et al. Time-course
in hormone-refractory prostate cancer. Cancer Sci. changes of steroidogenic gene expression and ste-
2008;99(1):81–86. roidogenesis of rat Leydig cells after acute immobi-
42. Jenkins EP, Hsieh CL, Milatovich A et al. lization stress. Int J Mol Sci. 2014;15(11):21028–21044.
Characterization and chromosomal mapping of a 57. Witorsch RJ. Effects of elevated glucocorticoids on
human steroid 5 alpha-reductase gene and pseu- reproduction and development: Relevance to endo-
dogene and mapping of the mouse homologue. crine disruptor screening. Crit Rev Toxicol. 2016;​
Genomics. 1991;11(4):1102–1112. 46(5):420–436.
43. Thigpen AE, Davis DL, Milatovich A et al. Molecular 58. Gao HB, Tong MH, Hu YQ, Guo QS, Ge R, Hardy
genetics of steroid 5 alpha-reductase 2 deficiency. MP. Glucocorticoid induces apoptosis in rat Leydig
J Clin Invest. 1992;90(3):799–809. cells. Endocrinology. 2002;143(1):130–138.
44. Cantagrel V, Lefeber DJ, Ng BG et al. SRD5A3 is 59. Ge RS, Gao HB, Nacharaju VL, Gunsalus GL, Hardy
required for converting polyprenol to dolichol and is MP. Identification of a kinetically distinct activity of
mutated in a congenital glycosylation disorder. Cell. 11beta-hydroxysteroid dehydrogenase in rat Leydig
142(2):203–217. cells. Endocrinology. 1997;138(6):2435–2442.
260  Environmental toxicants on Leydig cell function

60. Ge RS, Hardy DO, Catterall JF, Hardy MP. 73. Gilliland FD, Mandel JS. Mortality among employ-
Developmental changes in glucocorticoid recep- ees of a perfluorooctanoic acid production plant.
tor and 11b-hydroxysteroid dehydrogenase oxi- J Occup Med. 1993;35(9):950–954.
dative and reductive activities in rat Leydig cells. 74. Zhou Y, Hu LW, Qian ZM et al. Association of per-
Endocrinology. 1997;138(12):5089–5095. fluoroalkyl substances exposure with reproductive
61. Ge RS, Dong Q, Niu EM et al. 11{beta}-Hydroxysteroid hormone levels in adolescents: By sex status. Environ
dehydrogenase 2 in rat Leydig cells: Its role in blunt- Int. 2016;94:189–195.
ing glucocorticoid action at physiological levels of sub- 75. Joensen UN, Veyrand B, Antignac JP et al. PFOS
strate. Endocrinology. 2005;146(6):2657–2664. (perfluorooctanesulfonate) in serum is negatively
62. Shan LX, Hardy MP. Developmental changes in associated with testosterone levels, but not with
levels of luteinizing hormone receptor and andro- semen quality, in healthy men. Hum Reprod. 2013;​
gen receptor in rat Leydig cells. Endocrinology. 28(3):599–608.
1992;131(3):1107–1114. 76 Biegel LB, Liu RC, Hurtt ME, Cook JC. Effects of
63. Ariyaratne HB, Mendis-Handagama SM, Hales DB, ammonium perfluorooctanoate on Leydig cell func-
Mason IJ. Studies of the onset of Leydig cell differ- tion: In vitro, in vivo, and ex vivo studies. Toxicol Appl
entiation in the prepubertal rat testis. Biol Reprod. Pharmacol. 1995;134(1):18–25.
2000;63:165–171. 77. Shi Z, Zhang H, Liu Y, Xu M, Dai J. Alterations in
64. Hu GX, Lian QQ, Ge RS, Hardy DO, Li XK. gene expression and testosterone synthesis in the tes-
Phthalate-induced testicular dysgenesis syndrome: tes of male rats exposed to perfluorododecanoic acid.
Leydig cell influence. Trends Endocrinol Metab. Toxicol Sci. 2007;98(1):206–215.
2009;20(3):139–145. 78. Wan HT, Zhao YG, Wong MH et al. Testicular sig-
65. Abdellatif AG, Preat V, Vamecq J, Nilsson R, Roberfroid naling is the potential target of perfluorooctane-
M. Peroxisome proliferation and modulation of  rat sulfonate-mediated subfertility in male mice. Biol
liver carcinogenesis by 2,4-­ dichlorophenoxyacetic Reprod. 2011;84(5):1016–1023.
acid, 2,4,5-trichlorophenoxyacetic acid, perfluorooc- 79. Shi Z, Ding L, Zhang H, Feng Y, Xu M, Dai J. Chronic
tanoic acid and nafenopin. Carcinogenesis. 1990;11(11):​ exposure to perfluorododecanoic acid disrupts tes-
1899–1902. ticular steroidogenesis and the expression of related
66. Jensen AA, Leffers H. Emerging endocrine dis- genes in male rats. Toxicol Lett. 2009;188(3):192–200.
rupters: Perfluoroalkylated substances. Int J Androl. 80. Shi Z, Feng Y, Wang J, Zhang H, Ding L, Dai J.
2008;31(2):161–169. Perfluorododecanoic acid-induced steroidogenic inhi-
67. Johnson JD, Gibson SJ, Ober RE. Cholestyramine- bition is associated with steroidogenic acute regulatory
enhanced fecal elimination of carbon-14 in rats after protein and reactive oxygen species in cAMP-stimulated
administration of ammonium [14C]perfluorooc- Leydig cells. Toxicol Sci. 2010;114(2):285–294.
tanoate or potassium [14C]perfluorooctanesulfo- 81. Feng Y, Shi Z, Fang X, Xu M, Dai J. Perfluorononanoic
nate. Fundam Appl Toxicol. 1984;4(6):972–976. acid induces apoptosis involving the Fas death
68. Zhao B, Hu GX, Chu Y et al. Inhibition of human receptor signaling pathway in rat testis. Toxicol Lett.
and rat 3beta-hydroxysteroid dehydrogenase and 2009;190(2):224–230.
17beta-hydroxysteroid dehydrogenase 3 activities 82. Zhao B, Li L, Liu J et al. Exposure to perfluorooctane
by perfluoroalkylated substances. Chem Biol Interact. sulfonate in utero reduces testosterone production in
188(1):38–43. rat fetal Leydig cells. PLoS One. 2014;9(1):e78888.
69. Giesy JP, Kannan K. Perfluorochemical surfac- 83. Zhao B, Chu Y, Hardy DO, Li XK, Ge RS. Inhibition
tants in the environment. Environ Sci Technol. of 3beta- and 17beta-hydroxysteroid dehydrogenase
2002;36(7):146A–152A. activities in rat Leydig cells by perfluorooctane acid.
70. Olsen GW, Mair DC, Church TR et al. Decline J Steroid Biochem Mol Biol. 118(1–2):13–17.
in perfluorooctanesulfonate and other polyfluo- 84. Ye L, Zhao B, Cai XH, Chu Y, Li C, Ge RS. The inhib-
roalkyl chemicals in American Red Cross adult itory effects of perfluoroalkyl substances on human
blood donors, 2000–2006. Environ Sci Technol. and rat 11beta-hydroxysteroid dehydrogenase 1.
2008;42(13):4989–4995. Chem Biol Interact. 2011;195(2):114–118.
71. Wang B, Chen Q, Shen L, Zhao S, Pang W, Zhang J. 85. Zhao B, Lian Q, Chu Y, Hardy DO, Li XK, Ge RS. The
Perfluoroalkyl and polyfluoroalkyl substances in cord inhibition of human and rat 11beta-hydroxysteroid
blood of newborns in Shanghai, China: Implications dehydrogenase 2 by perfluoroalkylated substances.
for risk assessment. Environ Int. 2016;97:7–14. J Steroid Biochem Mol Biol. 2011;125(1–2):143–147.
72. Olsen GW, Gilliland FD, Burlew MM, Burris JM, 86. Kohn MC, Parham F, Masten SA et al. Human expo-
Mandel JS, Mandel JH. An epidemiologic investiga- sure estimates for phthalates. Environ Health Perspect.
tion of reproductive hormones in men with occupa- 2000;108(10):A440–A442.
tional exposure to perfluorooctanoic acid. J Occup 87. Hauser R, Calafat AM. Phthalates and human health.
Environ Med. 1998;40(7):614–622. Occup Environ Med. 2005;62(11):806–818.
References 261

88. Benjamin S, Masai E, Kamimura N et al. Phthalates 102. Zhang J, Jin S, Zhao J, Li H. Effect of dibutyl phthal-
impact human health: Epidemiological evidences ate on expression of connexin 43 and testosterone
and plausible mechanism of action. J Hazard Mater. production of Leydig cells in adult rats. Environ
2017;340:360–383. Toxicol Pharmacol. 2016;47:131–135.
89. Ma X, Lian QQ, Dong Q, Ge RS. Environmental 103. Howdeshell KL, Furr J, Lambright CR, Rider CV,
inhibitors of 11β-hydroxysteroid dehydrogenase Wilson VS, Gray LE Jr. Cumulative effects of dibu-
type 2. Toxicology 2011;285(3):83–9. tyl phthalate and diethylhexyl phthalate on male
90. Ge RS, Chen GR, Tanrikut C, Hardy MP. Phthalate rat reproductive tract development: Altered fetal
ester toxicity in Leydig cells: Developmental timing steroid hormones and genes. Toxicol Sci. 2007;99(1):​
and dosage considerations. Reprod Toxicol. 2007;23(3):​ 190–202.
366–373. 104. Creasy DM, Beech LM, Gray TJ, Butler WH.
91. Albro PW, Corbett JT, Schroeder JL, Jordan S, The ultrastructural effects of di-n-pentyl phthal-
Matthews HB. Pharmacokinetics, interactions with ate on the testis of the mature rat. Exp Mol Pathol.
macromolecules and species differences in metab- 1987;46(3):357–371.
olism of DEHP. Environ Health Perspect. 1982;45:​ 105. Gray TJ, Gangolli SD. Aspects of the testicular tox-
19–25. icity of phthalate esters. Environ Health Perspect.
92. Albro PW, Chapin RE, Corbett JT, Schroeder J, 1986;65:229–235.
Phelps JL. Mono-2-ethylhexyl phthalate, a metabo- 106. Sjoberg P, Bondesson U, Gray TJ, Ploen L. Effects of
lite of di-(2-ethylhexyl) phthalate, causally linked di-(2-ethylhexyl) phthalate and five of its metabolites
to testicular atrophy in rats. Toxicol Appl Pharmacol. on rat testis in vivo and in in vitro. Acta Pharmacol
1989;100(2):193–200. Toxicol (Copenh). 1986;58(3):225–233.
93. Li L, Chen X, Hu G et al. Comparison of the effects 107. Gray TJ, Beamand JA. Effect of some phthalate esters
of dibutyl and monobutyl phthalates on the steroido- and other testicular toxins on primary cultures
genesis of rat immature Leydig cells. Biomed Res Int. of testicular cells. Food Chem Toxicol. 1984;22(2):​
2016;2016:1376526. 123–131.
94. Erythropel HC, Maric M, Nicell JA, Leask RL, 108. Gray TJ, Butterworth KR. Testicular atrophy pro-
Yargeau V. Leaching of the plasticizer di(2-ethylhexyl)​ duced by phthalate esters. Arch Toxicol Suppl. 1980;​
phthalate (DEHP) from plastic containers and the 4:452–455.
question of human exposure. Appl Microbiol Biotechnol. 109. Mahood IK, Hallmark N, McKinnell C, Walker M,
2014;98(24):9967–9981. Fisher JS, Sharpe RM. Abnormal Leydig Cell aggre-
95. Heudorf U, Mersch-Sundermann V, Angerer J. gation in the fetal testis of rats exposed to di (n-butyl)
Phthalates: Toxicology and exposure. Int J Hyg Environ phthalate and its possible role in testicular dysgen-
Health. 2007;210(5):623–634. esis. Endocrinology. 2005;146(2):613–623.
96. Dobrzynska MM. Phthalates—Widespread occur- 110. Scott HM, Hutchison GR, Mahood IK et al. Role of
rence and the effect on male gametes. Part 1. General androgens in fetal testis development and dysgen-
characteristics, sources and human exposure. Rocz esis. Endocrinology. 2007;148(5):2027–2036.
Panstw Zakl Hig. 2016;67(2):97–103. 111. Hallmark N, Walker M, McKinnell C et al. Effects
97. Sharpe RM. Hormones and testis development and of monobutyl and di(n-butyl) phthalate in vitro
the possible adverse effects of environmental chemi- on steroidogenesis and Leydig cell aggregation in
cals. Toxicol Lett. 2001;120(1–3):221–232. fetal testis explants from the rat: Comparison with
98. Akingbemi BT, Hardy MP. Oestrogenic and anti- effects in vivo in the fetal rat and neonatal marmo-
androgenic chemicals in the environment: Effects set and in vitro in the human. Environ Health Perspect.
on male reproductive health. Ann Med. 2001;33(6):​ 2007;115(3):390–396.
391–403. 112. Li X, Chen X, Hu G et al. Effects of in utero exposure
99. Zhang Y, Lin L, Cao Y, Chen B, Zheng L, Ge RS. to dicyclohexyl phthalate on rat fetal Leydig cells. Int
Phthalate levels and low birth weight: A nested J Environ Res Public Health. 2016;13(3):246.
case-control study of Chinese newborns. J Pediatr. 113. Lin H, Ge RS, Chen GR et al. Involvement of testic-
2009;155(4):500–504. ular growth factors in fetal Leydig cell aggregation
100. Wang YX, Zeng Q, Sun Y et al. Phthalate exposure after exposure to phthalate in utero. Proc Natl Acad Sci
in association with serum hormone levels, sperm U S A. 2008;105(20):7218–7222.
DNA damage and spermatozoa apoptosis: A cross- 114. Lin H, Lian QQ, Hu GX et al. In utero and lactational
sectional study in China. Environ Res. 2015;150:​ exposures to diethylhexyl-phthalate affect two popu-
557–565. lations of Leydig cells in male long-evans rats. Biol
101. Pan Y, Jing J, Dong F et al. Association between Reprod. 2009;80(5):882–888.
phthalate metabolites and biomarkers of reproduc- 115. Li L, Bu T, Su H et al. Inutero exposure to diisononyl
tive function in 1066 Chinese men of reproductive phthalate caused testicular dysgenesis of rat fetal tes-
age. J Hazard Mater. 2015;300:729–736. tis. Toxicol Lett. 2014;232(2):466–474.
262  Environmental toxicants on Leydig cell function

116. Mahood IK, Scott HM, Brown R, Hallmark N, Walker 129. Loganathan SN, Kannan K. Occurrence of bisphenol
M, Sharpe RM. In utero exposure to di(n-butyl) a in indoor dust from two locations in the eastern
phthalate and testicular dysgenesis: Comparison of United States and implications for human exposures.
fetal and adult end points and their dose sensitivity. Arch Environ Contam Toxicol. 61(1):68–73.
Environ Health Perspect. 2007;​115(Suppl 1):​55–61. 130. Calafat AM, Kuklenyik Z, Reidy JA, Caudill SP,
117. Barlow NJ, Phillips SL, Wallace DG, Sar M, Gaido Ekong J, Needham LL. Urinary concentrations of
KW, Foster PM. Quantitative changes in gene bisphenol A and 4-nonylphenol in a human refer-
expression in fetal rat testes following exposure to ence population. Environ Health Perspect. 2005;113(4):​
di(n-butyl) phthalate. Toxicol Sci. 2003;73(2):​431–441. 391–395.
118. Wilson VS, Lambright C, Furr J et al. Phthalate ester- 131. Calafat AM, Ye X, Wong LY, Reidy JA, Needham LL.
induced gubernacular lesions are associated with Exposure of the U.S. population to bisphenol A and
reduced insl3 gene expression in the fetal rat testis. 4-tertiary-octylphenol: 2003–2004. Environ Health
Toxicol Lett. 2004;146(3):207–215. Perspect. 2008;116(1):39–44.
119. Beverly BE, Lambright CS, Furr JR et al. Simvastatin 132. Ouchi K, Watanabe S. Measurement of bisphenol A
and dipentyl phthalate lower ex vivo testicular tes- in human urine using liquid chromatography with
tosterone production and exhibit additive effects on multi-channel coulometric electrochemical detec-
testicular testosterone and gene expression via dis- tion. J Chromatogr B Analyt Technol Biomed Life Sci.
tinct mechanistic pathways in the fetal rat. Toxicol 2002;780(2):365–370.
Sci. 2014;141(2):524–537. 133. Takeuchi T, Tsutsumi O. Serum bisphenol a concen-
120. Gray LE Jr, Furr J, Tatum-Gibbs KR et al. Establishing trations showed gender differences, possibly linked
the “biological relevance” of dipentyl phthalate to androgen levels. Biochem Biophys Res Commun.
reductions in fetal rat testosterone production and 2002;291(1):76–78.
plasma and testis testosterone levels. Toxicol Sci. 134. Kuiper GG, Lemmen JG, Carlsson B et al.
2015;149(1):178–191. Interaction of estrogenic chemicals and phytoes-
121 Borch J, Metzdorff SB, Vinggaard AM, Brokken L, trogens with estrogen receptor beta. Endocrinology.
Dalgaard M. Mechanisms underlying the anti- 1998;139(10):4252–4263.
androgenic effects of diethylhexyl phthalate in fetal 135. Sohoni P, Sumpter JP. Several environmental oes-
rat testis. Toxicology. 2006;223(1–2):144–155. trogens are also anti-androgens. J Endocrinol.
122. Guo J, Li XW, Liang Y et al. The increased number 1998;158(3):327–339.
of Leydig cells by di(2-ethylhexyl) phthalate comes 136. Liang H, Xu W, Chen J et al. The association between
from the differentiation of stem cells into Leydig exposure to environmental bisphenol A and gonad-
cell lineage in the adult rat testis. Toxicology. 2013;​ otropic hormone levels among men. PLoS One.
306C:9–15. 2017;12(1):e0169217.
123. Li XW, Liang Y, Su Y et al. Adverse effects of di- 137. Scinicariello F, Buser MC. Serum ­testosterone con­
(2-ethylhexyl) phthalate on Leydig cell regeneration centrations and urinary bisphenol A, Benzophenone-3,
in the adult rat testis. Toxicol Lett. 2012;215(2):84–91. triclosan, and paraben levels in male and female chil-
124. Ge RS, Chen GR, Dong Q et al. Biphasic effects of dren and adolescents: NHANES 2011–2012. Environ
postnatal exposure to diethylhexylphthalate on the Health Perspect. 2016;124(12):1898–1904.
timing of puberty in male rats. J Androl. 2007;28(4):​ 138. O’Hara L, McInnes K, Simitsidellis I et al. Autocrine
513–520. androgen action is essential for Leydig cell matura-
125. Akingbemi BT, Ge R, Klinefelter GR, Zirkin BR, tion and function, and protects against late-onset
Hardy MP. Phthalate-induced Leydig cell hyper- Leydig cell apoptosis in both mice and men. FASEB J.
plasia is associated with multiple endocrine distur- 2014;29(3):894–910.
bances. Proc Natl Acad Sci U S A. 2004;101(3):775–780. 139. Kilcoyne KR, Smith LB, Atanassova N et al. Fetal
126. Heng K, Anand-Ivell R, Teerds K, Ivell R. The endo- programming of adult Leydig cell function by andro-
crine disruptors dibutyl phthalate (DBP) and dieth- genic effects on stem/progenitor cells. Proc Natl Acad
ylstilbestrol (DES) influence Leydig cell regeneration Sci U S A. 2014;111(18):E1924–1932.
following ethane dimethane sulphonate treatment of 140. Murphy L, O’Shaughnessy PJ. Testicular steroido-
adult male rats. Int J Androl. 2011;35(3):353–363. genesis in the testicular feminized (Tfm) mouse:
127. Yuan K, Zhao B, Li XW et al. Effects of phthalates Loss of 17 alpha-hydroxylase activity. J Endocrinol.
on 3beta-hydroxysteroid dehydrogenase and 17beta- 1991;131(3):443–449.
hydroxysteroid dehydrogenase 3 activities in human 141. Akingbemi BT, Sottas CM, Koulova AI, Klinefelter
and rat testes. Chem Biol Interact. 2012;195(3):​180–188. GR, Hardy MP. Inhibition of testicular steroidogene-
128. Zhao B, Chu Y, Huang Y, Hardy DO, Lin S, Ge RS. sis by the xenoestrogen bisphenol A is associated with
Structure-dependent inhibition of human and rat reduced pituitary luteinizing hormone secretion and
11beta-hydroxysteroid dehydrogenase 2 activities by decreased steroidogenic enzyme gene expression in
phthalates. Chem Biol Interact. 2010;183(1):79–84. rat Leydig cells. Endocrinology. 2004;​145(2):592–603.
References 263

142. Murono EP, Derk RC, de Leon JH. Differential 155. Nashev LG, Schuster D, Laggner C et al. The UV-filter
effects of octylphenol, 17beta-estradiol, endosulfan, benzophenone-1 inhibits 17beta-hydroxysteroid
or bisphenol A on the steroidogenic competence dehydrogenase type 3: Virtual screening as a strategy
of cultured adult rat Leydig cells. Reprod Toxicol. to identify potential endocrine disrupting chemicals.
2001;15(5):551–560. Biochem Pharmacol. 79(8):1189–1199.
143. Nakamura D, Yanagiba Y, Duan Z et al. Bisphenol A 156. Rune GM, de Souza P, Krowke R, Merker HJ,
may cause testosterone reduction by adversely affect- Neubert D. Morphological and histochemical pat-
ing both testis and pituitary systems similar to estra- tern of response in rat testes after administration of
diol. Toxicol Lett. 2010;194(1–2):16–25. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Histol
144. Gilibili RR, Vogl AW, Chang TK, Bandiera SM. Histopathol. 1991;6(4):459–467.
Localization of cytochrome P450 and related 157. Johnson L, Dickerson R, Safe SH, Nyberg CL,
enzymes in adult rat testis and downregulation Lewis  RP, Welsh TH Jr. Reduced Leydig cell vol-
by estradiol and bisphenol A. Toxicol Sci. 2014;​ ume and function in adult rats exposed to 2,3,7,​
140(1):26–39. 8-tetrachlo​ rodibenzo-p-dioxin without a signifi-
145. Chouhan S, Yadav SK, Prakash J et al. Increase in cant effect on spermatogenesis. Toxicology. 1992;​
the expression of inducible nitric oxide synthase on 76(2):103–118.
exposure to bisphenol A: A possible cause for decline 158. Dhanabalan S, Mathur PP, Latha P. TCDD and cor-
in steroidogenesis in male mice. Environ Toxicol ticosterone on testicular steroidogenesis and anti-
Pharmacol. 2015;39(1):405–416. oxidant system of epididymal sperm in rats. Toxicol
146. Zhou W, Liu J, Liao L, Han S. Effect of bisphenol A Ind Health. 2013;31(9):811–822.
on steroid hormone production in rat ovarian theca- 159. Adamsson A, Simanainen U, Viluksela M, Paranko
interstitial and granulosa cells. Mol Cell Endocrinol. J, Toppari J. The effects of 2,3,7,8-tetrachlorod-
2008;283(1–2):12–18. ibenzo-p-dioxin on foetal male rat steroidogenesis.
147. Adu-Kumi S, Kares R, Literak J et al. Levels and Int J Androl. 2009;32(5):575–585.
seasonal variations of organochlorine pesticides in 160. Lai KP, Wong MH, Wong CK. Inhibition of
urban and rural background air of southern Ghana. CYP450scc expression in dioxin-exposed rat Leydig
Environ Sci Pollut Res Int. 2012;19(6):1963–1970. cells. J Endocrinol. 2005;185(3):519–527.
148. Niwa T, Fujimoto M, Kishimoto K, Yabusaki Y, 161. Fukuzawa NH, Ohsako S, Wu Q et al. Testicular
Ishibashi F, Katagiri M. Metabolism and interac- cytochrome P450scc and LHR as possible targets
tion of bisphenol A in human hepatic cytochrome of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)
P450 and steroidogenic CYP17. Biol Pharm Bull. in the mouse. Mol Cell Endocrinol. 2004;221(1–2):​
2001;24(9):1064–1067. 87–96.
149. Guo J, Yuan X, Qiu L et al. Inhibition of human and 162. Sathish Kumar T, Sugantha Priya E, Raja Singh P,
rat 11beta-hydroxysteroid dehydrogenases activities Arunakaran J. Lactational exposure of polychlo-
by bisphenol A. Toxicol Lett. 2012;215(2):​126–130. rinated biphenyls downregulates critical genes in
150. Eladak S, Grisin T, Moison D et al. A new chapter Leydig cells of F1 male progeny (PND21). Andrologia.
in the bisphenol A story: Bisphenol S and bisphenol 2016;49(8):e12734.
F are not safe alternatives to this compound. Fertil 163. Murugesan P, Muthusamy T, Balasubramanian K,
Steril. 2014;103(1):11–21. Arunakaran J. Polychlorinated biphenyl (Aroclor
151. Ullah H, Jahan S, Ain QU, Shaheen G, Ahsan N. 1254) inhibits testosterone biosynthesis and anti-
Effect of bisphenol S exposure on male reproductive oxidant enzymes in cultured rat Leydig cells. Reprod
system of rats: A histological and biochemical study. Toxicol. 2008;25(4):447–454.
Chemosphere. 2016;152:383–391. 164. Murugesan P, Balaganesh M, Balasubramanian K,
152. Feng Y, Yin J, Jiao Z, Shi J, Li M, Shao B. Bisphenol Arunakaran J. Effects of polychlorinated biphenyl
AF may cause testosterone reduction by directly (Aroclor 1254) on steroidogenesis and antioxidant
affecting testis function in adult male rats. Toxicol system in cultured adult rat Leydig cells. J Endocrinol.
Lett. 2012;211(2):201–209. 2007;192(2):325–338.
153. Roelofs MJ, van den Berg M, Bovee TF, Piersma AH, 165. Yamamoto M, Narita A, Kagohata M, Shirai M,
van Duursen MB. Structural bisphenol analogues Akahori F, Arishima K. Effects of maternal expo-
differentially target steroidogenesis in murine MA-10 sure to 3,3’,4,4’,5-pentachlorobiphenyl (PCB126) or
Leydig cells as well as the glucocorticoid receptor. 3,3’,4,4’,5,5’-hexachlorobiphenyl (PCB169) on testic­
Toxicology. 2015;329:10–20. ular steroidogenesis and spermatogenesis in male
154. Rodriguez-Bernaldo de Quiros A, Paseiro-Cerrato offspring rats. J Androl. 2005;26(2):205–214.
R, Pastorelli S, Koivikko R, Simoneau C, Paseiro- 166. Andric N, Kostic T, Kaisarevic S, Fa S, Pogrmic K,
Losada P. Migration of photoinitiators by gas phase Kovacevic R. In vivo and in vitro effects of PCB126
into dry foods. J Agric Food Chem. 2009;57(21):​ and PCB153 on rat testicular androgenesis. Environ
10211–10215. Toxicol Pharmacol. 2008;25(2):222–226.
264  Environmental toxicants on Leydig cell function

167. Fukuzawa NH, Ohsako S, Nagano R et al. Effects 180. Tsujita M, Ichikawa Y. Substrate-binding region of
of 3,3’,4,4’,5-pentachlorobiphenyl, a coplanar poly- cytochrome P-450SCC (P-450 XIA1). Identification
chlorinated biphenyl congener, on cultured neonatal and primary structure of the cholesterol binding
mouse testis. Toxicol In Vitro. 2003;17(3):259–269. region in cytochrome P-450SCC. Biochim Biophys
168. Kim IS, Ariyaratne HB, Chamindrani Mendis- Acta. 1993;1161(2–3):124–130.
Handagama SM. Effects of continuous and intermit- 181. Hu GX, Zhao B, Chu Y et al. Effects of methoxychlor
tent exposure of lactating mothers to aroclor 1242 on and 2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroethane
testicular steroidogenic function in the adult male on 3beta-hydroxysteroid dehydrogenase and 17beta-
offspring. Tissue Cell. 2001;33(2):169–177. hydroxysteroid dehydrogenase-3 activities in human
169. Kalayou S, Granum C, Berntsen HF et al. Label- and rat testes. Int J Androl. 2011;34(2):138–144.
free based quantitative proteomics analysis of pri- 182. Ye L, Chen X, Li X et al. Effects of methoxychlor
mary neonatal porcine Leydig cells exposed to and its metabolite 2,2-bis(p-hydroxyphenyl)-1,1,1-
the  persistent contaminant 3-methylsulfonyl-DDE. trichloroethane on human and rat 17alpha-
J Proteomics. 2015;137:68–82. hydroxylase/17,20-lyase activity. Toxicol Lett. 2014;​
170. Castellanos CG, Sorvik IB, Tanum MB, Verhaegen 225(3):407–412.
S, Brandt I, Ropstad E. Differential effects of the 183. Guo J, Deng H, Li H et al. Effects of methoxychlor
persistent DDT metabolite methylsulfonyl-DDE in and its metabolite 2,2-bis(p-mhydroxyphenyl)-1,1,1-
non­s timulated and LH-stimulated neonatal por- trichloroethane on 11beta-hydroxysteroid dehydro-
cine Leydig cells. Toxicol Appl Pharmacol. 2013;267(3):​ genase activities in vitro. Toxicol Lett. 2013;218(1):​
247–255. 18–23.
171. Ye L, Guo J, Ge RS. Environmental pollutants 184. Shivanandappa T, Krishnakumari MK. Hexachloro​
and hydroxysteroid dehydrogenases. Vitam Horm. cyclohexane-induced testicular dysfunction in rats.
2014;94:349–390. Acta Pharmacol Toxicol (Copenh). 1983;52(1):12–17.
172. Ye L, Su ZJ, Ge RS. Inhibitors of testosterone biosyn- 185. Dalsenter PR, Faqi AS, Webb J, Merker HJ, Chahoud
thetic and metabolic activation enzymes. Molecules. I. Reproductive toxicity and tissue concentra-
2011;16(12):9983–10001. tions of lindane in adult male rats. Hum Exp Toxicol.
173. Akingbemi BT, Ge RS, Klinefelter GR, Gunsalus GL, 1996;15(5):​406–410.
Hardy MP. A metabolite of methoxychlor, 2,2-bis(p- 186. Ronco AM, Valdes K, Marcus D, Llanos M. The
hydroxyphenyl)-1,1, 1-trichloroethane, reduces tes- mechanism for lindane-induced inhibition of ste-
tosterone biosynthesis in rat leydig cells through roidogenesis in cultured rat Leydig cells. Toxicology.
suppression of steady-state messenger ribonucleic 2001;159(1–2):99–106.
acid levels of the cholesterol side-chain cleavage 187. Walsh LP, Stocco DM. Effects of lindane on steroido-
enzyme. Biol Reprod. 2000;62(3):571–578. genesis and steroidogenic acute regulatory protein
174. Gray LE, Jr., Ostby J, Ferrell J et al. A dose-response expression. Biol Reprod. 2000;63(4):1024–1033.
analysis of methoxychlor-induced alterations of 188. Wango EO, Onyango DW, Odongo H, Okindo E,
reproductive development and function in the rat. Mugweru J. In vitro production of testosterone and
Fundam Appl Toxicol. 1989;12(1):92–108. plasma levels of luteinising hormone, testosterone
175. Okazaki K, Okazaki S, Nishimura S et al. A repeated and cortisol in male rats treated with heptachlor.
28-day oral dose toxicity study of methoxychlor in Comp Biochem Physiol C Pharmacol Toxicol Endocrinol.
rats, based on the ‘enhanced OECD test guideline 1997;118(3):381–386.
407’ for screening endocrine-disrupting chemicals. 189. Sircar S, Lahiri P. Effect of lindane on mitochondrial
Arch Toxicol. 2001;75(9):513–521. side-chain cleavage of cholesterol in mice. Toxicology.
176. Murono EP, Derk RC, Akgul Y. In vivo exposure 1990;61(1):41–46.
of young adult male rats to methoxychlor reduces 190. Viswanath G, Chatterjee S, Dabral S, Nanguneri SR,
serum testosterone levels and ex vivo Leydig cell Divya G, Roy P. Anti-androgenic endocrine disrupt-
testosterone formation and cholesterol side-chain ing activities of chlorpyrifos and piperophos. J Steroid
cleavage activity. Reprod Toxicol. 2006;21(2):​148–153. Biochem Mol Biol. 2010;120(1):22–29.
177. Chen B, Chen D, Jiang Z et al. Effects of estradiol 191. Kumar V, Balomajumder C, Roy P. Disruption of
and methoxychlor on Leydig cell regeneration in the LH-induced testosterone biosynthesis in testicular
adult rat testis. Int J Mol Sci. 2014;15(5):7812–7826. Leydig cells by triclosan: Probable mechanism of
178. Liu S, Li C, Wang Y et al. In utero methoxychlor action. Toxicology. 2008;250(2–3):124–131.
exposure increases rat fetal Leydig cell number but 192. Manfo FP, Chao WF, Moundipa PF, Pugeat M, Wang
inhibits its function. Toxicology. 2016;370:31–40. PS. Effects of maneb on testosterone release in male
179. Murono EP, Derk RC. The effects of the reported rats. Drug Chem Toxicol. 2011;34(2):120–128.
active metabolite of methoxychlor, 2,2-bis(p- 193. Rajeswary S, Kumaran B, Ilangovan R et al.
hydroxyphenyl)-1,1,1-trichloroethane, on testoster- Modulation of antioxidant defense system by the
one formation by cultured Leydig cells from young environmental fungicide carbendazim in Leydig
adult rats. Reprod Toxicol. 2004;19(1):135–146. cells of rats. Reprod Toxicol. 2007;24(3–4):371–380.
References 265

194. Adedara IA, Vaithinathan S, Jubendradass R, 208. Chapin RE, Phelps JL, Somkuti SG, Heindel JJ, Burka
Mathur PP, Farombi EO. Kolaviron prevents car- LT. The interaction of Sertoli and Leydig cells in the
bendazim-induced steroidogenic dysfunction and testicular toxicity of tri-o-cresyl phosphate. Toxicol
apoptosis in testes of rats. Environ Toxicol Pharmacol. Appl Pharmacol. 1990;104(3):483–495.
2013;35(3):444–453. 209. Chen G, Zhang S, Jin Y et al. TPP and TCEP induce
195. Ellis MK, Richardson AG, Foster JR et al. The oxidative stress and alter steroidogenesis in TM3
reproductive toxicity of molinate and metabolites Leydig cells. Reprod Toxicol. 2015;57:100–110.
to the male rat: Effects on testosterone and sperm 210. Henry MJ, Sisler HD. Effects of sterol biosynthesis-
morphology. Toxicol Appl Pharmacol. 1998;151(1):​ inhibiting (SBI) fungicides on cytochrome P-450
22–32. oxygenations in fungi. Pesticide Biochem Physiol.
196. X, Mao B, Dong Y, Li Y et al. Effects of ziram on rat 1984;22:262–275.
and human 11beta-Hydroxysteroid dehydrogenase 211. Walsh LP, Kuratko CN, Stocco DM. Econazole and
isoforms. Chem Res Toxicol. 2016;29(3):398–405. miconazole inhibit steroidogenesis and disrupt ste-
197. Atanasov AG, Tam S, Rocken JM, Baker ME, roidogenic acute regulatory (StAR) protein expres-
Odermatt A. Inhibition of 11 beta-hydroxysteroid sion post-transcriptionally. J Steroid Biochem Mol Biol.
dehydrogenase type 2 by dithiocarbamates. Biochem 2000;75(4–5):229–236.
Biophys Res Commun. 2003;308(2):257–262. 212. Liu HC, Zhu D, Wang C et al. Effects of etomidate
198. Chung JY, Kim YJ, Kim JY et al. Benzo[a]pyrene on the steroidogenesis of rat immature Leydig cells.
reduces testosterone production in rat Leydig cells PLoS One. 2015;10(11):e0139311.
via a direct disturbance of testicular steroido- 213. De Coster R, Wouters W, Beerens D et al. Comparative
genic machinery. Environ Health Perspect. 2011;119 effects of etomidate and its fluoro analogue, R 8110
(11):1569–1574. on testicular, adrenal and ovarian steroid biosynthe-
199. Georgellis A, Rydstrom J. Cell-specific metabolic sis. J Vet Pharmacol Ther. 1988;11(4):345–353.
activation of 7,12-dimethylbenz[a]anthracene in rat 214. Couto JA, Saraiva KL, Barros CD et al. Effect of
testis. Chem Biol Interact. 1989;72(1–2):65–78. chronic treatment with Rosiglitazone on Leydig cell
200. Mandal PK, McDaniel LR, Prough RA, Clark BJ. steroidogenesis in rats: In vivo and ex vivo studies.
7,12-Dimethylbenz[a]anthracene inhibition of ste- Reprod Biol Endocrinol. 2010;8:13.
roid production in MA-10 mouse Leydig tumor cells 215. Blystone CR, Lambright CS, Howdeshell KL et
is not directly linked to induction of CYP1B1. Toxicol al. Sensitivity of fetal rat testicular steroidogen-
Appl Pharmacol. 2001;175(3):200–208. esis to maternal prochloraz exposure and the
201. Verma R, Mohanty B. Early-life exposure to dimethoate-­ underlying mechanism of inhibition. Toxicol Sci.
induced reproductive toxicity: Evaluation of effects 2007;97(2):512–519.
on pituitary-testicular axis of mice. Toxicol Sci. 216. Blystone CR, Furr J, Lambright CS et al. Prochloraz
2009;112(2):450–458. inhibits testosterone production at dosages below
202. Astiz M, Hurtado de Catalfo G, de Alaniz MJ, Marra those that affect androgen-dependent organ weights
CA. Exogenous arachidonate restores the dimethoate-​ or the onset of puberty in the male Sprague Dawley
induced inhibition of steroidogenesis in rat intersti- rat. Toxicol Sci. 2007;97(1):65–74.
tial cells. Lipids. 2012;47(6):557–569. 217. Laier P, Metzdorff SB, Borch J et al. Mechanisms
203. Astiz M, Hurtado de Catalfo GE, de Alaniz MJ, of action underlying the antiandrogenic effects of
Marra CA. Involvement of lipids in dimethoate- the fungicide prochloraz. Toxicol Appl Pharmacol.
induced inhibition of testosterone biosynthesis in rat 2006;213(2):160–171.
interstitial cells. Lipids. 2009;44(8):703–718. 218. Ohlsson A, Ulleras E, Oskarsson A. A bipha-
204. Walsh LP, Webster DR, Stocco DM. Dimethoate sic effect of the fungicide prochloraz on aldoste-
inhibits steroidogenesis by disrupting transcription rone, but not cortisol, secretion in human adrenal
of the steroidogenic acute regulatory (StAR) gene. H295R cells—Underlying mechanisms. Toxicol Lett.
J Endocrinol. 2000;167(2):253–263. 2009;191(2–3):174–180.
205. Krause W, Homola S. Alterations of the seminifer- 219. Taxvig C, Hass U, Axelstad M et al. Endocrine-
ous epithelium and the Leydig cells of the mouse tes- disrupting  activities in vivo of the fungicides tebu-
tis after the application of dichlorvos (DDVP). Bull conazole and epoxiconazole. Toxicol Sci. 2007;100(2):​
Environ Contam Toxicol. 1974;11(5):429–433. 464–473.
206. Krause W, Hamm K, Weissmuller J. Damage 220. Blystone CR, Lambright CS, Furr J, Wilson VS,
to spermatogenesis in juvenile rat treated with Gray LE Jr. Iprodione delays male rat pubertal devel-
DDVP and malathion. Bull Environ Contam Toxicol. opment, reduces serum testosterone levels, and
1976;15(4):458–462. decreases ex vivo testicular testosterone production.
207. Ali FA, Abdalla MH. Pathological changes in tes- Toxicol Lett. 2007;174(1–3):74–81.
tes and liver of male albino rats after dermal expo- 221. Kano Y, Natori S. Change in free amino acids and
sure to DDVP insecticide. J Egypt Public Health Assoc. phospholipids in the head of adult Sarcophaga per-
1992;67(5–6):565–578. egrina with age. Differentiation. 1983;24(1):9–12.
266  Environmental toxicants on Leydig cell function

222. Rao KS, Burek JD, Murray FJ et al. Toxicologic and repro- 237. Sen Gupta R, Sen Gupta E, Dhakal BK, Thakur AR,
ductive effects of inhaled 1,2-dibromo-3-­chloropropane Ahnn J. Vitamin C and vitamin E protect the rat tes-
in rats. Fundam Appl Toxicol. 1983;3(2):104–110. tes from cadmium-induced reactive oxygen species.
223. Shemi D, Marx Z, Kaplanski J, Potashnik G, Sod- Mol Cells. 2004;17(1):132–139.
Moriah UA. Testicular damage development in 238. Ji X, Li Z, Chen H et al. Cytotoxic mechanism related
rats injected with dibromochloropropane (DBCP). to dihydrolipoamide dehydrogenase in Leydig cells
Andrologia. 1988;20(4):331–337. exposed to heavy metals. Toxicology. 2015;334:22–32.
224. Potashnik G, Yanai-Inbar I, Sacks MI, Israeli R. 239. Liu MY, Lai HY, Yang BC, Tsai ML, Yang HY,
Effect of dibromochloropropane on human testicu- Huang  BM. The inhibitory effects of lead on ste-
lar function. Isr J Med Sci. 1979;15(5):438–442. roidogenesis in MA-10 mouse Leydig tumor cells.
225. Egnatz DG, Ott MG, Townsend JC, Olson RD, Johns Life Sci. 2001;68(8):849–859.
DB. DBCP and testicular effects in chemical workers: 240. Chowdhury AR. Spermatogenic and steroidogenic
An epidemiological survey in Midland, Michigan. impairment after chromium treatment in rats. Indian
J Occup Med. 1980;22(11):727–732. J Exp Biol. 1995;33(7):480–484.
226. Potashnik G, Yanai-Inbar I. Dibromochloropropane 241. Das J, Kang MH, Kim E, Kwon DN, Choi YJ, Kim
(DBCP): An 8-year reevaluation of testicular func- JH. Hexavalent chromium induces apoptosis in male
tion and reproductive performance. Fertil Steril. somatic and spermatogonial stem cells via redox
1987;47(2):317–323. imbalance. Sci Rep. 2015;5:13921.
227. Ahmad N, Wisner JR, Jr., Warren DW. Morphological 242. Ng TB, Liu WK. Toxic effect of heavy metals on cells
and biochemical changes in the adult male rat isolated from the rat adrenal and testis. In Vitro Cell
reproductive system following long-term treat- Dev Biol. 1990;26(1):24–28.
ment with 1,2-dibromo-3-chloropropane. Anat Rec. 243. Penna S, Pocino M, Marval MJ, Lloreta J, Gallardo L,
1988;222(4):340–349. Vila J. Modifications in rat testicular morphology
228. Kelce WR, Raisbeck MF, Ganjam VK. Gonadotoxic and increases in IFN-gamma serum levels by the oral
effects of 2-hexanone and 1,2-dibromo-3-­ administration of subtoxic doses of mercuric chlo-
chloropropane on the enzymatic activity of rat tes- ride. Syst Biol Reprod Med. 2009;55(2):69–84.
ticular 17 alpha-hydroxylase/C17,20-lyase. Toxicol 244. Chiou TJ, Chu ST, Tzeng WF, Huang YC, Liao CJ.
Lett. 1990;52(3):331–338. Arsenic trioxide impairs spermatogenesis via reduc-
229. Takiguchi M, Yoshihara S. New aspects of cad- ing gene expression levels in testosterone synthesis
mium as endocrine disruptor. Environ Sci. 2006;13(2):​ pathway. Chem Res Toxicol. 2008;21(8):1562–1569.
107–116. 245. Sanghamitra S, Hazra J, Upadhyay SN, Singh RK,
230. Zhang Q, Zou P, Zhan H et al. Dihydrolipoamide Amal RC. Arsenic induced toxicity on testicular tis-
dehydrogenase and cAMP are associated with sue of mice. 2008;52(1):84–90.
cadmium-mediated Leydig cell damage. Toxicol Lett. 246. Mukhopadhyay PK, Dey A, Mukherjee S, Pradhan
2011; 205(2):183–189. NK. The effect of coadministration of alpha-­
231. Danielsson BR, Dencker L, Lindgren A, Tjalve H. tocopherol and ascorbic acid on arsenic trioxide-
Accumulation of toxic metals in male reproduction induced testicular toxicity in adult rats. J Basic Clin
organs. Arch Toxicol Suppl. 1984;7:177–180. Physiol Pharmacol. 2015;24(4):245–253.
232. Blanco A, Moyano R, Vivo J et al. Quantitative 247. Bhosle NB, Garg A, Jadhav S et al. Butyltins in water,
changes in the testicular structure in mice exposed biofilm, animals and sediments of the west coast of
to low doses of cadmium. Environ Toxicol Pharmacol. India. Chemosphere. 2004;57(8):897–907.
2007;23(1):96–101. 248. McVey MJ, Cooke GM. Inhibition of rat testis
233. Blanco A, Moyano MR, Molina AM et al. Quantitative microsomal 3beta-hydroxysteroid ­dehydrogenase acti­
study of Leydig cell populations in mice exposed to vity by tributyltin. J Steroid Biochem Mol Biol. 2003;​
low doses of cadmium. Bull Environ Contam Toxicol. 86(1):99–105.
2009;82(6):756–760. 249. Ohno S, Nakajima Y, Nakajin S. Triphenyltin and tri-
234. Rajendar B, Bharavi K, Rao GS et al. Protective effect butyltin inhibit pig testicular 17beta-hydroxysteroid
of alpha-tocopheral on biochemical and histological dehydrogenase activity and suppress testicular tes-
alterations induced by cadmium in rat testes. Indian J tosterone biosynthesis. Steroids. 2005;70(9):645–651.
Physiol Pharmacol. 2012;55(3):213–220. 250. Doering DD, Steckelbroeck S, Doering T,
235. Khanna S, Mitra S, Lakhera PC, Khandelwal S. Klingmuller D. Effects of butyltins on human
N-acetylcysteine effectively mitigates cadmium- 5alpha-reductase type 1 and type 2 activity. Steroids.
induced oxidative damage and cell death in Leydig 2002;67(10):859–867.
cells in vitro. Drug Chem Toxicol. 2015;39(1):74–80. 251. Lo S, Allera A, Albers P et al. Dithioerythritol
236. Yang JM, Arnush M, Chen QY, Wu XD, Pang  B, (DTE) prevents inhibitory effects of triphenyltin
Jiang XZ. Cadmium-induced damage to pri- (TPT) on the key enzymes of the human sex ste-
mary cultures of rat Leydig cells. Reprod Toxicol. roid hormone metabolism. J Steroid Biochem Mol Biol.
2003;17(5):553–560. 2003;84(5):569–576.
References 267

252. Yu ZH, Chan HC. Gossypol as a male antifertility 265. Liu Q, Wang Y, Gu J et al. Zearalenone inhibits tes-
agent—Why studies should have been continued. tosterone biosynthesis in mouse Leydig cells via the
1998;21(1):2–7. crosstalk of estrogen receptor signaling and orphan
253. Comhaire FH. Male contraception: Hormonal, nuclear receptor Nur77 expression. Toxicol In Vitro.
mechanical and other. Hum Reprod. 1994;9(4):​ 2014;28(4):647–656.
586–590. 266. Wang Y, Zheng W, Bian X et al. Zearalenone induces
254. Gui-Yuan Z, Meng-Chun J, Jin-Lai C, Wen-Qing Y. apoptosis and cytoprotective autophagy in primary
The effect of long-term treatment with crude cotton Leydig cells. Toxicol Lett. 2014;226(2):182–191.
seed oil on pituitary and testicular function in men. 267. Lin P, Chen F, Sun J et al. Mycotoxin zearalenone
Int J Androl. 1989;12(6):404–410. induces apoptosis in mouse Leydig cells via an endo-
255. Mushtaq M, Kulp S, Chang W, Lin YC. Gossypol plasmic reticulum stress-dependent signalling path-
inhibits human chorionic gonadotropin-stimulated way. Reprod Toxicol. 2015;52:71–77.
testosterone production by cultured canine testicular 268. Li Y, Zhang B, Huang K et al. Mitochondrial pro-
interstitial cells. Res Commun Mol Pathol Pharmacol. teomic analysis reveals the molecular mechanisms
1996;91(3):259–272. underlying reproductive toxicity of zearalenone in
256. Donaldson A, Sufi SB, Jeffcoate SL. Inhibition MLTC-1 cells. Toxicology. 2014;324:55–67.
by gossypol of testosterone production by mouse 269. Savard C, Nogues P, Boyer A, Chorfi Y. Prevention of
Leydig cells in vitro. Contraception. 1985;31(2):​ deoxynivalenol- and zearalenone-associated oxida-
165–171. tive stress does not restore MA-10 Leydig cell func-
257. Sufi SB, Donaldson A, Jeffcoate SL, Matlin SA, Zhou tions. Toxicology. 2016;341–343:17–27.
RH. Inhibition by (+) and (-) isomers of gossypol of 270. Yang J, Zhang Y, Wang Y, Cui S. Toxic effects of zeara-
testosterone release from mouse Leydig cells in vitro. lenone and alpha-zearalenol on the regulation of ste-
Contraception. 1985;31(2):159–164. roidogenesis and testosterone production in mouse
258. Pearce S, Sufi SB, O’Shaughnessy PJ, Donaldson A, Leydig cells. Toxicol In Vitro. 2007;21(4):558–565.
Jeffcoate SL. Inhibition by gossypol of cyclic AMP 271. Egbunike GN. Steroidogenic and spermatogenic
production in mouse Leydig cells. Contraception. potentials of the male rat after acute treatment with
1986;34(6):639–646. aflatoxin B1. Andrologia. 1982;14(5):440–446.
259. Cuellar A, Diaz-Sanchez V, Ramirez J. Cholesterol 272. Adedara IA, Nanjappa MK, Farombi EO, Akingbemi
side-chain cleavage and 11 beta-hydroxylation are BT. Aflatoxin B1 disrupts the androgen biosynthetic
inhibited by gossypol in adrenal cortex mitochon- pathway in rat Leydig cells. Food Chem Toxicol. 2014;
dria. J Steroid Biochem Mol Biol. 1990;37(4):581–585. 65:252–259.
260. Hu GX, Zhou HY, Li XW et al. The (+)- and (-)-­gossypols 273. Yang JY, Zhang YF, Li YX et al. Effects of T-2 toxin
potently inhibit both 3beta-hydroxysteroid dehydro- on the regulation of steroidogenesis in mouse Leydig
genase and 17beta-hydroxysteroid dehydrogenase 3 cells. Toxicol Ind Health. 2015;32(10):​1801–1807.
in human and rat testes. J Steroid Biochem Mol Biol. 274. Yang JY, Zhang YF, Meng XP, Li YX, Ma KW, Bai
2009;115(1–2):14–19. XF. T-2 toxin inhibits gene expression and activity of
261. Hiipakka RA, Zhang HZ, Dai W, Dai Q, Liao S. key steroidogenesis enzymes in mouse Leydig cells.
Structure-activity relationships for inhibition of Toxicol In Vitro. 2015;29(5):1166–1171.
human 5alpha-reductases by polyphenols. Biochem 275. Yang J, Zhang Y, Jing A, Ma K, Gong Q, Qin C.
Pharmacol 2002;63(6):1165–1176. Effects of T-2 toxin on testosterone biosynthesis in
262. Chen BB, Lin H, Hu GX, Su Y, Zhou HY, Lian QQ mouse Leydig cells. Toxicol Ind Health. 2012;30(10):​
et al. The (+)- and (-)-gossypols potently inhibit 873–877.
human and rat 11beta-hydroxysteroid dehydroge- 276. Liu S, Wang D, Zhang J et al. Citrinin reduces testos-
nase type 2. J Steroid Biochem Mol Biol, 2009;113 terone secretion by inducing apoptosis in rat Leydig
(3–5):177–181. cells. Toxicol In Vitro. 2012;26(6):856–861.
263. Svechnikov K, Supornsilchai V, Strand ML, Wahlgren 277. Kalayou S, Ndossi D, Frizzell C et al. An inves-
A, Seidlova-Wuttke D, Wuttke W et al. Influence of tigation of the endocrine disrupting potential of
long-term dietary administration of procymidone, a enniatin B using in vitro bioassays. Toxicol Lett.
fungicide with anti-androgenic effects, or the phy- 2015;233(2):84–94.
toestrogen genistein to rats on the pituitary-gonadal 278. Murono EP, Lin T, Osterman J, Nankin HR. The
axis and Leydig cell steroidogenesis. J Endocrinol effects of cytochalasin B on testosterone synthesis
2005; 187(1):117–124. by interstitial cells of rat testis. Biochim Biophys Acta.
264. Hu GX, Zhao BH, Chu YH et al. Effects of genis- 1980;633(2):228–236.
tein and  equol on human and rat testicular 279. Murono EP, Lin T, Osterman J, Nankin HR.
3beta-hydroxysteroid dehydrogenase and 17beta- Relationship between inhibition of interstitial cell
hydroxysteroid dehydrogenase 3 activities. Asian J testosterone synthesis by cytochalasin B and glucose.
Androl. 12(4):​519–526. Biochem Biophys Res Commun. 1982;104(1):​299–306.
Index
Page numbers followed by f and t indicate figures and tables, respectively.

A androgen receptor functional HIV and, 185 Autophagy-related gene 7 (Atg7),


Abdominal examination, 139 domains, 40 Antisperm antibodies (ASA), 15
Acquired immune deficiency cell- and temporal-specific 63, 126 Autoregulatory UBA domain,
syndrome, 63 expression of AR, 43 Antithyroid agents, 235–236; 227
Acrosome development, 193 spermatogenesis, 40 see also Endocrine Axial elements (AE), 107
Acrosome reaction, 127 testis gene expression disruption Axl receptor, 55; see also Tyro3,
Actin, 191 is regulated by mechanisms Axl, and Mer (TAM)
crossing linking proteins, 160 testosterone, 46–47 Antiviral proteins, 57–58 receptors
microfilaments, 215 testosterone and AR are AP1, 24 α-Zearalenol, 257
monomers, 191 required to complete AP2, 24 Azole compounds, 255
Actin-based cytoskeletons, meiosis, 44–45 Apale spermatogonia, 124 Azoospermia; see also Fertility/
174f, 175; see also testosterone and number of Apermatogenesis associated 3 infertility in humans,
Spermiation gonocytes, 43–44 (SPATA3), 221 regulation of
Actin-like protein 7A (ACTL7A), testosterone is required for Apical tubulobulbar complex about, 125–126
222 spermatid attachment (apical TBC), 172 acrosome reaction/
Actin-like protein 7B (ACTL7B), and release of sperm, Apoptotic germ cells, capacitation, 127
222 45 phagocytosis of, asthenozoospermia,
Actin-related protein 3 (Arp3), testosterone is required to 53–54 126–127
224 maintain the blood- Aquaporin 1 and 9 (AQP1 and DNA fragmentation, 127
Activator protein-2 (AP-2), 123 testis barrier, 45–46 AQP9), 119 etiology of, 144t
Active spermatid transport, 172 testosterone production, 42 5α-reductase (SRD5A1), 246, oligozoospermia, 126
Acyline, 85 testosterone signaling 246f, 247 pretesticular, 144
ADAM3, 10 pathways, 42–43 Argonaute proteins (AGO), 128 teratozoospermia, 127
Adark spermatogonia, 124 testosterone supports ARKO mice, 43, 44 treatment, 126
Adenovirus, 42 the expansion of Aromatase (Cyp19a1), 116, 117 Azoospermia factor (AZF), 127,
Adherens junction (AJ) spermatogonia, 44 Aromatase inhibitors (AI), 125, 128
hybrid, 175 Androgen response elements 145
proteins, 160 (AREs), 40, 234 Arpc1b, knockdown of, 196 B
testis-specific actin-rich, 171 Androgens, 116 Arp2/3 complex, 191 Barrier functions, classical JAMs
Adhesion proteins, 194 Anejaculation(AE), 147; see also Arsenic chemicals, 256 and CAR, 75–76
Adjudin 1-(2,4-dichlorobenzyl)- Male infertility ART, see Assisted reproductive Basal compartment, 184
1H-indazole-3- about, 149 techniques (ART) Basal ectoplasmic specialization
carbohydrazide, 225 and failure of emission, Arteriole smooth muscle (basal ES), 205
Adluminal compartment, 184 management of, 149 AR expression, 43 Basal inter-Sertoli cell junctions,
Adrenal chromaffin, 85–86 Angiotensin, regulation of JAMs Aryl hydrocarbon receptor 194
Adult Leydig cells, 245 and CAR, 77 (AhR), 157 Basic fibroblast growth factor
17α-ethinylestradiol (EE2), 157 Animal early development, Assisted reproductive techniques (bFGF), 97
Aflatoxins, 257 epigenetic changes (ART), 130, 130t; Β-COP (coatomer protein
AGFG1, 14 on, 157 see also Chemicals, complexers), 5
Agouti gene, 157 Annexin A1, 24 environmental Benzophenones, 253
AIP1, actin disassembly factor, Annexin A6, 11, 12, 13f, 19 motile sperm counts in, 130t 17β-estradiol (E2), 206
193 Antero-posterial (A/P) axis, 211 oligozoospermia and, 126 concentrations, 116, 117
Air pollution, on fertility, 129 Antiandrogenic chemicals, Asthenospermia; see also Male 17β-estradiol (E2) in male
A-kinase anchor protein 4 235; see also infertility reproduction, role of
(AKAP4), 221, 222, 223 Endocrine disruption causes, 142–143 environmental estrogens
Albumin, 24, 28f mechanisms management on human testicular
Amino acid sequence, 191 Antiandrogens, 252, 255 semen volume and pH, development,
Aminoglycosides, 129 Antibiotics, 129, 141t 143 119–120
AMP-activated kinase (AMPK), for fertility/infertility in postejaculate urinalysis, knockouts of ESR2 or GPER,
201 humans, 129 143 119
Anaerobic glycolysis, 101 Antibodies, to spermatic concentration and sperm loss of ESR1 causing male
Analytical ultracentrifugation antigens, 63 count, 143 reproductive defects,
(AUC), 75 Antigen-presenting cells (APCs), Asthenozoospermia, 126–127; 118
Anaphylatoxins, release of, 86 86, 88 see also Azoospermia loss of membrane ESR1
Androgen Antihypertensives, 129, 141t defined, 126 expression causing
endogenous, 235 for fertility/infertility in etiologies of, 126 male reproductive
Androgen, luteinizing hormone humans, 129 male infertility, 222 abnormalities,
(LH) and, 239; see also Anti-Ifnar1 MAb (IFNα receptor Atlastin 2, 24 118–119
Endocrine disrupting 1 monoclonal α-tubulin, 24 3β-HSD, see 3β-hydroxysteroid
chemicals (EDC) antibody), 186 Autoantigens, 184 dehydrogenase (HSD)
Androgen-binding protein (ABP), Anti-JAM-A Fab BV11, Autocrine regulation of 17β-HSD3, see
124, 237 monoclonal antibody, spermatogonia, 97–99 17β-hydroxysteroid
Androgen biosynthesis, 245–246 73, 75 Autoimmune orchitis, 59 dehydrogenase 3
Androgen receptor antagonist, Anti-Müllerian hormone experimental models of, (HSD3)
252 (AMH), 124, 63–64 3-β-hydroxysteroid
Androgen receptor- (AR-) 170, 239; see pathogenesis, 64, 65–69 dehydrogenase
mediated activity, 234 also Endocrine BTB, 65–66 (3β-HSD), 225, 245,
Androgen receptors (ARs), 156 disrupting chemicals germ cell apoptosis 246–247
Androgen regulation of (EDC) mediators, 67–69 17β-hydroxysteroid
spermatogenesis, Antioxidants, 127 immune cells, 66, 67f dehydrogenase
40–47 Antiretroviral therapy (ART) Autonomic dysreflexia (AD), 150 (17β- HSD), 225, 253

268
Index 269

17β-hydroxysteroid BTB, see Blood-testis barrier antiandrogenic Clathrin assembly protein


dehydrogenase 3 (BTB) chemicals, 235 (CALM), 16
(HSD3), 247 β-TrCP, 99 antithyroid agents, Clathrin heavy chain (CHC), 21
11β-hydroxysteroid 235–236 Claudin, 84, 85
dehydrogenase C endocrine disrupting Claudin-3, 171
isoform 1 (11β- Cadmium, 256 chemical (EDC) Claudin-11, 171, 206, 214, 215, 225
HSD1), 248 Cadmium (batteries and affection of Clusterin (CLU), 223
Binding immunoglobulin protein pigments), 129 neuroendocrine Coat protein complex II (COPII),
(BIP), 21, 24 Cadmium-based model systems, control of testicular 16
Bioinformatics 225 function, 236 Cobalt, 256
for biomarkers identified Caenorhabditis elegans (C. estrogenic chemicals, Cofilin, 191
using omics elegans), 202 234–235 Cohesins, 109
approaches, 226–227, Caenorhabditis elegans (C. expression of steroid Colony-stimulating factor 1
227t–229t elegans) model, 158 hormone receptors in (CSF-1), 97, 170
in translational research of Calmodulin protein kinase IV testicular cells, 234 Complement cascade activation,
male reproduction, (CaMKIV), 221 paracrine regulation of 86
220–221 Calnexin, 21, 24 testicular cells, 240f Computational characterization/
Biomarkers and drug targets, Calpain 11 (CAPN11), 221 Chemical pollutants on integrative analysis of
227t–229t Calspermin (CAS), 221 spermatogenesis and proteins
Biomarkers identification; see Calyx, 194 male infertility bioinformatics for
also Computational Capacitation, 127 EDC, mechanistic actions of biomarkers
characterization/ CAR, see Coxsackievirus and epigenetic changes identified using
integrative analysis of adenovirus receptor on animal early omics approaches,
proteins (CAR) development, 157 role of, 226–227,
bioinformatics for, using Carbamates, 254 receptor mediated 227t–229t
omics approaches, Carbendazim, 254 pathways, 156–157 bisphenol-A (BPA)-
226–227, 227t–229t Carbonic anhydrase II (CAR2), 119 environmental pollution and based model for
exogenous compounds-based Carboxypeptidase D, 19 reproductive health identification of
models for, 223–224 Catsper1-4, 172 human reproductive crucial drug targets,
genomic, transcriptomic, and CBAVD, 128 health, declining, 224–225
proteomic approaches C-C chemokine receptor 5 155–156 cadmium-based model
in, 221 (CCR5), 185 overview, 155 systems, 225
BIP, see Binding immunoglobulin CCDC136 (coiled-coil domain PFC, human exposure risk, exogenous compounds-
protein (BIP) containing 136), 16 and reproductive based models
Bisphenol A (BPA), 130, 156, 224, CD147, 96, 97 health for biomarkers
225, 252 Cell adhesion, function of about, 158–160 identification,
Bisphenol-A (BPA)-based model classical JAMs and perfluoroalkyl chemicals/ 223–224
for crucial drug CAR, 76 mechanistic activities, genomic, transcriptomic, and
targets identification, Cell map, 5 160 proteomic approaches
224–225 Cell mediated immune response, reproductive toxicity of EDC in biomarkers
Bisphenols, 252–253 88 from laboratory identification, 221
2-bis-(p-hydroxyphenyl)- Cell polarity, 211 animal and cell studies genomic and transcriptomic
trichloroethane function of classical JAMs hypothalamic-pituitary biomarkers, 221–222
(HPTE), 238 and CAR, 76 gonadal axis, 157–158 male contraceptive-based
Blood-testis barrier (BTB), 40, Cell signaling for testosterone spermatogenesis, 158 model, 225–226
42, 95, 124, 158, 168, biosynthesis in Leydig Chemicals, environmental; omics/bioinformatics
171, 185, 194, 205; cells, 247–248 see also Fertility/ methods in
see also Germ cell Cellular metabolism, 101 infertility in humans, translational research
development Celsr, 211 regulation of of male reproduction,
defined, 83 Central element (CE), 107 about, 129–130 220–221
germ cell development, 83–84 Central nervous system (CNS), 185 assisted reproductive overview, 220
integrity, disruption of, 55 Ceruloplasmin, 124 techniques (ART), proteomic markers and drug
JAMs, 73, 75 CFTR gene mutations, 128 130, 130t targets, 222–223
maintenance of testicular CGN (cis Golgi network), 5 Chemokine receptor type 4 Congenital bilateral absence
immunoprivilege, 63, Chemical exposure effects, 236 (CXCR4), 98 of vas deferens
65–66 Chemical exposures on testis cell- Chemotherapeutic agents, 129 (CBAVD), 126, 146
male germ cells, localization, cell interactions and fertility/infertility in humans, Connexin-43, 65, 160, 194, 196,
58 endocrine function 129 224
movement of germ cells EDC, classes of, 234, 235t Chlamydia spp., 89 Constitutive androstane
across, 84–85 EDC disruption of paracrine Chlamydia trachomatis, 59, 63 receptors (CAR), 156
permeability, 55 relationships between Chlorpyrifos, 254 Copper, 256
restructuring of junctions, testicular cells Cholesterol Cortactin, knockdown of, 196
73, 75 anti-Müllerian hormone chemical structures of, 247f Corticoids, for orchitis, 69
spermatogonia and (AMH), 239 transporters, 246f Coxsackievirus and adenovirus
spermatocytes on, 58 estrogen, 239 Cholesterol-transporting protein, receptor (CAR)
structure and function, 65–66 follicle-stimulating 248 functions
testis immune privilege, 85 hormone (FSH), Chromatids, 106 barrier, 75–76
testosterone to maintain, 45–46 238–239 Chromium, 256 cell adhesion, 76
Body mass index (BMI), 129 growth factors, 239 Chromosomal synapsis, 109 cell polarity, 76
Bone morphogenetic protein 4 LH and androgen, 239 Chromosomal synapsis, 107, 109; molecular structure, 73–75
(BMP4), 98, 124, 170 paracrine regulators of see also Mammalian overview, 73, 74f
Bordetella pertussis, 64, 85 testicular cells, 238t meiosis, genetics of tissue distribution and
Bortezomib, 99 EDC targeting of testicular Chromosome movement, 106 localization in testis,
Brain-derived neurotrophic cells Cigarette smokers, 256 74t, 75
factor (BDNF), 170 about, 236 cis Golgi network (CGN), 5 overview, 73
Branched actin polymerization, germ cells, 237 Citrinin, 257 regulation, in testis, 77
194 Leydig cells, 237–238 c-Kit genes, 44 CREB transcription factor, 42
Breast cancer related protein Sertoli cells, 237 c-Kit receptor, 98 Crossover formation, 110;
(BCRP), 225 endocrine disruption, Classical pathway, testosterone see also Meiotic
Breastfeeding, 159 mechanisms of signaling, 42 recombination
270 Index

Crucial drug target identification, Defensin, beta 119 (DEFB119), Ejaculatory duct obstruction Environmental estrogens on
bisphenol-A (BPA)- 222 (EDO), 143, 146 human testicular
based model for, Deleted-in-azoospermia-like Ejaculatory dysfunction; see also development, 119–120
224–225 (DAZL) gene, 222 Male infertility Environmental pollution and
Cryptorchidism, 145 Dendritic cells (DCs), 52, 64, 66 about, 147 reproductive health
CXCL12, 98 Deoxynivalenol, 257 anejaculation, 149 human reproductive health,
Cyclic-AMP-(cAMP)-dependent Desmoglein 2 (DSG2), 222 anejaculation/failure declining, 155–156
protein kinase A, 223 Desmosome (DS), 205 of emission, overview, 155
Cyclin-dependent kinases (CDK), Deubiquitinating enzymes management of, 149 Environmental toxicants on
107 (DUB), 99 retrograde ejaculation, Leydig cell
Cyclin-related protein CNTD1, 110 Diabetes mellitus (DM), 129 147–148 about, 248, 249t–251t, 250
CYP11A1, see Cytochrome P450 obesity and, 129 Electroejaculation, 150 benzophenones, 253
cholesterol side chain Diagnosis, of human Electrophoresis, two-dimensional bisphenols, 252–253
cleavage enzyme autoimmune orchitis, (2-DE), 220 carbamates, 254
(CYP11A1) 69 EM analysis, 5 1,2-dibromo-3-
CYP17A1, see Cytochrome P450 1,2-dibromo-3-chloropropane Embrel , 67–68
® chloropropane
17α-hydroxylase/17,​ (DBCP), 129, 255 Emp70 (endomembrane protein (DBCP), 255
20-lyase (CYP17A1) Dibutyl phthalate (DBP), 235 70), 7 metals, 255–256
cyp11a1 expressions, 253 Dichlorodiphenyltrichloroethane End-binding protein 1 (EB1), 175 organochlorines, 253–254
cyp19 gene, 175, 176 (DTT), 155, 253 Endocrine disrupting chemicals organophosphates, 254–255
cyp19KO, 118 Dichlorvos, 255 (EDC); see also perfluoroalkyl substances
Cysteine 451 (C451A), 118 Di(2-ethylhexyl) phthalates Chemical exposures (PFAS), 248–249
Cystic fibrosis transmembrane (DEHP), 158, 235, on testis cell-cell phthalates, 251–252
regulatory (CFTR) 251, 252 interactions and plant toxicants, 256–257
protein, 126, 146 Diethylstilbestrol (DES), 118, 156 endocrine function polycyclic aromatic
Cytidine 5-monophosphatase Differential-in-gel electrophoresis affection of neuroendocrine hydrocarbons (PAH),
(CMPase), 5 (DIGE), 220 control of testicular 254
Cytochalasin D, 196 Digital rectal examination, 143 function, 236 Enzymes for testosterone
Cytochalasins, 257 Dihydrotestosterone (DHT), 42, classes of, 234, 235t biosynthesis and
Cytochrome b5, 247 234, 245, 246 disruption of paracrine metabolism; see also
Cytochrome c oxidase subunit 6B Dimethoate, 255 relationships between Leydig cell function,
(COX6B), 223 7,12-dimethylbenz[a] anthracene testicular cells environmental
Cytochrome P450 (DMBA), 254 anti-Müllerian hormone toxicants on
17α-hydroxylase/​ Diphtheria toxin, 83 (AMH), 239 3β-HSD, 246–247
17,20-lyase Disordered spacer region, 227 estrogen, 239 17β-HSD3, 247
(CYP17A1), 245, 247 DMC1, 45, 110 follicle-stimulating CYP11A1, 246
Cytochrome P450 cholesterol DMRT, 45 hormone (FSH), CYP17A1, 247
side chain cleavage DNA-binding domain (DBD), 40 238–239 SRD5A, 247
enzyme (CYP11A1), DNA-break detection, 127 growth factors, 239 Epidermal growth-factor (EGF),
246 DNA fragmentation, 127, 128; see LH and androgen, 239 52, 53f
Cytochrome P450 (CYP) also Azoospermia; paracrine regulators of Epidermal growth factor receptor
isoforms, 160 Male infertility testicular cells, 238t substrate 8 (Eps8), 224
Cytoplasmic tails, of classical about, 150 effects of, 155 Epididymal obstruction,
JAMs, 73 management, 150 mechanistic actions of posttesticular
Cytoskeletal reorganization DNA integrity, 150 epigenetic changes azoospermia, 146, 147
genetic regulation, 176 DNA replication, 7 on animal early Epididymis, 85, 139
hormonal regulation, 175–176 Docosahexaenoic acid (DHA), 16 development, 157 Epididymitis, 59
Cytoskeletons, actin- and Doublesex- and mab-3-related receptor mediated Epithelial cycle of
MT-based, 174f, 175; transcription factor 1 pathways, 156–157 spermatogenesis1, 211
see also Spermiation (DMRT1), 222 reproductive toxicity of, from Epithelial-mesenchymal
Cytoskeletons (F-actin) and Double-strand breaks (DSB) laboratory animal transition (EMT), 97
spermatogenesis repair, 109–110; and cell studies Epoxiconazole, 255
F-actin biogenesis, dynamics, see also Meiotic hypothalamic-pituitary Epoxyeicosatrienoic acids (EET),
and function, 191–192 recombination gonadal axis, 157–158 160
F-actin in Sertoli cells Dpy19l2, 17 spermatogenesis, 158 ER-associated protein degradation
in ectoplasmic Drosophila, 99, 193, 202 targeting of testicular cells (ERAD), 10
specializations, Drosophilia, 211 about, 236 Erp44 (endoplasmic reticulum
194–195 Drug targets, proteomic markers germ cells, 237 resident protein), 10
in seminiferous tubule and, 222–223 Leydig cells, 237–238 ERP57 (ER protein 57), 23
contraction, role of, Dynamic light scattering (DLS) Sertoli cells, 237 Escherichia coli, 58–59, 63
197 analysis, 75 Endocrine disruption Esr1 knockout (Esr1KO) mouse,
Sertoli cell F-actin- mechanisms 118
containing structures, E antiandrogenic chemicals, Esr2 knockouts (Esr2KOs), 119
dynamics of (during E1 activating enzyme, 99 235 Esr1KO, 119
spermatogenesis), EAO (experimental autoimmune antithyroid agents, 235–236 Estradiol, 129
196–197 orchitis) model, 55, EDC affection of treatment, 196
in tubulobulbar complex 63–64, 66, 84 neuroendocrine Estrogen
(TBC), 195–196 Ebola virus, 88 control of testicular in EDC disruption
F-actin in spermatogenic cells ECM protein, 96 function, 236 of paracrine
in spermatogonia and E2 conjugating enzyme, 99 estrogenic chemicals, relationships, 239;
meiotic cells, 192–193 Ectoplasmic specialization (ES), 234–235 see also Endocrine
during spermiogenesis, 75, 171 Endocytic vesicle-mediated disrupting chemicals
193–194 adherans junction, 45 protein trafficking (EDC)
Cytosolic DNA sensors, 55, 57 apical, 225 event, 172 environmental, on
core PCP proteins regulating, Endoplasmic reticulum (ER), 3 human testicular
D 216f Endosomalsorting complex development, 119–120
Daf-2 gene, 202 F-actin in, 194–195 required for treatment in males, early, 118
Database for Annotation, EDC, see Endocrine disrupting transport-0 ­(ESCRT-0), Estrogenic chemicals, 234–235;
Visualization and chemicals (EDC) 13 see also Endocrine
Integrated Discovery EF1α, elongation factor, 21 Endosulfan, 254 disruption
(DAVID), 221 EGF receptor (EGFR), 42 Enniatin B, 257 mechanisms
Index 271

Estrogen receptor 1 (ESR1) in spermatogenic cells Fibroblast growth factor 9 Germ cells (GCs), 183, 237;
loss of, causing male in spermatogonia and (FGF9), 106 see also Endocrine
reproductive defects, meiotic cells, 192–193 Fibroblast growth factor receptor disrupting chemicals
118 during spermiogenesis, 3 (FGFR3), 222 (EDC)
in male reproduction, role of, 193–194 Flagella biogenesis, 194 across BTB/SCB, movement,
see 17β-estradiol (E2) F-actin biogenesis Flame retardants, 156 84–85
in male reproduction, dynamics, and function, Flavones, 257 apoptosis mediators, 67–69
role of 191–192 Fluorescence in situ hybridization generation of, 1, 2f–3f
Estrogen receptor 1 (ESR1), 117, F-actin disorganization, 214 (FISH), 127 Germ cell tumors (GCTs), 88
120 F-actin elongation, 191 Focal adhesion kinase (FAK), gfra1 gene, 101
Estrogen receptor 2 (ESR2), 117 Fads2 gene, 16 196 GIANTIN, 8–9
knockouts of, 119 FAM3C, see Family with Focal contact, 174 GL54D, 8–9, 12
Estrogen receptor 1 (ESR1) sequence similarity Focal orchitis, 64 Glial cell-derived neurotrophic
expression, loss of 3, member C Follicle-stimulating hormone factor (GDNF), 44
membrane (FAM3C) (FSH), 44, 123, production, 44
causing male reproductive Family with sequence similarity 124, 143, 157, 206, Glial cell line-derived
abnormalities, 3, member C 236, 238–239; see neurotrophic factor
118–119 (FAM3C), 11, 12, 14f also Endocrine (GDNF), 192, 193,
Estrogen receptors (ESRs), 116, Family with sequence similarity disrupting chemicals 202
156, 234 71, member F1 (EDC) Glial-derived neurotrophic factor
Estrogen receptors (ER) (FAM71F1), 221 FSCN3, 194 (GDNF), 95
expression in male Fascin protein (Fascin 1-3), 225 Functions, classical JAMs and about, 95, 96f, 97
reproductive organs, F-box protein, 99, 100 CAR role of SC and, 124
117–118 Fbxw11, 99 barrier, 75–76 treatment of, 98
Estrogen response elements Fertility cell adhesion, 76 Globozoospermia, 14
(ERE), 234 as function of age, 128 cell polarity, 76 Globular actin (G-actin), 191
Estrogens history of, 123 Fungicide (vinclozolin), 157 Glucocorticoid metabolizing
in males, 116 and pollution, 129 Fungicides, 254 enzymes, 248
Estrogen sources in Fertility/infertility in humans, Fz/Dgo/Dsh complex, 213 Glucocorticoid receptors (GR),
males; see also regulation of 156
Spermatogenesis, azoospermia G Glucose metabolism, 223
membrane/nuclear about, 125–126 Galectin-1, 88 Glucose-regulated protein 170
estrogen receptors in acrosome reaction/ Galnt3, 16 (GRP170), 23
about, 116–117 capacitation, 127 GALNTL5, 16 Glucose transporter 3 (GLUT-3),
early estrogen treatment in asthenozoospermia, Galntl5, 16 20–21
males, effects of, 118 126–127 Gametogenetin (GGN), 221 Glucose transporter 8 (GLUT-8),
estrogen receptors (ER) DNA fragmentation, 127 Gamma-aminobutyric acid 225
expression in male oligozoospermia, 126 (GABA), 124 GLUT-4, 21
reproductive organs, teratozoospermia, 127 Gap junction (GJ), 171, 205 Glutathione S transferase (GST)
117–118 treatment, 126 Gap junction (GJ) protein isoforms, 24
Etanercept (Embrel®), 67–68 environmental chemicals connexin-43 (Cx43), Glutathione S-transferase Mu 3
Ethylene dibromide (gasoline), about, 129–130 160 (GSTMu3), 223
129 assisted reproductive Gba2, 172 Glyceraldehyde- 3-phospahte
Etomidate, 255 techniques (ART), GBF1, 7–8, 12, 19 dehydrogenase
E3 ubiquitin ligase, 99 130, 130t GC1-spg cells, 101 sperm-specific, 172
Eukaryotic initiation factor 4E fertilization, requirements GDNF, see Glial cell-derived Glyceraldehyde-3-phosphate
binding protein-1 for, 125, 125t neurotrophic factor dehydrogenase
(4E-BP1), 201 genetic factors, 127–128 (GDNF) (GAPDHS), 222, 223
European Chemicals Bureau, history of fertility, 123 Gdnf transgene, 44 Glyceraldehyde-3-phosphate
224 hormonal regulation of Gene array analysis, 99 dehydrogenase, testis-
Exogenous compounds- spermatogenesis, 125 Gene knockout, 96 specific (GAPDS), 223
based models leydig cells, role of, 124–125 Gene ontology, 221 Glycolysis, 101
for biomarkers medications Generation of germ cells, 1, 2f–3f Glycoproteins, 125
identification, antibiotics, 129 Genes/nucleic acid, 100 GnRH antagonist, 42
223–224 antihypertensives, 129 Genetic anomalies, male treatment, 44
Exonuclease 1 (EXO1), 110 chemotherapeutic agents, infertility, 173t Golgi apparatus regulation of
Experimental autoimmune 129 Genetic mutations, in TAM differentiation, 1–31
orchitis (EAO) model, fertility and pollution, receptors, 52 data from other sources,
55, 63–64, 66, 84 129 Genetic regulation, cytoskeletal 14–16
Extracellular matrix (ECM) hormonal, 129 reorganization, 176 fragmentation of the Golgi
proteins, 95 psychiatric-related, 129 Genetics of mammalian meiosis, apparatus in late
Extracellular regulated kinase obesity and diabetes mellitus, see Mammalian spermatids, 18–19
(ERK), 237 129 meiosis, genetics of Golgi apparatus
pregnancy and infertility, Genistein, 257 and acrosome formation,
F epidemiology of, 123 Genomic approaches in 3–5
F-actin Sertoli cells (SC), role of, 124 biomarkers molecular dissection of, 5
in Sertoli cells spermatogenesis and identification, 221 structure and function, 5
in ectoplasmic spermiogenesis, Genomic biomarkers, 221–222 Golgi proteins
specializations, 123–124 Germ cell aromatase expression, coincidental temporal
194–195 varicoceles 117 expression of non-
in seminiferous tubule about, 128 Germ cell arrest (GA) Golgi and, 21, 23–24
contraction, role of, fertility as function of phenotypes, 221 defining in spermatids,
197 age, 128 Germ cell development; 17–18
Sertoli cell F-actin- Fetal Leydig cells, 237, 245 see also Human in early spermatids
containing Fetal male germ cells, 106 spermatogenesis during acrosome
structures, FG repeats-containing protein 1 blood-testis barrier (BTB), formation, 9–10
dynamics of (during (HRB), 14 171 forming Hermes body at
spermatogenesis), Fibroblast growth factor 2 from undifferentiated step 19 spermatids,
196–197 (FGF2), 97 spermatogonia to 19–20
in tubulobulbar complex Fibroblast growth factor 8 haploid spermatids, localized to
(TBC), 195–196 (FGF8), 98 168–171 spermatocytes, 7–9
272 Index

matching protein GPR30, 234 Human reproductive health, Immunoelectron microscopy, 107
expression profiles of G protein-coupled estrogen declining, 155–156 Immunoglobulin A (IgA), 126
Golgi and non-Golgi receptor (GPER), 117 Human Sertoli cells, 206 Immunoglobulin G (IgG), 126
proteins during knockouts of, 119 Human spermatogenesis Immunoglobulin M (IgM), 126
spermatogenesis, G-protein-coupled receptor germ cell development Immunoglobulin superfamily
20–21 (GPR), 154 blood-testis barrier (IgSF), 73
overlapping expression Granulomas, 64, 65 (BTB), 171 Immunoregulation, testicular,
profiles of non-Golgi Granzymes, 86 from undifferentiated see Testicular
and, 24–25 GRASP55, 9–10, 12 spermatogonia to immunoregulation
proteins involved in Growth and differentiation factor haploid spermatids, Immunoregulatory mechanisms,
acrosome binding to 3 (GDF3), 123 168–171 183
the nuclear surface, Growth-arrest–specific gene 6 overview, 168 Immunosuppression, in testis, 55
16–17 (Gas6), 52, 53 round spermatids to Indoleamine-pyrrole
proteins of unknown Growth factors, endocrine functional 2,3-dioxygenase
function, 20 disrupting chemicals spermatozoa, (IDO), 88
for residual bodies at step (EDC), 239 development of Infection and inflammation, 63
19 spermatids, 20 Guillain-Barré syndrome, 186 cytoskeletal Infectious orchitis, 59
segregation to acrosome, reorganization, Infertility
12 H 175–176 etiological factors, 63, 69
Golgi role in chromatoid Haploid spermatids, 168–171; spermiation, 172, 174–175 in humans, regulation of, see
body and MVB see also Germ cell spermiogenesis, 171–172, Fertility/infertility in
formation, 13–14 development 173t humans, regulation of
introduction, 1 Heat shock proteins (HSPs), 54 Human T-cell leukemia virus male, see Male infertility
spermatogenesis, 1–3 Heat shock-related 70 kDa type 1 (HTLV-1), 188 pregnancy and, epidemiology
defined, 1 protein 2 (HSPA2), Human tubal fluid (HTF), 150 of, 123
spermiogenesis, 3 223 Humoral immune response, Inflammation, spermatogenesis
unexpected ER resident Heavy metals, 255 86–88 and, 63–69
proteins with Hedgehog signaling, 98 HUWE1 protein, 100 antibodies to spermatic
Golgi/ acrosome HEI10, 110 Hydroxychlor, 254 antigens, 63
localizations, 10 Hemivasotomy, 147t Hydroxyeicosatetraenoic acids autoimmune orchitis
unexpected proteins with Hepatitis virus, 58 (HETEs), 160 BTB, 65–66
Golgi/ acrosome Hermes body, 5, 6f, 10, 19, 20 Hypophysectomy, 44 experimental models,
localizations, 11–12 Heterophilic trans-interaction, Hypoplastic seminal vesicles, 143 63–64
Golgi cisternae, 8, 9 JAMs, 73, 74t, 75 Hypospermatogenesis, 126, 144, germ cell apoptosis
Golgi markers, 7, 8 2,5-hexanedione and 145 mediators, 67–69
Golgi proteins, 5 carbendazim, 173 Hypothalamic kisspeptin (KiSS1), immune cells, 66, 67f
coincidental temporal High endothelial venules (HEVs), 157 pathogenesis, 64, 65–69
expression of non- 75 Hypothalamic pathways, 236 human orchitis, 63
Golgi and, 21, 23–24 High-intensity focused Hypothalamic-pituitary gonadal perspectives, 69
defining in spermatids, ultrasonography axis, 157–158 treatment of orchitis, 69
17–18 (HIFU), 146 Hypothalamic-pituitary-testes Inhibin, 125
in early spermatids during High-mobility group box 1 (HPT) axis, 143 Inhibin B, 124
acrosome formation, (HMGB1), 54 Innate defense system
9–10 High-risk chemotherapeutic I PRR-initiated innate immune
forming Hermes body at agents, 129 ICA69, 16 responses
step 19 spermatids, H4K44 acetylation (H4K44ac), IHO1, protein complex, 110 Leydig cells, 57–58
19–20 110 Imatinib, 141t male germ cells, 58
localized to spermatocytes, Homophilic trans-interaction, Immotility (IM), 142 in testis, PRRs, 55–57
7–9 JAMs, 73, 74t, 75 Immune cells TLR-initiated innate immune
matching protein expression Homozygous mutations, 111 autoimmune orchitis, 66, 67f responses in Sertoli
profiles of Golgi HORMAD1, 109 in promoting germ cell death, cells, 57
and non-Golgi Hormonal medications, 129, 140t 67 TLR signaling in testicular
proteins during fertility/infertility in humans, Immune homeostasis, TAM macrophages, 58–59
spermatogenesis, 129 receptors in Innate immune responses
20–21 Hormonal regulation regulating, 52–53 by microbial pathogens, 52
overlapping expression cytoskeletal reorganization, Immune privilege, 183 PRR-initiated
profiles of non-Golgi 175–176 potential downside of, 88–89 Leydig cells, 57–58
and, 24–25 of spermatogenesis, 125 of testis, 183–184, 184f; see male germ cells, 58
proteins involved in acrosome Hotspot, meiotic recombination, also Testis, HIV/ Sertoli cells, TLR-initiated, 57
binding to the nuclear 110; see also Meiotic ZIKV viruses in testicular cells, TAM
surface, 16–17 recombination Immune regulation within testis, inhibition, 54–55
proteins of unknown HSP70.2 (HspA2), 11, 12, 14f SCs, 81–89 Insecticides, 254
function, 20 Human chorionic gonadotropin BTB/SCB, 83–85 Insulin-like 3, 245
for residual bodies at step 19 (hCG), 125 movement of germ cells Insulin-like growth factor, 124
spermatids, 20 Human genes encode type 1, 247 across, 84–85 Insulin-like peptide (ILP), 202
segregation to acrosome, 12 Human HSD3B1, 246 testis immune privilege, 85 Insulin receptor, 235
Golgi ribbon, 5, 10, 18, 20 Human immunodeficiency virus cell mediated immune Insulin receptor subtype-2 (IRS-2),
Golgi trafficking, 7 (HIV), 58, 59, 63, response, 88 225
GOLPH3 (Golgi phophoprotein), 88, 185–186; see also humoral immune response, Integrative analysis of proteins,
see GPP34 Testis, HIV/ZIKV 86–88 see Computational
Gonadotropin, 157 viruses in overview, 81 characterization/
Gonadotropin-releasing Human infertility, implications testis immune privilege, 81, integrative analysis of
hormone (GnRH), in, 111; see also 82f, 83f proteins
125, 144, 236 Mammalian meiosis, Immune responses Interferons (IFN), 185
Gonocytes, 94 genetics of cell mediated, 88 IFN-gamma (IFNγ),
testosterone and number of, Human menopausal humoral, 86–88 regulation of JAMs
43–44 gonadotropins innate, see Innate immune and CAR, 77
GOPC, 15 (hMG), 125 responses IFN-stimulating gene 15
Gopc, 172 Human orchitis, 63 Immune tolerance to male germ (ISG15), 58
Gossypol exposure, 256–257 Human primordial germ cells cells, TAM receptors Interleukin (IL), 65, 66, 68, 86
GPP34, 9–10, 11, 19 (hPGC), 168 in, 55 Interleukin-1alpha (IL-1α), 124
Index 273

Intermediate cellular Levonorgestrel (LNG), 171 Long noncoding RNA (lncRNA), Male infertility
compartment theory, Leydig cell function, 128 causes of, 220
85 environmental Low-risk chemotherapeutic Male reproductive organs,
Intermediate chemotherapeutic toxicants on agents, 129 estrogen receptors
risk agents, 129 androgen biosynthesis, Luteinizing hormone (LH), 123, (ER) expression in,
Intracytoplasmic sperm injection 245–246 143, 157, 236 117–118
(ICSI), 126, 130t, cell signaling for and androgen, 239; see also Male reproductive system,
139, 172 testosterone Endocrine disrupting knockout phenotypes
Intratesticular testosterone, 175 biosynthesis in chemicals (EDC) in, 76–77
Intrauterine insemination (IUI), Leydig cells, 247–248 Lymphomonocyte infiltration, Mammalian genome, 100
128, 130, 130t, 142 environmental toxicants 63 Mammalian meiosis, genetics of
In vitro fertilization (IVF), 123, targeting Leydig cell chromosomal synapsis, 107,
130t, 139, 158 about, 248, 249t–251t, M 109
In vitro meiosis, 110–111, 111t; 250 Macrophages human infertility,
see also Mammalian benzophenones, 253 antigen-presenting cells, 66 implications in, 111
meiosis, genetics of bisphenols, 252–253 testicular, TLR signaling in, meiotic initiation, 106
Ionomycin-stimulated carbamates, 254 58–59 meiotic recombination
conditions, 86 1,2-dibromo-3- Madin-Darby canine kidney crossover formation, 110
Ionotropic P2X receptors chloropropane (MDCK) cells, 75 DSB repair, 109–110
(P2XRs), 99 (DBCP), 255 MAJIN, DNA-binding meiotic recombination
Islet cell auto antigen 1-like metals, 255–256 transmembrane hotspot, 110
protein (ICA1L), 16 organochlorines, protein, 106 programmed meiotic DSB
Isoaspartyl peptidase/ 253–254 Malathion, 255 formation, 109
lasparaginase organophosphates, Male contraceptive-based model, telomere-led meiotic
(ASRGL1), 223 254–255 225–226 chromosome
Isoflavones, 257 perfluoroalkyl substances Malectin, 24 movement, 106–107,
(PFAS), 248–249 Male fertility, connecting mTOR 107f, 108t
J phthalates, 251–252 signaling to, 202, 203t in vitro meiosis, 110–111,
JAMs, see Junctional adhesion plant toxicants, 256–257 Male germ cells 111t
molecules (JAMs) polycyclic aromatic division, 193 Mammalian target of rapamycin
Junctional adhesion molecules hydrocarbons (PAH), immune tolerance to, TAM (mTOR), 101
(JAMs), 73–78 254 receptors, 55 Man2á1, 12
functions enzymes for testosterone innate immune responses in ManIIX, 12
barrier, 75–76 biosynthesis and testicular cells, 52 differential expression, 17–18
cell adhesion, 76 metabolism PRR-initiated innate immune MARK4, structure of, 226, 227
cell polarity, 76 3β-HSD, 246–247 responses in, 58 Matrix-assisted laser desorption
future perspectives, 77–78 17β-HSD3, 247 TAM receptors in, 53–54 mass spectrometry
general properties, 73–77 CYP11A1, 246 Male gonadal development, 168 (MALDI-MS), 220
knockout phenotypes in male CYP17A1, 247 Male infertility Matrix metalloproteinases
reproductive system, SRD5A, 247 asthenospermia (MMP), 96, 188
76–77 Leydig cell development, causes, 142–143 MAX, 106
molecular structure, 73–75 248 management, 143 MCT2, 24
homophilic and overview, 245 postejaculate urinalysis, Mechanistic target of rapamycin
heterophilic trans- Leydig cell hyperplasia, 118 143 (mTOR) signaling in
interaction, 73, 74t, 75 Leydig cells, 7, 9, 40, 41f, 42 azoospermia spermatogenesis
overview, 73, 74f AR expression, 43 pretesticular, 144 in maintaining
tissue distribution and and EDC targeting of chemical pollutants on spermatogonial
localization in testis, testicular cells, spermatogenesis function, role of
74t, 75 237–238; see also and, see Chemical about, 202, 204–205
overview, 73 Endocrine disrupting pollutants on mTOR signaling in
regulation, in testis, 77 chemicals (EDC); spermatogenesis and meiosis, 205
Junctional restructuring theory, Environmental male infertility mTOR signaling in Sertoli
85 toxicants on Leydig DNA fragmentation cells, 205–206
cell about, 150 mTOR complexes and
K development, 248 management, 150 inhibitor rapamycin,
Kallman syndrome, 126, 128, 144 innate immune responses in ejaculatory dysfunction 201
Kartagener’s syndrome, 126, 142 testicular cells, 52 about, 147 mTOR signaling to male
KASH5, 106 PRR-initiated innate immune anejaculation, 149 fertility, connecting,
Keratin, 223 responses in, 57–58 anejaculation/failure 202, 203t
Ketoconazole, 255 source of androgens, 234 of emission, rapamycin and rapalogs
Kisspeptin (KiSS1), 157 testis immune privilege, 81, management of, 149 in human disease,
Klinefelter syndrome (KS), 126, 82f, 83f retrograde ejaculation, pharmacological
127, 145 testosterone production, 42 147–148 applications of,
Knockout phenotypes, in male TLRs in, 55 evaluation 201–202
reproductive system, Tyro3 in, 53 history, 139, 140t–141t Medications; see also Fertility/
76–77 Ligand-binding domain (LBD), physical exam, 139 infertility in humans,
Kyoto Encyclopedia of Genes and 40 investigations regulation of
Genomes (KEGG), Light microscope (LM) semen analysis, 139–142, antibiotics, 129
221 observations, 1, 4 142t antihypertensives, 129
Lindane, 254 oligospermia chemotherapeutic agents, 129
L Liquid chromatography and mass causes, 143 fertility and pollution, 129
Lactacystin, 99 spectrometry (LC–MS), management, 143 hormonal, 129
Lactate dehydrogenase C 220 testicular azoospermia psychiatric-related, 129
(LDHC), 24 Listeria monocytogenes, 64 causes, 145 MEIOB, 110
LAMAN, 12 Lkb1, 202 management, 145–146 MEIOC complex, 106
Laminin, 96 LncRNA, 100 posttesticular Meiosis, 7, 205
Lateral elements (LE), 107 Localization, in testis, 74t, 75 azoospermia, Meiosis
Lead, 256 Longitudinal intussusception 146–147, 147t mTOR signaling in, 205
Lead (paint), 129 vasoepididymostomy varicocele in vitro, 110–111, 111t; see also
Leptotene spermatocytes, (LIVE) technique, about, 150–151 Mammalian meiosis,
movement, 84–85 147, 148f management of, 151 genetics of
274 Index

Meiosis-specific genes in human Microtubule (MT)-based Myosin-based vesicles, 194 Nuclear localization signal (NLS),
infertility, 111t cytoskeletons, 174f, Myosins, 191, 192 40
Meiosis-specific proteins, 106 175, 216; see also Myxoma virus, 64 Nuclear-only estrogen receptor 1
Meiotic cells, F-actin in Spermiation (NOER), 118, 119
spermatogonia and, Minichromosome maintenance N Nuclear receptors (NR), 156
192–193 (MCM) helicase N-acetylgalactosaminyl­ N-WASP, 175, 194
Meiotic chromosome family, 110 transferase 3, 16 Nω-Nitro-L-arginine methyl
movement, telomere- MiR-145, overexpression, 77 NADPH for electron transfers, ester hydrochloride
led, 106–107, 107f, MiR-106b-25, 101 246, 247 (L-NAME), 67
108t Mitogen-activated protein NADPH oxidase 1 (Nox1), 101
Meiotic division, 1 kinases (MAPK), 235 NADPH oxidase 3 (Nox3), 101 O
of spermatocytes, 3 pathways, 118 National Health and Nutrition Obesity and diabetes mellitus,
Meiotic double-strand Mitotic phase of spermatogenesis Examination 129
breaks formation autocrine/paracrine Survey (NHANES) Obesity-associated
programmed, 109; regulation of 2011–2012, 252 asthenozoospermic
see also Meiotic spermatogonia, 97–99 National Institute on Aging patients, 223
recombination interaction between Interventions Testing Obstructed ejaculatory ducts, 143
Meiotic initiation, 106; see also spermatogonia and Program, 202 Obstructive azoospermia (OA),
Mammalian meiosis, niche, 96–97 Natural fertility, 126 125
genetics of metabolism of Natural fertilization, 125 Occludin, 65, 66, 84, 85, 224, 225
Meiotic recombination, spermatogonia, 101 Natural killer (NK) cells, 86, 88 Oligoasthenoteratospermia
106, 109; see also noncoding RNA in N-butyldeoxynojirimycin (OAT), 151
Mammalian meiosis, spermatogonia (NB-DNJ), 13 Oligoastheoteratozoospermia
genetics of regulation, 100–101 N-Cadherin, 224 (OAT), 128
crossover formation, 110 origin and subpopulations Nectin-2, 175, 194, 196 2’5’-oligodenylate synthetase1
DSB repair, 109–110 of spermatogonia, Neisseria gonorrhoeae, 59, 63 (OAS1), 58
meiotic recombination 94–95 Neonatal Leydig cells, 245 Oligospermia; see also Male
hotspot, 110 overview, 94 Neonatal pig islets (NPIs), 86, 88 infertility
programmed meiotic DSB perspectives, 102 Neonatal pig SCs (NPSCs), 86 causes, 143
formation, 109 spermatogonia, niche of, 95 Nerve growth factor (NGF), 170 management, 143
Meiotic recombination hot spot ubiquitin proteasome system Neuroendocrine control of Oligozoospermia, 63, 126; see also
locus (mrhl), 101 in spermatogonia testicular function, Azoospermia
Meiotic sex chromatin silencing regulation, 99–100 EDC affection of, 236 defined, 126
(MSCI), 109 Mitotic/proliferation phase, 1 Neurogenin 3-Cre, 205 Omics methods in translational
MEI4-REC114, 109 Mixed atrophy (MA), 221 Neurotrophin (NT), 170 research of male
Melanoma differentiation- Molecular characterization of Nicotinamide adenine reproduction,
associated protein 5 proteins, 221 dinucleotide 220–221
(MDA5), 57 Molecular structure, classical phosphatase Oncologic agents, 141t
Membrane attach complex JAMs and CAR, (NADPase), 5 Oocytes, 125
(MAC), 86 73–75 Nifedipine, 150 Orchidometer, 139
Membrane-bound Fas L (mFasL), homophilic and heterophilic NIMAlike kinase-1 (NEK1), 109 Orchitis, 59
68 trans-interaction, 73, NIPBL, 109 autoimmune
Membrane ESR1 (mESR1), 118 74t, 75 Nitric oxide synthase (NOS) experimental models of,
Mercury pollution, 256 overview, 73, 74f activity, 67 63–64
Mer receptor; see also Tyro3, tissue distribution and Nonclassical (nongenomic) pathogenesis, 64, 65–69
Axl, and Mer (TAM) localization in testis, pathway, testosterone focal, 64
receptors 74t, 75 signaling, 42–43 human, 63
immune tolerance to male Molinate, 254 Noncoding RNA (ncRNA), 100, perspectives, 69
germ cells, 55 Mono-butyl phthalate (MBP), 128 treatment of, 69
for triggering phagocytotic 251 Noncoding RNA in Organochlorines, 253–254
signaling, 53, 54f Monocarboxylate transporters spermatogonia Organohalogen chemicals, 236
Mesenchymal stem cells (MSC), (MCTs), 21 regulation, 100–101; Organophosphates, 254–255
237 Mono-ethylhexyl phthalate see also Mitotic phase flame retardants, 156
Metabolism of spermatogonia, (MEHP), 251, 252 of spermatogenesis Organotins, 256
101; see also Monomers, 191 Nongenomic (nonclassical) Orotidine 50-monophosphatase, 5
Mitotic phase of Motile sperm counts in ART, pathway, testosterone Outer dense fiber protein 2
spermatogenesis 130t signaling, 42–43 (ODF2), 223
Metabotropic P2Y receptors Motility and progressive motility Non-Golgi proteins, 20 Ovaries, 116
(P2YRs), 99 (PM), 142 Non-obese diabetic (NOD) mice,
Metals, 255–256 Mouse genes in meiosis, 108t 88 P
Methanol, 251 Mouse models Nonobstructive azoospermia Pachytene piRNA, 128
Methoxychlor, 253, 254 Leydig cells, 55, 57 (NOA), 125, 126, Pachytene stage, 111
3-Methylsulfonyl-DDE, 253 male germ cells, 58 144, 171 Pancreatic islets, for diabetes,
MFSD14A, 16 NOD mice, 88 Non-progressive motility (NPM), 85–86
MG160, 7–8 Sertoli cells, 54–55 142 P antigen family member 1
Microbial pathogens, innate TAM receptors, 52–55 Nonsister chromatids, 106 (PAGE1), 222
immune responses Movement, of germ cells across Normal sperm morphology, 127 Paracrine regulation
by, 52 BTB/SCB, 84–85 Notch gene homologue 1 of spermatogonia, 97–99
Microdissection testicular sperm Multidimensional protein (NOTCH1), 222 of testicular cells, 238t, 240f
extraction (mTESE), identification N-terminal domain (NTD), 40 Paracrine regulators of testicular
145, 145f technology N-terminal gamma-carboxylated cells, 238t, 240t
MicroRNA (miRNA), 77, 100, (MUDPIT), 220 glutamic acid (GLA), Paracrine signaling, 98
128, 170 Multidrug resistance gene 52, 53f Parathyroid allografts, 81
Microsurgical epididymal (MDR1), 225 N-terminal kinase domain, 227 Pathogen-associated molecular
sperm aspiration Multidrug resistance-related Nuclear ESR1 (nESR1) patterns (PAMPs), 55
(MESA), 126, 147, protein 1(MRP1), 225 expression, 118 Pathogenesis, of autoimmune
147f Mumps virus (MuV), 58, 59, Nuclear factor kappa B (NF-κB) orchitis, 64, 65–69
MicroTESE, 126 63, 64 MyD88-dependent pathway BTB, 65–66
Microtubule affinity regulating Mx GTPase1 (MX1), 58 and, 57 germ cell apoptosis
kinase 4 (MARK4), Myeloid differentiation protein 88 TLR signaling in testicular mediators, 67–69
225 (MyD88), 57 macrophages, 59 immune cells, 66, 67f
Index 275

Pattern recognition receptors Phosphoinositide 3-kinase Privileged sites, 183 phagocytosis of apoptotic
(PRRs), 55–59 (PI3K)-related kinase Privileged tissues, 183 germ cells, 53–54
activation, 52 family, 201 Prochloraz, 255 in regulating immune
families, 55, 56f Phospholipid hydroperoxide Profilin, 191 homeostasis, 52–53
immune responses in glutathione peroxida­ Progressive motility (PM), 142 in testis, 53–54
testicular cells, 52 semitochondrial Proline-rich region (PRR), 40 Recombinant soluble JAM-A
overview, 55–57 (PHGPx), 223 Properties, JAMs, 73–77 (rsJAM-A), 73, 75
PRR-initiated innate immune Phosphorylated ERK kinase Prosaposin, 10 Redd1 expression, 101
responses (p-ERK), 42 Prospermatogonia, 94 Regulation of JAMs and CAR in
Leydig cells, 57–58 Phospho-S6, 206 Prostaglandin (PG), 88 testis, 77
male germ cells, 58 Phthalates, 130, 251–252 signaling pathway, 160 Reproductive health,
signaling pathways, 57 Piperophos, 254 Prostate medications, 140t environmental
PCB126, 253 Pituitary adenylate cyclase- Protein c-Src, 226 pollution and
P450 cytochrome cholesterol activating Protein disulfide isomerase human reproductive health,
side chain cleavage polypeptide (PACAP), ER60, 63 declining, 155–156
enzyme (CYP11A1), 170 Protein interacting with C kinase overview, 155
245, 246f PIWI-interacting RNA (piRNA), 1 (PICK1), 15, 16 Rete testis (RT), 116
PDILT, 10 100, 128 Protein kinase C (PKC), 235 Retinoic acid (RA), 98, 106, 111,
Pelvic surgery, 126 Planar cell polarity (PCP) core Protein-protein docking software, 124, 170
Penile examination, 139 proteins, 211, 213f, 221 Retinoic acid-inducible gene
Pentachlorophenol (PCP), 253 214t Protein S (ProS), ligand of TAM (RIG-I)-like receptors
Percutaneous epididymal sperm Planar cell polarity (PCP) matter receptors, 52, 53f (RLRs), 55, 57
aspiration (PESA), PCP proteins and Proteomic approaches in Retinoid signaling, 106
126, 146, 146f spermatogenesis biomarkers Retinoid X receptors (RXRs), 156
Perfluorinated compounds (PFC); background, 211–213, 214t identification, 221 Retrograde ejaculation, 147–148;
see also Chemical study in vitro, 213, 214–215 Proteomic markers and drug see also Ejaculatory
pollutants on study in vivo, 215 targets, 222–223 dysfunction
spermatogenesis and Planar cell polarity matter and Proteomics analysis, 257 causes, 148
male infertility spermatogenesis Proteomics methods, 220, 223 management, 148–149
human exposure risk, and overview, 211 PRRs, see Pattern recognition Retroperitoneal lymph node
reproductive health Plant toxicants, 256–257 receptors (PRRs) dissection (RPLND),
about, 158–160 Plasma membrane receptors, Pseudoephedrine, 143 126
perfluoroalkyl chemicals/ 234 P204 signaling, 58 Rho-associated kinase (ROK), 213
mechanistic activities, Plasminogen activator, 124 Psychiatric-related medications, Ribosomal protein S6, 206
160 Plasticizers, 251 129, 141t Ribosomal protein S6 kinase
Perfluorinated compounds (PFC), Plzf, 202, 205 fertility/infertility in humans, (S6K), 201
156 Polarity, cell, 76 129 Rictor depletion, 202
Perfluoroalkyl chemicals/ Polarity proteins, 158 PTEN, 201, 204f, 206 RNA, noncoding, in
mechanistic activities, Polo-like kinase 1 (PLK1), 109 Purinergic signaling, 98 spermatogonia
160 Polychlorinated biphenyls regulation, 100–101
Perfluoroalkyl substances (PFAS), (PCBs), 155, 156, 253 Q RNA virus sensor signaling, 59
248–249 Polycomb repressive complex 1 Quantitative proteomics, 5 Rosiglitazone, 255
Perfluorohexane sulfonate (PRC1.6), 106 Quiescent stem cells, 170 Round spermatids, 193
(PFHS), 156, 159, 160 Polycyclic aromatic hydrocarbons Round spermatids to functional
Perfluorooctane sulfonate (PAH), 254 R spermatozoa,
(PFOS), 156, 250 Polyglandular autoimmune RAB14, 21 development of;
Perfluorooctanesulfonic acid syndrome, 63 RAD51, 45 see also Human
(PFOS), 237 Polyubiquitination of K48 chain, RAD51/DMC1, 109 spermatogenesis
Perfluorooctanoic acid (PFOA), 99 Radiotherapy, 223 cytoskeletal reorganization,
156, 159, 160, 250 Postejaculate urinalysis; see also RanBP5, 23, 24 175–176
Perforin, 86 Asthenospermia Rapamycin, 201 spermiation, 172, 174–175
Perinatal exposure to BPA, 158 causes, 143 inhibitor, mTOR complexes spermiogenesis, 171–172, 173t
Periodic acid-Schiff (PAS) concentration and sperm and, 201 RPA, 109, 110
reaction, 172 count, 143 and rapalogs in human RTKs, see Receptor tyrosine
Periodic acid Schiff (PAS) management, 143 disease, kinases (RTKs), TAM
technique, 1, 3–4, 4f Postoperative hydroceles, 151 pharmacological
Peritubular cell-specific AR Posttesticular azoospermia applications of, S
knockout (PTM- management of 201–202 Sapreticulin, 10
ARKO) mice, 44 epididymal obstruction treatment, 204 Scarb1 gene, 248
Peritubular myoid cells and vasal obstruction, Raptor-deficient testes, 205 Scavenger receptor class B
(PTMC), 116, 123, 146 RA receptors (RAR), 98 member 1(SCARB1),
124, 168 reconstruction, 147, 147t RA response element (RARE), 246f
AR expression, 43 sperm retrieval in OA, 98 Schiff reaction, 2f, 3
Peroxiredoxin 1, 24 146–147 Rattus, 99 Scrotal examination, 139
Peroxiredoxin 6, 24 obstructive causes, 146 Rbfox2 (RNA binding protein), SCs, see Sertoli cells (SCs)
Peroxisome proliferator-activated PRDM9, 110 101 Seager Electroejaculator, 150
receptors (PPAR), Prdm9 gene, 110 Reactive oxygen species (ROS), Sec23, 16, 21
130, 156, 160 Pregnancy and infertility, 101, 253, 257 SEC23IP, 16
Peroxisome proliferators, 160 epidemiology of, 123 Real-time polymerase chain Secondary spermatocytes, 126
Pesticide (methoxychlor), 157 Pregnane X receptors (PXR), 156 reaction (RT-PCR), Selective estrogen receptor
Peutz-Jeghers syndrome (PJS), Pregnenolone, 245 220 modulators (SERM),
201, 206 Preleptotene spermatocytes, 124 REC8, 109 125, 143
P-glycoprotein, 225, 226 movement, 84–85 Rec8, 106 Selective 5-hydroxytryptamine
Phagocytosis of apoptotic germ Prepachytene piRNAs, 128 Receiver operating characteristics reuptake inhibitors,
cells, TAM receptors Prespermatogonia, 94, 168 (ROC), 142 129
in, 53–54 Pretesticular azoospermia, 125 Receptor mediated pathways, Semen analysis; see also Male
Phorbol 12-myristate 13-acetate Primary immotile-cilia 156–157 infertility
(PMA), 86 syndrome, 126 Receptor tyrosine kinases about, 139, 142, 142t
Phosphati-dylinositol- 3-kinase Primary spermatocytes, 126 (RTKs), TAM, 52–53 morphology, 142
(PI3K), 118 Primordial germ cells (PGC), categories, 52 motility and progressive
Phosphofructokinase, 225 123, 237 overview, 52, 53f motility (PM), 142
276 Index

Semen analysis, World Health Sertoli cells barrier (SCB), 83–85 PCP proteins and; see also Spermatogonial niches in mouse
Organization 2010, defined, 83 Planar cell polarity testis, 96f
125t germ cell development, 83–84 (PCP) matter Spermatogonial progenitor cells
Semen analysis abnormalities, movement of germ cells background, 211–213, (SPC), 202
defined, 142 across, 84–85 214t Spermatogonial stem cells (SSC)
Semen volume and pH, 143 testis immune privilege, 85 study in vitro, 213, about, 94
Seminal proteomes, 128 Sertolionly syndrome, 144 214–215 fertility and infertility
Seminiferous epithelium, 1, 3, 3f Sertoli-spermatid adhesion study in vivo, 215 regulation, 123–124
Seminiferous tubule contraction, complex, 45 regulation of, 174 functional activity of, 95
F-actin in, 197 Sertoli valve, 95 reproductive toxicity of EDC human, 171
Seminiferous tubules (STs), 1, 40, Serum antisperm antibodies, 146 and, 158 within seminiferous tubule,
63, 81 Serum/glucocorticoid induced SC immune regulation within 116
about, 111 kinase (SGK), 201 testis, see Sertoli cells transplantation of, 96
cross-sectional profiles, 2f, 3f Sex chromosomes, 109, 127 (SCs) Spermatogonial transplantation,
structure of, 183, 184 Sex-hormone-binding globulin and spermiogenesis, 123–124 192
Sendai virus, 64 (SHBG)-like domains, testicular immunoregulation, Spermatozoa, 1, 3, 124, 191
Septin 4, 221 52, 53f see Testicular identification of, 123
Sertoli cell AR knockout Sexual transmission of viruses, immunoregulation Sperm chromatin, 158
(SCARKO) mice, 44, 185 Spermatogenesis, membrane/ Sperm chromatin dispersion
45, 46, 47 Short ncRNA, 100 nuclear estrogen (SCD), 127, 150
Sertoli cell F-actin-containing Signaling pathways, PRRs, 57 receptors in Sperm chromatin structure assay
structures, Simian immunodeficiency virus E2/ESR1 in male (SCSA), 150
dynamics of (during (SIV), 185 reproduction, role of Sperm count, concentration and,
spermatogenesis), Single-cell gel electrophoresis, environmental 143
196–197 127 estrogens impact Sperm DNA fragmentation, 127;
Sertoli cell only (SCO), 126 Single-stranded DNA (ssDNA), human testicular see also Azoospermia
Sertoli-cell-only (SCO) 109 development, 119–120 Sperm DNA integrity, 150
syndrome, 221, 222 Sirolimus, 201 knockouts of ESR2 or Spermiation
Sertoli cells (SCs), 3, 7, 9, 12, 19, Sirt1, 15 GPER, 119 about, 172
20, 40, 42, 45 S6 kinase 1 (S6K1), 202 loss of ESR1 causing male actin- and MT-based
and androgen receptor (AR), Skp1-Cullin-F-box (SCF) reproductive defects, cytoskeletons, role
116 complex, 99 118 of, 175
AR expression, 43 Slc9a3 expression, 119 loss of membrane ESR1 regulation of
BTB function, 65–66 Smad signaling, 77 expression causing spermatogenesis, 174
in EDC targeting of testicular Small interfering RNA (siRNA), male reproductive Spermiogenesis, 1
cells, 237; see also 128 abnormalities, defined, 124
Endocrine disrupting Small RNAs, 128 118–119 F-actin during, 193–194
chemicals (EDC) sequencing methods, 220 estrogen sources in males round spermatids to
immune regulation within SMAP2, 16 about, 116–117 functional
testis, 81–89 Sodium/hydrogen exchanger effects of early estrogen spermatozoa,
BTB/SCB, 83–85 (NHE3; SLC9A3), 119 treatment in males, development of,
cell mediated immune Soluble form Fas L (sFasL), 68–69 118 171–172, 173t
response, 88 Solvents, 251 estrogen receptors (ER) spermatogenesis and,
humoral immune Somatic cells, 95 expression in male 123–124
response, 86–88 Somatic Sertoli cell, 191 reproductive organs, Sperm mitochondria-associated
overview, 81 Sororin, 109 117–118 cysteine-rich protein
testis immune privilege, SORTILIN, differential Spermatogenic cells, F-actin in (SMCP), 222
81, 82f, 83f expression, 17–18 F-actin during Sperm morphology, 142
JAMs in, 75 Sox9 genes, 168 spermiogenesis, Sperm motility, 126, 142
mTOR signaling in, 205–206 SPACA4, 222 193–194 defects, 77
phagocytosis of apoptotic SPATA16 protein, 16 F-actin in spermatogonia and Sperm protein associated
germ cells, 53–54 Sperm acrosome associated 1 meiotic cells, 192–193 with nucleus on
potential downside of (Spaca1), 16 Spermatogonia; see also X chromosome B
immune privilege, Sperm acrosome associated 4 Mitotic phase of (SPANXB), 223
88–89 (SPACA4), 221 spermatogenesis Sperm release, 173
role of, 124 Spermatic antigens, antibodies autocrine regulation of, Sperm retrieval in OA, 146–147;
SC androgen receptor to, 63 97–99 see also Posttesticular
knockout (SCARKO) Spermatic cord, 139 F-actin in/meiotic cells, azoospermia
mice, 84 Spermatid-associated protein 192–193 Spinal cord injury, 126
SC conditioned media (SPERT), 222 metabolism of, 101 SPO11, 109
(SCCM), 88 Spermatids, 3, 67, 81, 84, 172 and niche, interaction src kinase, 40
TAM receptors in testis, 52, 53 and actin monomers, 193 between, 96–97 SRD5A, see 5α-reductase
testosterone signaling Spermatocytes, 1, 58, 67, 81, 84, niche of, 95 (SRD5A1)
pathways, 42–43 124 origin and subpopulations of, sry genes, 168
TLR-initiated innate immune meiotic division of, 3 94–95 STAG1/STAG2/STAG3, 109
responses in, 54–55, Spermatogenesis, 123 paracrine regulation of, 97–99 Stem cell factor (SCF), 123, 170,
57 cytoskeletons (F-actin) and, regulation 202
Sertoli cells, F-actin in see Cytoskeletons ubiquitin proteasome Stem cell renewal theory, 1
in ectoplasmic (F-actin) and system in, 99–100 Steroid hormone receptors
specializations, spermatogenesis undifferentiated, 168–171 expression in
194–195 hormonal regulation of, 125 Spermatogonia (SPG), 1, 58, 123, testicular cells, 234
in seminiferous tubule human, see Human 125 Steroid hormones, 234
contraction, role of, spermatogenesis type B, 168 receptor knockout, 118
197 inflammation and, see Spermatogonial function, role of Steroid metabolizing enzyme
Sertoli cell F-actin- Inflammation mTOR signaling in activities, toxicants
containing structures, JAMs, see Junctional maintaining on, 251t
dynamics of (during adhesion molecules about, 202, 204–205 Steroidogenic acute regulatory
spermatogenesis), (JAMs) mTOR signaling in meiosis, protein (StAR), 158,
196–197 mitotic phase of, see 205 225, 236, 246f, 248
tubulobulbar complex (TBC), Mitotic phase of mTOR signaling in Sertoli Stimulator of IFN gene (STING),
195–196 spermatogenesis cells, 205–206 57
Index 277

STING-dependent pathway, 58 PRRs, see Pattern Thiram, 254 Trimethylation of lysine 36


Stra8, 106 recognition receptors Thyroid hormone receptors (TR), of histone H3
Strep-Tag molecule, 68 (PRRs) 156, 235 (H3K36me3), 110
Streptomyces bygroscopicus, 201 TLRs, see Toll-like Thyroid hormone response Triphenyltin, 256
Stress-70 protein, 223 receptors (TLRs) elements (TRE), 236 tsc2-deleted testes, 204
Stromal cell derived factor 1 orchitis, 59 Tight junctions (TJs), 75–76, tsc1 knockout testes, 204
(SDF-1), 123 overview, 52–55 83, 84 T-2 toxin, 257
Structural maintenance of TAM receptors, see Tyro3, actin-based, 171 Tuberous sclerosis (TSC) 1/TSC2
chromosome (SMC) Axl, and Mer (TAM) proteins, 160 complex, 201
proteins, 109 receptors with Sertoli cells, 205, 213 Tubulin beta 2B (TUBB2B), 223
Study in vitro, planar cell polarity Testicular macrophages, TLR Tissue distribution, in testis, Tubuli rectus, 95
(PCP) proteins, 213, signaling in, 58–59 74t, 75 Tubulobulbar complex (TBC),
214–215 Testicular sperm aspiration TLRs, see Toll-like receptors 173
Study in vivo, planar cell polarity (TESA), 144, 144f (TLRs) F-actin in, 195–196
(PCP) proteins, 215 Testicular sperm extraction TMED4/P25, 5–7, 12 Tumor necrosis factor alpha
Subfertility, etiological factors, (TESE), 144 TMED4/p25, 21 (TNF-α)
63 Testis TMED7/P27, 5–7, 12 germ cell apoptosis, 55
Subinguinal varicocelectomy, immune privilege, Sertoli TMED7/p27, 21 regulation of JAMs and
151f cells, 81, 82f, 83f, 85 TM9SF3, 5–7, 12, 21 CAR, 77
Substrate S6 kinase 1 (S6K1), 202 regulation of JAMs and CAR Toll-like receptors (TLRs) testicular tight junction
Subtilosin, 129 in, 77 initiated innate immune function, 66
Sulfated glycoproteins (SGP), 237 Sertoli cell immune responses in testis, Tyro3, Axl, and Mer (TAM)
Sulfoglycolipids, 10 regulation within, see 52–53, 54–55 receptors, 52–55
SUN1, 106, 107 Sertoli cells innate defense system, 55, 57 in immune tolerance to male
Superresolution imaging, 107 TAM receptors in, 53–54 pattern recognition receptors, germ cells, 55
Surface- enhanced desorption/ tissue distribution and 88 inhibition of innate immune
ionization mass localization in, PRR signaling pathways, 57 responses in testicular
spectrometry 74t, 75 TLR-initiated innate immune cells, 54–55
(SELDI-MS), 220 Testis, HIV/ZIKV viruses in responses in Sertoli overview, 52, 53f
Surgical sperm retrieval, 144 immune privilege of testis, cells, 57 phagocytosis of apoptotic
SYCP1, 107 contributing factors, TLR signaling in testicular germ cells, 53–54
SYCP2, 107 183–184, 184f macrophages, 58–59 in regulating immune
SYCP3, 107 overview, 183 TopoVI DNA topoisomerase homeostasis, 52–53
Synapsis, 107 viruses and testis subunit A (TopoVIA), schematic structure, 52, 53f
Synaptonemal complex (SC) about, 184–185 109 in testis, 53–54
about, 107 human TOR pathway in reproduction, Tyro3 receptor, 55
tripartite structure of, 107 immunodeficiency 203t Tyrosine kinase receptors, 235
Syncytial state, 94 virus (HIV), 185–186 TOR/Raptor-dependent
Syntaxin 2, 16 Zika virus (ZIKV), 186, mechanism, 202 U
Syntaxin 13, 24 187t Total motile count (TMC), 126 Ubiquilin 3 (UBQLN3), 221
Synthetic fluorinated Testis-expressed 11 (TEX11) Toxoplasma gondii, 58, 59 Ubiquitin, 21
hydrocarbons, 158 gene, 128 Transcriptomic approaches Ubiquitin-activating enzyme E1
Synthetic organochlorine Testis gene expression, 46–47; in biomarkers (Uba1), 15
pesticide, 253 see also Androgen identification, 221 Ubiquitination, 99
Systemic immune tolerance, 183 regulation of Transcriptomic biomarkers, Ubiquitin-directed AAA-ATPase
spermatogenesis 221–222 P97, 21
T Testis-specific profilins, 194 Transepithelial resistance (TER), Ubiquitin-proteasome system
TATA element modulatory factor Testis-specific serine kinase 6 76 (UPS)
(TMF), 17 (TSSK6), 222 Transferrin, 24, 28f, 124, 237 about, 99
Tbc1d20 gene, 14 Testosterone; see also Androgen Transforming growth factor- functional role of, 99
Tek4 (Tektin 4), 172 regulation of alpha (TGF-α), 124 Ubiquitin proteasome system
Telomere-led meiotic spermatogenesis Transforming growth factor-β in spermatogonia
chromosome biosynthetic enzyme (TGF-β), 124, 170 regulation, 99–100
movement, 106–107, activities, 249t–250t regulation of JAMs and Ubiquitin-specific peptidase 8
107f, 108t concentrations, 116, 125 CAR, 77 (USP8), 13
Telomeres, 106 downregulation, 59 testicular tight junction Ubiquitous environmental
Teratozoospermia, 127; see also to maintain the blood-testis function, 66 contaminants, 156
Azoospermia barrier, 45–46 Trans Golgi networks (TGNs), 5 UBQLN3, 222
TERB1, 106 and number of gonocytes, Trans-interaction, homophilic UBXD8, 10
TERB2, 106 43–44 and heterophilic, 73, UDP-glucose: glycoprotein
Terminal deoxynucleotidyl production, 42, 245 74t, 75 glucosyltransferase
transferase-mediated signaling pathways, 42–43 Transition protein 2 (TNP2), (UGGT), 23
dUTP nick end for spermatid attachment and 222 Ultrastructural defects (USD),
labeling (TUNEL), release of sperm, 45 Transition protein 1 (TNP1) gene, 142
127, 150 testis gene expression, 46–47 222 UniProtKB/Swiss-Prot, 8
Terminal kinase-associated Testosterone biosynthesis Transrectal ultrasound (TRUS), United Nations Environment
domain, 227 in Leydig cells, 143 Programme/World
Testicular azoospermia; see also cell signaling for, Transurethral resection of Health Organization
Male infertility 247–248 ejaculatory duct (UNEP/WHO)
causes, 145 Testosterone signaling pathways (TURED), 143, 146 report, 155
management, 145–146 classical pathway, 42 Transverse elements (TF), 107 UPEC (uropathogenic Escherichia
posttesticular azoospermia, nonclassical (nongenomic) Treg cells, 66, 67f coli), 58–59
146–147, 147t pathway, 42–43 Tributyltin, 256 Uridine 5’-diphospho (UDP), 236
Testicular cells, paracrine Tetrachlorodibenzo-p-dioxin, Tricellulin, 84 Uropathogenic Escherichia coli
regulators of, 238t 129 Triclosan, 254 (UPEC), 58–59
Testicular dysgenesis syndrome 2,3,7,8-tetrachlorodibenzo-p- Tricresyl phosphate, 255
(TDS) theory, 120 dioxin (TCDD), 253 Tridecyl alcohol, 251 V
Testicular immunoregulation, Tetraiodothyronine, 235 Triiodothyronine, 235 Vaccination, MuV orchitis, 59
52–59 TGNs (trans Golgi networks), 5 Trimethylation of lysine 4 Valsalva, 128, 151
innate defense system, Thiamine pyrophosphatase of histone H3 Vangl, 211–216, 214t
55–59 (TPPase), 5 (H3K4me3), 110 Vang/Pk complex, 213
278 Index

Varicocele; see also Fertility/ Vectorial asymmetry, 211 Voltage-dependent anionselective Z


infertility in humans, Vesicle-associated membrane channel protein 2 ZBTB16 (PLZF), 44
regulation of; Male protein 3 (VAMP3), (VDAC2), 223 Zearalenone, 257
infertility 21 VPS54, 14 Zika virus (ZIKV), 88, 186, 187t;
about, 128, 150–151 Vibrostimulation, 150 see also Testis, HIV/
fertility as function of age, Vinclozolin, 158 W ZIKV viruses in
128 Viral orchitis, 59 WAPL, 109 infection in male
management of, 151 Viruses and testis; see also Wnt6, 98 genitourinary tract,
Varicocelectomy, 150 Testis, HIV/ZIKV Wnt5a, 98 187t
subinguinal, 151 viruses in Wt1 gene, 170 Zinc metallopeptidase, 221
Varicocele repair (VR), 151 about, 184–185 ZIP9, 43
Vasal obstruction human immunodeficiency X Zipper theory, 85
posttesticular azoospermia, virus (HIV), 185–186 Xenoestrogens, 234 Ziram, 254
146 Zika virus (ZIKV), 186, 187t Zonadhesin, 64
vasovasostomy for, 147 Vitamin C Y Zona pellucida, 125, 127
Vas deferens, 139 in DNA fragmentation, 150 Y-chromosome microdeletion, Zonula occludens (ZOs), 84
Vasoepididymostomy (VE), 147 Vitamin E 126, 127, 145
VASP proteins, 191, 193 in DNA fragmentation, 150 YTHDC2 complex, 106

Potrebbero piacerti anche