Sei sulla pagina 1di 201

Bone Resorption

Felix Bronner, Mary C Farach-Carson and


Janet Rubin (Eds)

Bone
Resorption
Volume 2 in the Series
Topics in Bone Biology
Series Editors:
Felix Bronner and Mary C. Farach-Carson
Felix Bronner, PhD
Department of BioStructure and Function,
Department of Pharmacology,
University of Connecticut Health Center,
Farmington, CT,
USA

Mary C Farach-Carson, PhD


Department of Biology,
University of Delaware,
Newark, DE, USA

Janet Rubin, MD
Division of Endocrinology and Metabolism,
Emory University School of Medicine,
Veterans Affairs Medical Center,
Decatur, GA,
USA

British Library Cataloguing in Publication Data


A catalogue record for this book is available from the British Library

Library of Congress Control Number: 2005924008

Apart from any fair dealing for the purposes of research or private study, or criticism or review, as permitted under the
Copyright, Designs and Patents Act 1988, this publication may only be reproduced, stored or transmitted, in any form or by
any means, with the prior permission in writing of the publishers, or in the case of reprographic reproduction in accordance
with the terms of licences issued by the Copyright Licensing Agency. Enquiries concerning reproduction outside those terms
should be sent to the publishers.

ISBN 1-85233-812-1
Springer Science+Business Media
springeronline.com

Springer-Verlag London Limited 2005

The use of registered names, trademarks, etc. in this publication does not imply, even in the absence of a specic statement,
that such names are exempt from the relevant laws and regulations and therefore free for general use.

Product liability: The publisher can give no guarantee for information about drug dosage and application thereof contained
in this book. In every individual case the respective user must check its accuracy by consulting other pharmaceutical
literature.

Typeset by SNP Best-set Typesetter Ltd., Hong Kong


Printed and bound in the United States of America
28/3830-543210 Printed on acid-free paper SPIN 10943977
Preface

Bone Resorption, the second volume of the series Topics in Bone Biology, is
centered on the osteoclast, the bone-resorbing cell. The volume thus com-
plements the rst volume of the series, Bone Formation, which discussed
origin, function, and pathology of the bone-forming cell, the osteoblast. Both
volumes are addressed to scientists and clinicians who are active in or wish
to enter the eld of skeletal function; this group encompasses a wide variety
of specialties, including orthopaedics, rheumatology, endocrinology,
nephrology, oncology, dentistry, nursing, and chiropractic medicine.
Rubin and Greeneld, in the rst chapter, discuss the origin and differen-
tiation of the osteoclast. Recognized as a multinuclear cell about 150 years
ago, its origin was not uncovered until a century later when, as a result of
an experiment in which quail and chicken cells were co-cultured, it became
clear that osteoclasts arose from hematopoietic stem cells. The authors then
discuss the osteoclastogenic factor, now known as RANKL; the inhibitory
binding protein, osteoprotegerin (OPG); and the complex signaling pathway
that leads to differentiation and late differentiation of this complicated cell.
A section of the chapter is devoted to a concise discussion of regulators and
repressors of osteoclast differentiation, including sex steroids, calcitonin,
nitric oxide, and mechanical factors. The chapter concludes that the osteo-
clast modulates bone development, bone growth, and bone disease.
Bone cells resorb bone by spreading their membrane over the bone surface
and then secreting enzymes that cause bone salt to be solubilized and the
organic matrix to be disassembled. Oursler, in Chapter 2, discusses the oste-
olytic enzymes of the osteoclast. One pivotal enzyme is the tartrate-resistant
acid phosphatase, yet its precise role remains unresolved. Recent data indi-
cate that this enzyme can dephosphorylate osteopontin and may therefore
play a role in suppressing bone resorption. Oursler proceeds to discuss the
cathepsins and points out the important role of cathepsin K in basic bone
resorption.Although most matrix metalloproteases (MMPs) are produced by
osteoblasts, osteoclasts produce MMP 1 and 9, which may therefore be impli-
cated in osteoclast-mediated bone degradation. As yet it is difcult to gen-
eralize about the roles played by MMPs and cathepsins. Future studies may
reveal that different molecules of these two enzyme classes bring about oste-
olysis when resorption is due to remodeling, as compared to the molecules
that are implicated in disuse or other bone-destroying processes.
How osteoclast activity is regulated is discussed by Baron and Horne in
the third chapter. Five steps are required for the bone-resorbing cell to do its
job: migration to the portion of bone that is to be resorbed, adhesion of the
podosome membrane, secretion of the various lytic and other enzymes,
acidication of the uid under the podosome, and internalization, i.e., ret-
rograde transport or transcytosis of some products under the podosome.
vi Preface

The essentiality of these ve steps becomes obvious when constituent steps


or molecules are genetically altered. The authors discuss the many signaling
molecules and cascades that regulate these ve steps. They conclude that
signaling from the integrins and from the receptors for RANK, M-CSF, and
calcitonin constitute the key regulatory pathways and that most signaling
cascades involve sequential phosphorylation and dephosphorylation events.
As these steps are identied, they may then become targets for therapeutic
approaches that attempt to re-equilibrate the relationship between bone for-
mation and resorption.
For bone tissue to be able to change its shape and structure, pre-existing
bone mineral and matrix must rst be removed. Bain and Gross, in the fourth
chapter, provide an overview of the structural aspects of bone remodeling,
with specic focus on how the osteoclast modulates bone mass and archi-
tecture. In cortical bone, the activation of the osteoclast machinery leads to
a linear cellular assembly that can tunnel through the cortex. This osteoclast-
generated tunnel then is lled by osteoblastic bone formation, leading to the
formation of what is called a bone structural unit (BSU). The authors indi-
cate that on the basis of available data a cortical BSU has a life span of 200
days and suggest that the bone-remodeling rate has been genetically pro-
grammed to maintain the skeletal competence of the skeleton. In cancellous
bone, the BSU is hemi-osteonal in nature; and, rather than boring a tunnel,
the osteoclasts effect a trench-like structure on the surface of cancellous
bone. In contrast with the relatively long time period it takes for bone loss
to weaken the structure of bone as a result of aging or of the hormonal
changes due to the menopause, loss of bone mass due to disuse is rather
rapid, the result of profound osteoclastic resorption on endo-and intracor-
tical surfaces. As a result of disuse, bone porosity is twice as great as normal.
Hyperactive osteoclasts and excessive bone resorption can lead to irrever-
sible bone loss and structurally weakened bone.
The role of inammatory cytokines in bone disease is discussed in
Chapter 5 by Nanes and Pacici, who emphasize that inammatory cytokines
play a major role in bringing about bone disease. For example, estrogen
deciency, typical of the menopause, leads to the elevated production of the
tumor necrosis factor (TNF) and of interleukin-1 (IL-1). These in turn stim-
ulate osteoclastogenesis and inhibit osteoblast differentiation and function.
The signaling pathways of TNF and IL-1, though different, converge, acti-
vating a transcription factor and stimulating a mitogen-activated protein
kinase. The chapter reviews in detail the data on the effect of estrogen de-
ciency on cytokine production; on the central role for TNF, its source, on
estrogen regulation of T cell clonal expression; and on interferon gamma and
the skeletal effects of this interferon. TNF signaling and its relation to IL-1
signaling, and to RANKL, is detailed, and the authors discuss how osteoclast
survival is enhanced by the effects of these cytokines. A section on the effects
of cytokines on osteoblast differentiation, function, and survival concludes
the chapter.
A detailed analysis and description of how mutations affect the various
aspects of osteoclastogenesis and osteoclast function as delineated in
transgenic mouse models is provided in Chapter 6 by Horowitz, Kacena, and
Lorenzo. In the early part of the chapter the authors describe transcriptional
regulation, based in part on events occurring in the course of the more
widely explored B-cell differentiation. The authors cover over-expression of
some transcription factors, the role of proto-oncogenes, e.g., c-fos, and the
effects of nuclear-factor-activated T cellsc1, a critical signaling molecule
in osteoclastogenic responses. Although the initial response of the bone-
Preface vii

resorbing cell to signals is mediated by surface membrane receptors, the


initial signals are followed by intracellular events that are then described. An
example is the colony-stimulating factor-1 (CSF-1 or MCSF) that may play a
role in minimizing osteoclast apoptosis. The role of OPG and its relationship
to RANK and RANKL and their function in osteoclastogenesis is described
in detail. This is followed by discussion of the roles of tumor necrosis factor
(TNF), the interleukins, prostaglandins, and prostaglandin receptors. The
last part of the chapter deals with specic mutations and their effects on
the development and function of the mutant mice and on specic bone-
resorbing processes. The role and function of cathepsin K is illustrated by
discussion of what happens to cathepsin K knockout mice. A nal section
deals with the role of integrins in bone resorption.
Rubin and Nanes, in Chapter 7, discuss diseases of unchecked osteoclast
activity that lead to a marked increase in bone fragility, as well as conditions
and diseases of too little osteoclast activity, in which bone mineral content
has increased beyond the normal and bone structure has become disordered.
Diseases of relatively excessive osteoclastic activity include postmenopausal
osteoporosis, hyperparathyroidism, and Pagets disease. The latter is an
example of a genetic disorder. Other inherited diseases with an overactive
osteoclastic phenotype that are discussed are familial expansile osteolysis,
NasuHakola disease, and Rett syndrome. Inherited diseases with an under-
active osteoclastic phenotype include three types of malignant osteopetrosis,
two types of osteopetrosis that are autosomal dominant, as well as the rare
diseases pycnodysostosis, osteopetrosis with renal tubular acidosis, and
CammuratiEngelmann disease. The chapter discusses the known genetic
basis in detail and functional and clinical aspects of these diseases, whether
relatively common, e.g., hyperparathyroidism, or quite rare, as are the
osteopetroses. The chapter contains illustrations of many of these conditions.
Osteoporosis is the most common bone disease that affects people over
50 years of age. Because the bone resorption rate exceeds that of bone for-
mation, pharmaceutical research has been directed at agents that target
recruitment and function of the osteoclast. Fitzpatrick, in Chapter 8, reviews
the principal compounds that have been tested and have become drugs used
to treat bone loss. A major category of these drugs includes the bisphos-
phonates, reviewed in the chapter as a class and invidually, including alen-
dronate, risedronate, ibandronate, etidronate, tiludronate, and zolendroic
acid. Clinical trials involving these drugs are described and their results ana-
lyzed and evaluated. Because bisphosphonates are relatively new drugs, their
long-term efcacy, not yet well known, is discussed and evaluated. Estrogens
and estrogen-like compounds are an entirely different class of agents that are
used or proposed as anti-resorptive agents. Results from the Womens Health
Initiative have demonstrated unequivocally that estrogen reduces fracture
risk and the details of that study and related studies of selected estrogen
receptor modulators (SERMs) are discussed, as are the results of treatment
with calcitonin and of combination antiresoptive therapies. The chapter con-
cludes with a brief outlook of future therapies.
The last chapter, by Clines, Chirgwin, and Guise, describes and analyzes
the role of the osteoclast in causing skeletal complications of malignancy.
The principal syndromes discussed are bone metastases, the most common,
the hypercalcemia of malignancy, and oncogenic osteomalacia. The authors
discuss humoral mediators of malignancy, i.e., parathyroid hormone-related
protein, 1,25-dihyroxyvitamin D3, parathyroid hormone, as well as some
cytokines, the tumor necrosis factor, and the granulocyte-colony stimulat-
ing factor. The authors then outline treatment of hypercalcemia due to malig-
viii Preface

nancy and proceed to a discussion of solid tumor metastases in which they


classify them, describing the clinical consequences, the steps leading to the
metastases, and the development of the skeletal lesions. They illustrate the
vicious cycle between tumor cells and the skeleton and then turn to osteob-
lastic and mixed metastases. The chapter concludes with a discussion of
experimental and clinical therapies and an evaluation of how therapy affects
the skeleton of cancer patients who have lost bone.
All chapters contain extensive bibliographies, thereby permitting the
interested reader to explore a given topic further. Illustrations and summa-
rizing tables are in included in many chapters. We thank the contributing
authors for their willingness to undertake the task of making available to the
scientic community their knowledge and experience in this fast-develop-
ing eld and for providing a rational summary of what is known or yet to
be known, thereby contributing to the long-term quality of life for patients
with skeletal disease. We also thank the publisher for being patient and for
helping us to make this a pleasing and readable book

May, 2005.

Felix Bronner Mary C Farach-Carson Janet Rubin


Farmington, CT Newark, DE Decatur, GA
Contents

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi

1. Osteoclast: Origin and Differentiation


Janet Rubin and Edward M. Greeneld . . . . . . . . . . . . . . . . . . . . . 1

2. Osteolytic Enzymes of Osteoclasts


Merry Jo Oursler . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24

3. Regulation of Osteoclast Activity


Roland Baron and William C. Horne . . . . . . . . . . . . . . . . . . . . . . . 34

4. Structural Aspects of Bone Resorption


Steven D. Bain and Ted S. Gross . . . . . . . . . . . . . . . . . . . . . . . . . . . 58

5. Inammatory Cytokines
Mark S. Nanes and Roberto Pacici . . . . . . . . . . . . . . . . . . . . . . . . 67

6. Genetics and Mutations Affecting Osteoclast Development


and Function
Mark C. Horowitz, Melissa A. Kacena, Joseph A. Lorenzo . . . . . . . 91

7. Clinical Disorders Associated with Alterations in


Bone Resorption
Janet Rubin and Mark S. Nanes . . . . . . . . . . . . . . . . . . . . . . . . . . . 108

8. Pharmaceuticals for Bone Disease Targeting the Osteoclast


Lorraine A. Fitzpatrick . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128

9. Skeletal Complications of Malignancy: Central Role for


the Osteoclast
Gregory A. Clines, John M. Chirgwin, Theresa A. Guise . . . . . . . . . 151

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
Contributors

Steven D. Bain, PhD


SkeleTech, Inc
Bothell, WA
USA

Roland Baron, DDS, PhD


Department of Orthopedics and Rehabilitation
Department of Cell Biology
Yale University School of Medicine
New Haven, CT
USA

Felix Bronner, PhD


Department of BioStructure and Function
Department of Pharmacology
The University of Connecticut Health Center
Farmington, CT
USA

John M. Chirgwin, PhD


Department of Internal Medicine
Division of Endocrinology
University of Virginia
Charlottesville, VA
USA

Gregory A. Clines, MD, PhD


Department of Internal Medicine
Division of Endocrinology
University of Virginia
Charlottesville, VA
USA

Mary C. Farach-Carson, PhD


Department of Biology
University of Delaware
Newark, DE
USA
xii Contributors

Lorraine A. Fitzpatrick, MD
Global Development
General Medicine Therapeutic Area
Amgen, Inc.
Thousand Oaks, CA
USA

Edward M. Greeneld, PhD


Department of Orthopedics
Case Western Reserve University
Cleveland, OH
USA

Ted S. Gross, PhD


Department of Orthopedics and Sports Medicine
University of Washington
Seattle, WA
USA

Theresa A. Guise, MD
Department of Internal Medicine
Division of Endocrinology
University of Virginia
Charlottesville, VA
USA

William C. Horne, PhD


Department of Orthopedics and Rehabilitation
Yale University School of Medicine
New Haven, CT
USA

Mark C. Horowitz, PhD


Department of Orthopedics and Rehabilitation
Yale University School of Medicine
New Haven, CT
USA

Melissa A. Kacena, PhD


Department of Orthopedics and Rehabilitation
Yale University School of Medicine
New Haven, CT
USA

Joseph A. Lorenzo, MD
Department of Medicine
Division of Endocrinology
University of Connecticut Health Center
Farmington, CT
USA
Contributors xiii

Mark S. Nanes, MD, PhD


Department of Medicine
Division of Endocrinology and Metabolism
Veterans Affairs Medical Center
Emory University School of Medicine
Decatur, GA
USA

Merry Jo Oursler, PhD


Endocrine Research Unit
Mayo Clinic College of Medicine
Rochester, MN
USA

Roberto Pacici, MD
Department of Medicine
Division of Endocrinology and Metabolism
Emory University School of Medicine
Atlanta, GA
USA

Janet Rubin, MD
Division of Endocrinology and Metabolism
Emory University School of Medicine
Veterans Affairs Medical Center
Decatur, GA
USA
1.
Osteoclast: Origin and Differentiation
Janet Rubin and Edward M. Greeneld

hydroxyapatite surface has led to a fuller under-


Introduction standing of this important cell.
In this introductory chapter we set out the
The skeleton is an engineering feat: it is strong basic aspects of osteoclast origin and differ-
but light enough to permit locomotion, rigid entiation. Beginning with a description of the
to allow muscles to be xed but capable of bend- multinuclear cell found in bone and a brief
ing without breaking, and its structure is pro- introduction to its typing through functional
grammed for variable loading (Figure 1.1). In characteristics, we discuss developing research
addition to its structural role, the skeleton from a historical viewpoint. However, attention
serves as a reservoir and sink for calcium, an ion will be focused on the current understanding of
that plays an important role in cell metabolism osteoclastogenesis, shown in Figure 1.2.
and function. This means that bone must be
malleable, that its calcium stores must be acces-
sible, and that its structure can be sampled, The Osteoclast: Lineage of the
adapted, or ne-tuned, in a process termed
remodeling. The primary step in remodeling is
Multinucleated Macrophage
the resorption of bone. The multinucleated
osteoclast, a highly specialized cell, is res- The osteoclast was recognized in 1873 as the
ponsible for resorbing the mineral phase of the principal multinuclear cell that digested bone
skeleton. [87], but its origin was not uncovered until more
The osteoclast, therefore, is a basic element in than a century later. Kahn and Simmons were
skeletal physiology. Abnormalities in osteoclast able to separate the lineage of osteoclasts from
development or function that severely limit its the other key cell in remodeling, the osteoblast,
ability to resorb bone are incompatible with with a clever experiment in 1975. They took
normal locomotion, and, if severe enough, with advantage of the morphological differences
life. Defects in osteoclastogenesis can result in between cell nuclei of quail and chicken by
lethal neonatal disease because the bones lack a growing quail bone rudiments on well-
marrow space for hematopoiesis. Milder defects vascularized portions of chorioallantoic mem-
in osteoclastogenesis lead to crippling skele- branes of chicken embryos. The osteoblasts
tal lesions in childhood. In adulthood, excess appearing a week later were of quail origin, but
osteoclast action due either to the presence of the nuclei of the osteoclasts had a predomi-
too many osteoclasts or hyperactivity of those nantly chicken morphology [75]. This was the
present is the predominant cause of most rst convincing evidence that osteoclasts were
skeletal pathology. Within the past ten years, hematogenous in origin. Several years later,
research into the origin of the osteoclast and the building on work showing that cure of osteopet-
signals that direct its formation at a specic rosis in mice was possible by transplantation of
1
2 Bone Resorption

contact with the calcied matrix [17]. This


demonstrated that recreation of the bone envi-
ronment provided sufciency for transforma-
tion of hematologic precursors into osteoclasts.
The need for elements of the bone environ-
ment to allow osteoclastogenesis has become
key to understanding the intricate relationship
between bone osteoblasts/stromal cells and
osteoclasts.
The understanding that osteoclasts arose
from hematopoietic stem cells (HSC) created a
need for adequate typing to ensure that multi-
nucleated osteoclasts were not other cells
arising from the HSC. Indeed, many character-
istics of osteoclasts are shared by macroph-
ages, including the ability to fuse with like cells
and the ability to phagocytose. However, macro-
phages associated with bone surfaces do not
have the rufed border that is characteristic
of the osteoclast, nor do they respond to calci-
tonin with a change in shape. Moreover, and
most convincingly, macrophages are unable to
rescue an osteopetrotic decit [184]. Cell mor-
phology, staining for functional characteristics
such as tartrate resistant acid phosphatase, and
the ability to respond to calcitonin, were all
found to be useful for differentiating osteoclasts
from macrophages. Many laboratories con-
Figure 1.1 The skeleton as interpreted by Vesalius. tributed to a more exacting denition of cell
type by examining cell surface characteristics.
For instance, osteoclasts lack the Fc and C3
surface receptors that typify macrophages. As
normal bone marrow [187], the rst successful pre-osteoclasts differentiate, they loose their
bone-marrow transplantation was performed in early macrophage characteristics, decreasing
a female human infant with malignant osteopet- expression of macrophage-specic surface anti-
rosis. In the normally remodeling bone, the gens F4/80, Mac-1. Their ability to phagocytose
osteoclasts appearing in situ lacked Barr bodies, is also diminished [20, 185]. In comparison with
as would be expected in male nuclei: indeed, the macrophage polykaryons, osteoclasts express a
bone marrow had been harvested from the variable but restricted range of macrophage-
patients brother [24]. associated antigens (e.g., CD13, CD15A, CD44,
With evidence that the osteoclast arose from ICAM-1), whereas macrophage polykaryons
fusion of hematogenous cells, the next step in strongly express CD18, CD14, CD31 [10]. This
determining the lineage was to develop an in allows for a discrete separation of osteoclasts
vitro process to obtain polykaryons capable of from macrophage multinuclear cells.
resorbing calcied bone matrix. Neither mono- Cell typing that largely pertains to the
cytes nor macrophages t this denition, available and developed antibodies has led to
although they are able to resorb some mineral. further demarcation between osteoclasts and
Burger et al. [17], culturing hematopoietic cells of macrophage lineage [59, 60]. Currently
tissue, showed that at two weeks the culture markers used during ow cytometry delineate
consisted largely of immature and mature the extended progression from stem cell to
mononuclear phagocytes, but contained no the osteoclast phenotype. Indeed these steps
osteoclast-like cells. When, however, stripped constitute a road map of the differentiation
bone rudiments were added to the hematopoi- program. (These are shown along the top of
etic cell culture, osteoclasts appeared in close Figure 1.2.)
Osteoclast: Origin and Differentiation 3

HSC CMP CFU-GM CFU-M pre- migration/ multinucleated osteoclast


Sca-1+ CD34+ EpoR- GATA-2- osteoclast adhesion cell
FCR-hi GATA-2+ NF-E2-
IL-7R- NF-E2+ C/EBP+

?
PU.1 MCSF RANKL TRAF6
MCSF RANKL Fusion src
RANKL
Fos molecules 3 integrin
p50/p52 RANKL
mi/Mitf

F4/80

MCSF-R
RANK
MMP-9 Nonspecific
esterase
TRAP
CTR
CAII
3 integrin

Figure 1.2 Osteoclast Differentiation. The diagram shows the progression from the hematopoietic stem cell (HSC) through the
common myeloid progenitor (CMP) to the colony-forming unit for granulocytes and macrophages (CFU-GM) to the CFU for
macrophages and into the osteoclast lineage. Markers that can be used to identify early stem cells are shown above the cells. Factors
that stimulate progressing are shown in the arrowed boxes. The boxes on the bottom of the diagram show appearance of markers
associated with osteoclast lineage.

In vitro co-culture systems that adequately enhance osteoclast formation [95]. The effect of
simulate the bone microenvironment have been these CSFs gave support to the idea that
a major tool for study of progression along the macrophages and osteoclasts share a common
osteoclast lineage. Using variations on the origin. Experiments also made it possible to
Dexter marrow culture [28, 29], bone biologists identify three stages in osteoclast differentia-
found that it was possible to generate osteoclasts tion: early differentiation, where hematopoietic
in the presence of osteoclastogenic agents in stem cells proliferate within the macrophage
primate [141], feline [65], and murine cultures lineage [53]; progression into the early osteo-
[95, 162, 168]. Such culture systems allowed the clast precursor stage with expression of calci-
identication of factors that enhanced osteoclast tonin receptors and TRAP [51]; and a later stage
development such as 1,25-dihydroxyvitamin D where fusion occurs [13]. The extensive osteo-
[141], prostaglandins [5], and cyclic AMP [148], clastogenic marrow culture literature adds to the
as well as parathyroid hormone [6]. Within conclusion that the osteoclast precursor begins
the context of murine marrow cultures, other as a nonadherent hematopoietic stem cell and
cytokines such as IL-6 and TNF have also been progresses through the colony-forming unit for
shown to regulate osteoclast differentiation [3, granulocytes and macrophages (CFU-GM) to
182]. Dexters culture [29] had opened the way become a CFU-macrophage (CFU-M) before
to understanding the role of colony-stimulating entering the osteoclast lineage.
factors (CSFs) in blood cell proliferation and As stated above, accessory cells representing
differentiation. It therefore seemed logical to bone stromal elements are necessary for osteo-
expect that colony-stimulating factors would clast formation. In addition, cellcell contact is
also play a role in osteoclast development. required between the myelocytic precursor and
Macrophage CSF (MCSF) was found to be essen- the bone stroma [163, 203]. The critical factor
tial for osteoclastogenesis [86] and granulocyte- contributed by bone stromal cells for terminal
macrophage-CSF (GM-CSF) was shown to osteoclast differentiation was identied in 1998
4 Bone Resorption

as RANKL, a member of the tumor necrosis myeloid precursor, CMP, from the common
factor family [97, 206]. The discovery of this lymphoid precursor as an IL-7Ra-, GATA-2+,
factor has permitted osteoclast biology to shift NF-E2+, GATA-1+, GATA-3- cell [4]. It is this cell
into high gear, as will be described below. that responds to signals that generate prolifera-
tion of the CFU-GM, as distinguished from the
lineage providing cells for megakaryocyte or
erythrocyte lineage, or the earlier fork where
Early Differentiation the HSC enters the lymphoid pathway.
Interestingly, the absence of the Pax5 gene, a
The rst process we will examine is the pro- key to the B-lymphoid lineage, leads to enrich-
gression from the pluripotent hematopoietic ment of the myeloid lineage in transgenic
stem cell to the colony-forming unit for Pax5-/- animals [132]. The Pax5-/- pro-B cells
macrophages. respond to M-CSF with expression of
macrophage markers Mac-1 and F4/80, and,
Myeloid Differentiation in the presence of RANKL, into osteoclasts.
Although scientists were unable to force the
The pluripotent hematopoietic stem cell (HSC) pro-B cells into erythroid or megakaryocytic
in the marrow cavity awaits signals that direct lineage, pro-B cells clearly maintain a degree of
its development into the myeloid lineage. This plasticity even after they are committed to a
requires expression of molecules that allow lymphoid lineage [132].
response to osteoclastogenic factors. The HSC The factors responsible for herding HSC into
bears stem cell antigen-1 (Sca-1+), which indi- the common myeloid progenitor lineage and
cates a self-renewing capacity [135, 159]. The then into the CFU-GM are multiple and very
rst way station in HSC differentiation is a likely redundant. By means of targeted dele-
choice between entry into the myeloid or the tions of early lineage markers from embryonic
lymphoid progenitor pool. Expression of the stem cells, GATA-2 and SCL/tal-1 were shown
interleukin-7 receptor a-chain (IL-7Ra) differ- to be important for early development [202].
entiates the common lymphoid progenitor The SCL/tal-1 transcription factor was rst
(CLP) that reconstitutes T-cells, B-cells, and described as an abnormal translocation in T-cell
natural killer cells from cells of the myeloid leukemia and is necessary for generation of all
lineage [88]. hematopoietic cells. GATA-2, while not neces-
The immediate precursor of the myeloid sary for later expansion of osteoclast progeni-
progenitor population is identied by the ab- tors, has some role in separating CFU-M from
sence of the IL-7Ra. In a search for the earliest those cells that are most responsive to RANKL.
myeloid cell, Akashi and colleagues recently The colony-stimulating factors for macrophage
used cell sorting to discard IL-7Ra-positive cells and/or the granulocyte lineage cause cultured
that have entered lymphoid lineage. They next osteoclast numbers to swell in numbers by
selected an IL-7Ra-negative fraction expressing increasing the number of precursors [34, 53, 95,
the myelomonocytic Fcg receptor [4]. In the 160, 167, 175].
presence of GM-CSF and a mixture of cytokines
(steel factor, Flt-3 ligand, IL-11, IL-3, erythro- Becoming a Macrophage Progenitor
poietin, and thrombopoietin) 80% of this frac-
tion commits to myeloid lineage [4]. If this As the colony differentiates, a number of
lineage is further separated into a fraction with markers identify the emergence of the
high FcgR expression that also expresses the macrophage lineage. Macrophage colonies are
cell surface molecule sialomucin CD34, this readily identiable because they express a
fraction becomes almost exclusively myeloid. non-specic esterase, along with the receptor
FcgRhiCD34+ cells divide into three lineages that for the macrophage colony-stimulating factor.
contain macrophages and/or granulocytes, i.e., The movement from the common myeloid pre-
CFU-M, CFU-G, and CFU-GM. In contrast the cursor (CMP) through the CFU-GM and into the
FcgRloCD34+ behave like HSCs, and the FcgR- macrophage lineage is in fact, largely due to the
lo
CD34- give rise to colonies that contain only macrophage colony-stimulating factor (MCSF).
megakaryocytes and/or erythrocytes. Thus, The importance of MCSF to osteoclast lineage
Akashi was able to differentiate the common became apparent as the result of the discovery
Osteoclast: Origin and Differentiation 5

that the osteopetrotic (op/op) mouse, whose proliferation of the CFU-M [155]. The down-
marble bone disease and lack of dental eruption stream proliferative response to the MCSF
are lethal consequences of total osteoclast de- ligand includes induction of G2 progression
ciency, has a mutation in the gene that codes for through activation of cyclin genes. It is difcult
MCSF [193, 208]. Similarly, in the toothless rat, to separate the differentiative from the prolifer-
which displays the osteopetrotic phenotype, a ative responses in MCSF-R-bearing cells [74,
frameshift mutation in the MCSF gene causes 154]. Myeloid cells become macrophage, in
a loss of function that brings about a profound part, because the myeloid and B-cell-specic
deciency of osteoclasts and macrophages transcription factor PU.1 also increases. The
[186]. In culture, MCSF is more potent than GM- nding that PU.1-decient mice have a
CSF or interleukin-3 in stimulating osteoclast decreased number of macrophages and no
formation in primate bone marrow [137]. GM- osteoclasts gives support to the role played by
CSF cannot rescue the op/op MCSF decient PU.1 in macrophage recruitment [179].
mouse, but does support the development of The transcription factor fos, and perhaps
osteoclast-like cells if vitamin D is present in the other members of the AP-1 transcription factor
culture [93]. In contrast,VEGF can substitute for family, play an important role in osteoclast dif-
MCSF when administered to op/op mice and ferentiation. It has been long known that the v-
appears to be the molecule responsible for the fos oncogene induced by murine sarcoma virus
age-related resolution of osteopetrosis seen in causes osteosarcoma and that overexpression of
mice nursed through their early otherwise lethal the cellular homolog, c-fos, led to the develop-
inability to wean [131]. ment of bone tumors [149]. Because these fos
MCSF has importance as a growth and dif- lesions were in osteoprogenitor cells, not in the
ferentiating factor. It is also required for survival osteoclast lineage or in HSCs, it was surprising
of cells of the macrophage lineage [175]. In a that mice lacking c-fos developed osteopetrosis,
culture containing high levels of MCSF, mono- essentially an overgrowth of bone [48]. In fact,
cytes will increase in number at the expense c-fos is not required for normal osteoprogeni-
of osteoclastogenesis, probably because MCSF tor development but is required for osteoclast
strongly promotes the development of mononu- differentiation: c-fos-negative HSC progress
clear monocytes [34, 136]. Yet, in the case of normally into CFU-M and from there to
estrogen deciency, increased levels of MCSF in become terminally differentiated macrophages,
bone marrow may enhance osteoclastogenesis but osteoclasts are totally absent in this mouse.
[81]. These discrepancies may be due to differ- Whether the requirement for fos is in the
ences in the expression patterns of MCSF, result- stromal accessory cell (e.g., for normal ex-
ing from at least two splice variants. One of pression of RANKL), or only in the osteoclast
these is a secreted soluble factor. The other, rep- progenitor (e.g., required for response to osteo-
resenting a much smaller amount of the total clastogenic signals) has not yet been worked
MCSF, ~5%, is expressed on the surface of bone out. However, overexpression of c-fos in osteo-
stromal cells [35, 204]. Because the insoluble clast precursors can increase osteoclast differ-
form of MCSF is anchored and displayed by cells entiation [122] and RANKL does induce
within bone, this isoform may represent a local transcription of genes through c-fos activation.
signal that regulates osteoclast development. This suggests a link between RANKL signaling
Indeed in the op/op MCSF-decient mouse, the and c-fos activation [115]. RANKL has, in fact,
membrane-bound form of MCSF can correct been shown to induce interferon-B in osteoclast
the osteoclast defect [205]. Perhaps the best precursor cells. This interferes with RANKL-
way to summarize the literature is to state that induced expression of c-fos and decreases
MCSF expands the precursor pool capable of osteoclastogenesis [172].
responding to osteoclastogenic signals from the Other members of the AP-1 family are also
bone marrow and, at the same time, enhances involved: Fra-1 can rescue the c-fos defect in
macrophage differentiation and survival. osteoclast precursors [38]; this may suggest
The CFU-GM expresses the MCSF receptor, some redundancy between AP-1 members in
which is a membrane tyrosine kinase encoded intracellular signaling leading to osteoclastoge-
by the gene c-fms [153]. Multiple second mes- nesis. Overexpression of Fra-1 by itself, also,
sengers that are activated through this mem- leads to increased osteoblast differentiation and
brane tyrosine kinase receptor trigger thus to a progressive increase in bone mass [73].
6 Bone Resorption

Expression of AP-1 proteins therefore plays crit- nous precursors [35, 190]. Moreover the acces-
ical roles in osteoclast and osteoblast progeni- sory cells actually had to be in direct contact
tors and is necessary for normal coupling with the osteoclast progenitor cells. Separation
between the remodeling cells. by a membrane prevented the process [164].
This meant that the stimulated stromal cells,
even when xed, provided a contact signal to the
CFU-M into Pre-Osteoclast: CFU-M. What was this signal?
Laboratories working in Japan and
The RANKL:RANK Signal California discovered the answer simultane-
ously. TNF-related activation-induced cytokine,
RANKL Comes Calling or TRANCE, originally described as a factor that
activated signals in T-lymphocytes [197], was
The progression of the macrophage colony- found to be the osteoclastogenic factor that is
forming unit into the osteoclast lineage has now known as Receptor Activator of NFkB
been traditionally characterized by develop- Ligand or RANKL [97, 206] (Figure 1.3). Dis-
ment of a panel of osteoclastic phenotypic fea- covery of RANKL involved similar strategies
tures. These include expression of the calcitonin by both groups, using a previously identied
receptor, of enzymes such as tartrate-resistant inhibitory binding factor to screen for the
acid phosphatase and carbonic anhydrase II, osteoclastogenic factor. Yasudas team used an
and the ability to resorb bone [12, 52, 102, 103]. osteoclast inhibitory factor from bone-cell-
Other established markers are the vitronectin conditioned media that eventually was shown to
receptor and the vacuolar-type proton pump be the TNF-R-like decoy receptor osteoprote-
[94]. Although the progression from the mono- gerin, to tag expression libraries encoding the
cytic precursor to the osteoclast progenitor ligand partner for the inhibitory protein [206].
was thus clearly established, the critical They called their gene product osteoclast dif-
factor responsible for this metamorphosis was ferentiation factor and showed that it induced
unknown. osteoclast formation from spleen cells in the
Experiments had shown that when the absence of stromal elements. Osteoclast dif-
CFU-M (or its earlier precursors) collected from ferentiation factor expression was upregulated
bone marrow or spleen was added to a culture in stromal cells by osteoactive factors such as
of osteoprogenitor cells, to which any one of parathyroid hormone and 1,25-dihydroxyvita-
a handful of osteoclastogenic hormones or min D. Laceys group in California had dis-
cytokines was added, osteoclastogenesis could covered the same soluble inhibitory binding
proceed [112]; the osteoclastogenic factors protein, named it osteoprotegerin (OPG), and
acted synergistically [139] and furthermore showed that it prevented osteoclastogenesis in
appeared to target the stromal accessory cells, vivo [157]. Searching for the binding partner of
since the accessory cells could be pretreated OPG, they cloned osteoprotegerin-ligand
with agents such as 1,25-dihydroxyvitamin D (OPGL) from a murine expression library [97].
and xed prior to culture with the hematoge- OPGL bound to hematopoietic progenitor cells

Figure 1.3 RANKL. The drawing shows the


protein structure of RANKL.
Osteoclast: Origin and Differentiation 7

and rapidly induced osteoclast genes. The nal associates into a homotrimer, similarly to other
selection of the term RANKL, or receptor acti- TNF molecules with four unique surface loops
vator of NFkB ligand [7] as the name of the OPG that are necessary for full activation of RANK.
binding partner in place of ODF, OPGL or the Once the identity of RANKL as the factor
earliest manifestation, TRANCE, helped to clear necessary for osteoclastogenesis was conrmed,
the alphabet confusion. osteoclast biology became much more trans-
RANKL thus was identied as a membrane- parent. First, the role of the accessory cell, and
associated protein of the TNF ligand family that indeed the cellcell contact necessary between
is expressed by stromal cells after stimulation by the support cells and the osteoclast precursors,
bone resorbing cytokines/hormones. Anderson was found to be a function of the presence of
et al. also cloned by direct expression a gene that RANKL expressed on the accessory cell mem-
they called RANKL (the earliest and nal desig- brane. Indeed, RANKL is the bone environment.
nation) as the ligand partner to a receptor When RANKL was expressed in non-bone cells
termed RANK, that they had previously char- osteoclastogenesis was induced [101]. Further-
acterized [7]. The RANKL cDNA clone has an more it was possible to x these non-bone cells
open reading frame that encodes a 316 amino with paraformaldehyde prior to adding the HSC
acid type II transmembrane protein (Figure responders [206], as is also true for bone cells
1.4). A 48 amino acid intracellular amino- [35]. Interestingly, the need for a stromal cell
terminus precedes the 24-residue hydrophobic could also be overcome by the addition of the
domain that passes through the plasma mem- extracellular domain of RANKL [97, 206]. MCSF,
brane. The carboxy-terminal residues have which also is made by the stromal cells, is nec-
signicant homology to members of the TNF essary for proliferation of the progenitor pool,
ligand family. Wong et al. remark on similarity as well as for survival of cells of macrophage
of the mouse TRANCE with TRAIL, FasL, and lineage; in the absence of stromal cells, a source
TNF, especially in those regions that form the of MCSF must also be provided [181]. Soon it
beta strands seen in the TNF crystal structure was realized that soluble RANKL, either
[197]. Furthermore, there are putative N-linked secreted directly by some cells [89], or cleaved
glycosylation sites, all of which are highly from its membrane position by a metallopro-
conserved between mouse and human. Lam teinase [110, 129], also had a role in osteoclas-
and colleagues successfully crystallized the togenesis. Bone stromal cells have not been
ectodomain of murine RANKL [98]: RANKL shown to generate secreted RANKL under

Signal transduction

Transcription regulators

Figure 1.4 RANK signal transduction.


RANK signaling is complex and involves
multiple cascades. This diagram does not
rule out interaction between the signals,
some of which may be redundant.
8 Bone Resorption

known osteoclastogenic stimuli: conditioned tiation appears to be the osteoclast precursors


media from stromal cells are ineffective in gen- themselves, rather than the accessory stromal
erating osteoclastogenesis. Spot cultures of cells [99, 139].
stromal cells, in fact, generated osteoclasts only RANKL mRNA expression by stromal cells
in the local area of RANKL-expressing cells. has become a reliable marker of the osteoclas-
This further underscores that cellcell interac- togenic potential of the microenvironment or
tions are involved in bone cell RANKL:RANK culture system. IGF-I, which stimulates long
signaling [69]. bone apposition during growth, upregulates
The murine gene for RANKL exists as a single RANKL [144]. Equally important is the fact that
copy of ve exons spanning about 40 kilobases when a load is applied to cultured cells, RANKL
[85]. The intracellular and transmembrane expression decreases [146], whereas when
domains are encoded in the rst exon, with the weightlessness is simulated in culture, RANKL
important ligand domain beginning in the rst increases [76]. Disease states are also associated
exon and continuing through the fth. The with changes in RANKL expression. In Pagets
organizational structure is similar to that of the disease, where localized and intense bone
other members of the TNF family, with mouse remodeling occurs, RANKL expression is
RANKL having most homology to CD40L, increased in marrow samples from affected, but
which also has ve exons. Both molecules play a not from unaffected, bones [120]. The expres-
role in dendritic cell function [85]. Ablation of sion of RANKL in the estrogen deciency state,
the RANKL gene led to some surprising pheno- where increased resorption leads to decreased
typic effects in the transgenic mouse [90]. The bone mass, was not convincingly increased in
RANKL-null mouse had severe osteopetrosis the marrow [156]. Rather, as discussed below,
due to a complete absence of osteoclasts, but it the response of the osteoclast precursor to
also had unexpected defects in both T and B RANKL signaling via RANK is diminished.
lymphocytes and no lymph nodes. The rele- Both early bone stromal cells and osteoblasts
vance of RANKLs regulation of lymph-node express RANKL, and several authors have sug-
organogenesis for osteoclastogenesis has not yet gested that the differentiation state of the bone
been worked out, but may be important in situ- cell may affect the level of response to the stim-
ations where osteopetrosis is accompanied by ulatory agent. While this certainly would hold in
impaired lymphocyte development. terms of response to PTH, where PTH-R are not
expressed until after the cell is well on its ter-
minal differentiation path [117], this is less clear
Control of RANKL Expression by for unstimulated expression of RANKL. RUNX2,
Accessory Cells an essential transcription factor for osteoblast
differentiation, must be present before RANKL
Before RANKL, the response of the osteoclast- is expressed [42]. This suggests that the earliest
generating unit, i.e., HSC plus accessory cells, stromal cells may not have the machinery to
was studied as a response of one or both of these induce RANKL transcription. However, less
elements to agents known to increase bone mature osteoprogenitor cells may have an
resorption in the whole animal. Thus, hormones increased basal expression of RANKL [44,
such as 1,25-(OH)2D3 and PTH were known to 178].
stimulate osteoclastogenesis, as were inamma- It should be noted that it has been difcult
tory cytokines, such as tumor necrosis factor to measure RANKL protein in stromal cells.
and many of the interleukins [163, 165]. Of the While soluble RANKL can be measured by a
factors that increase osteoclastogenesis in commercial ELISA, both ELISA and Western
culture, most, if not all, increase RANKL expres- analysis of membrane-associated RANKL have
sion by the accessory cell. Parathyroid hormone been exceedingly difcult. An OPG-based pull-
increases RANKL expression in a dose-depen- down assay echoing original strategies for iden-
dent fashion within at least 24 hours [105], as tifying RANKL has been useful in some cases
does 1,25-(OH)2D3 [61, 147]. The major excep- [130] but its tendency to bind other members of
tion to this paradigm is stimulation of osteo- the TNF family limits its usefulness. Because
clast differentiation by TNF. Although TNF there exist three isoforms of RANKL, one with
stimulates RANKL expression, its primary a shorter intracellular domain, and one with no
target during stimulation of osteoclast differen- transmembrane domain at all that may repre-
Osteoclast: Origin and Differentiation 9

sent the secreted factor [66], the assay is com- terminal kinase (JNK), ERK1/2 kinase, p38
plicated further. kinase, and c-src (see Figure 1.4).
Despite problems with measurement of the The extracellular domain of RANK binds
RANKL protein, the endogenous mRNA for both the membrane and soluble forms of
RANKL in stromal cells responds to stimulation RANKL as well as other members of the TNF
by increasing robustly. This in turn is associated family. The intracellular portion of the molecule
with an increase in the cultures potential to has domains that interact with the TRAF family
generate osteoclasts from precursor cells [147]. (TNFR-associated factor) proteins. TRAF pro-
Attention to the RANKL promoter should there- teins serve as adaptor proteins that recruit and
fore have led to an understanding of the mech- activate downstream transducers [62, 198]. The
anism by which bone stromal cells regulate amino termini of TRAFs are characterized
RANKL expression. This has not been the case. by a RING nger domain that is required for
Nearly 1,000 base pairs of murine and human interaction with other proteins, and for the sub-
RANKL promoters were cloned shortly after sequent release of NFkB from its cytoplasmic
RANKL was identied [82]. However, RANKL anchor. The signal cascade activated through
expression should be tissue restricted, and the TRAF binding is complex, but certainly involves
available promoter sequences are not sufcient TRAFs 2, 5, and 6 through multiple associations
to limit expression or to provide the expected on the RANK intracellular domain [26]. TRAF2
response to agents known to increase RANKL knockout mice do not have obvious problems
mRNA expression [133]. Novel methods, with osteoclast recruitment [128, 207], while
perhaps involving study of the local chromatin TRAF6 knockout mice have osteopetrosis [106].
structure, may be needed to understand tissue TRAF6 thus is recognized as the key adaptor to
expression and its regulation. activated RANK.
In vivo, non-bone cells can also express Experiments using RANK constructs mut-
RANKL, as shown by localization in white cells ated for selective binding of various TRAF
and the effects of RANKL in lymph nodes and proteins have shown that while some TRAF
dendritic cells [7, 191, 197]. T-lymphocytes proteins are redundant, TRAF6 was absolutely
secrete a soluble form of RANKL into inamed required for the formation of cytoskeletal struc-
rheumatic joints, causing osteoclastic bone tures and to permit the osteoclast to resorb
destruction [89]. There is evidence that T cells bone [9]. Kobayashis study of TRAF6 structure
with RANKL expression may be key elements suggests that the RING nger domain of the
in the bone loss caused by estrogen deciency protein, while necessary for IL-1 and LPS signal
[19, 180]. activation of NFkB, may not be required for
early osteoclast maturation [84]. Thus, while in
the absence of the RING nger several osteo-
RANK: RANKL Signal Transduction in clast marker genes such as cathepsin K, calci-
the Monocytic Cell tonin receptor and TRAP, are expressed, the
actin-sealing machinery necessary to complete
Stimulation of the Receptor Activator of NFkB, the seal of the resorption pit is not functional.
or RANK, with activation of NFkB, leads to the The RING mutant TRAF still activates NFkB,
development of the full osteoclast phenotype, but perhaps does not allow association with a
including fusion. RANK signaling also increases signicant adapter protein required for late dif-
the net resorptive activity, and, like MCSF, ferentiation. TRAF6 also activates transforming
promotes the survival of osteoclast cells. RANK growth factor b-activated kinase 1 (TAK1).
was rst identied in dendritic cells, whose Interestingly TAK1 mediates JNK and p38
immune surveillance mechanism processes and MAP-kinases. This may explain the ability of
presents antigens to T cells [7]. The osteoclast transforming growth factor b to enhance osteo-
precursor sequentially expresses the receptor clastogenesis in the presence of RANKL [152].
for MCSF and then RANK [8]. RANK is highly To complicate issues, MAPK activates TAK1!
expressed in isolated bone marrow-derived Thus Kobayashis nding that TRAF6 was nec-
osteoclast progenitor cells, as well as in mature essary for the terminal maturation steps, as well
osteoclasts [62]. Cognate ligand binding has as Wongs earlier work that showed TRAF acti-
been shown to activate multiple downstream vated c-src, a kinase necessary for the actin ring
signaling cascades, including NFkB, c-jun N- structure of the osteoclast [196], suggest that
10 Bone Resorption

TRAF6 is a specic transducer of later effects of homology [157]. Its importance was recognized
RANKL. even before its target ligand was identied,
The TRAFs have also been implicated in when OPG was used to identify RANKL as its
modulating the effects of many of the bone binding partner in experiments using expres-
active cytokines. For instance, interferon- sion libraries. The binding of RANKL by this
gamma strongly suppresses osteoclastogenesis soluble decoy receptor served to prevent
by silencing RANK signal transduction. This RANKL binding to its target on the osteoclast
process invokes interferon-gamma activation precursor thus the name, osteoprotegerin
of a ubiquitin-proteasome system, whereby (OPG), signies that it preserves bone. Indeed,
TRAF6 signals are abrogated when TRAF6 is OPG is often regulated in vivo and in vitro in
degraded [173]. inverse proportion to the levels of RANKL [57].
The rest of the seemingly redundant TRAFs OPG contains 401 amino acids, with a signal
play roles in the earlier stages of osteoclast for- peptide encoding its status as a secreted mole-
mation [9, 26]. Because multiple signaling cas- cule expressed in many tissues including liver,
cades are involved, (e.g., NFkB, JNK, p38), one lung, heart, and kidney and, importantly in bone
approach to disentangling the important signals and cartilage. It contains no hydrophobic amino
is to perform gene expression proling after acids that would make for a transmembrane-
RANKL stimulation of RANK. Four of more spanning domain, so after secretion it is not
than 100 early RANKL-inducible genes have cell-associated. The N-terminal protein has high
been found to be linked to induction of osteo- homology with other members of the TNFR
clast maturation [67]. One important gene can- superfamily, allowing binding of RANKL [97]
didate arising from that study was NFAT2 and of at least one other TNF family member,
(nuclear factor of activated T cells-2); suppres- TRAIL [30].
sion of NFAT2 with antisense interfered with Transgenic mice that overexpress OPG in
osteoclast formation. NFAT2-decient embry- the liver under the control of the human
onic stem cells do not respond to RANKL with apolipoprotein E gene promoter display
formation of osteoclasts. Moreover, overexpres- increases in bone density, even osteopetrosis, if
sion of NFAT differentiates precursors in the the hepatic OPG output is very high [157].
absence of RANKL [171]. NFAT molecules act as Recombinant OPG can be used to simulate the
co-factors with AP-1 or fos/jun proteins to bind phenotype of OPG-overexpressing transgenic
to regulatory cis DNA elements [171]. NFATs mice. It protects rats against ovariectomy-
may one day be shown to be part of the patho- associated bone loss. Osteoclastogenesis can be
physiology responsible for the osteopetrotic entirely inhibited if OPG is added to culture or
phenotype seen in the fosless mouse [48]. What is expressed by stromal cells and osteoblasts
are the postulated targets and partners of [183]. Conversely, OPG-decient mice develop
NFAT2? Clearly osteoclast marker genes such as an early-onset osteoporosis [16]. In fact, many
TRAP, calcitonin receptor and carbonic anhy- studies have shown that OPG regulates osteo-
drase II have multiple sites recognized by NFAT clastogenesis both in vivo and in vitro; it does
as well as its partner AP-1 [171]. so by preventing the effects of RANKL [96, 183,
P38 MAPK is another important downstream 210]. Interestingly, endothelial cells also express
signal of RANK, as it is involved in expression of OPG. This local OPG expression appears not
carbonic anhydrase II and TRAP through acti- only to modulate inammatory bone states [25]
vation of mi/Mitf [113]. As will be discussed but can also prevent the calcication of arteries
below, estrogen may exert its effects by damping as encountered in atherosclerosis [121]. OPG-
the action of the p38 MAPK. decient mice exhibit calcication of the large
arteries [16].
The regulation of osteoprotegerin secretion
Osteoprotegerin: A Second Binding from bone cells is thus of great interest. The
Partner for RANKL OPG promoter has twelve RUNX2 binding ele-
ments that underwrite OPGs strong expression
Osteoprotegerin (OPG) is the third element of in osteoblasts, which express RUNX2 after dif-
the RANK: RANKL: OPG triumvirate. Simonet ferentiation. Not surprisingly then, overexpres-
originally identied OPG as a novel member sion of RUNX2 increases OPG promoter activity
of the TNFR superfamily through sequence [177]. Transforming growth factor-b, which can
Osteoclast: Origin and Differentiation 11

both stimulate and inhibit osteoclastogenesis, clast differentiation induced by TNFa [139].
increases OPG expression [170]. Estradiol stim- Studies using either stromal cells [99] or osteo-
ulates OPG expression in vitro [150] and may clast precursors [139] from mice lacking TNF
increase OPG levels in vivo [166]. Leptin, a receptors have shown that it is the osteoclast
hormone of lipid metabolism that modulates precursors rather than the accessory stromal
bone remodeling by an as yet poorly under- cells that are the primary target of TNFa stim-
stood mechanism, also increases OPG [18]. As ulation. However, TNFa cannot fully replace
expected, factors that increase osteoclastogene- RANKL during osteoclast differentiation.
sis and bone resorption decrease OPG secretion Rather, the action of TNFa requires that a per-
by bone cells. Glucocorticoids, which cause missive amount of RANKL either be present in
osteoporosis by stimulating bone resorption the culture or be added with the TNFa [139].
and inhibiting bone formation, inhibit OPG Recent studies have shown that molecules other
while also stimulating RANKL expression [56]. than TNFa are synergistic with, or can replace,
Parathyroid hormone also decreases OPG and RANKL or MCSF in stimulating osteoclast dif-
increases RANKL in vitro [104]. It does so also ferentiation. These molecules include TGFb [41,
in vivo, provided the hormone is administered 152], BMP-2 [68], activin A [40],VEGF [131], t3
continuously [111]. Insulin-like growth factor-I ligand [100], MIP-1a [49], and prostaglandin E2
(IGF-I) also appears to downregulate OPG in [188]. Even this more complex view of the reg-
vitro and in vivo. This may account for the ulation of osteoclast differentiation is likely to
remodeling and increase in bone markers seen be an oversimplication. For example, the effect
in trials where IGF-I was given in an attempt to of TGFb appears to depend on the mixture of
increase bone density [144]. cell types and cytokines that are present in the
There is a growing appreciation of the role microenvironment in which osteoclast dif-
of OPG in rare metabolic bone diseases. In the ferentiation occurs [114]. In vivo osteoclast
autosomal recessive Juvenile Pagets disease, differentiation therefore must involve multiple
also known as hyperostosis corticalis defor- cytokines acting in a complex regulatory
mans juvenilis, a progressive osteopenic skeletal network. Nonetheless, it is the balance between
deformity arises out of intense bone remodel- RANKL and OPG that must be considered the
ing. Two unrelated patients with this disease primary regulator of osteoclastogenesis.
were shown to have a defect in the OPG gene,
resulting in undetectable serum OPG levels
[192]. Autosomal dominant familial expansile
osteolysis is another rare bone disorder associ-
Late Differentiation
ated with focal areas of osteolysis that arise
from a deletion of the OPG coding sequence Once the CFU-M has been exposed to RANKL
[64]. and has committed to osteoclast lineage, addi-
tional differentiation steps must occur, the most
striking of which is fusion of multiple commit-
Exceptions: Osteoclastogenesis ted cells into the osteoclast polykaryon. The
Without RANKL:RANK number of osteoclast nuclei that occupy the
same fused cell appears to inuence the ability
Although RANKL and MCSF are sufcient to of the polykaryons to resorb bone. For instance,
induce osteoclast differentiation in cell culture, in Pagets disease, those pagetic osteoclasts
it would be surprising, given the complexity of responsible for site-specic increases in bone
most other paracrine regulatory systems, if the turnover have more nuclei than normal osteo-
control of osteoclast differentiation in vivo clasts [91]. The increase in activity associated
involved only these two factors. The best- with a cell that contains several nuclei may be
characterized exception to the RANKL/MCSF due to the increase in cytoplasm and membrane
paradigm is stimulation of osteoclast differenti- area, allowing for the formation of resorption
ation by TNFa. Although TNFa stimulates pits in the bone area covered by the osteoclast
RANKL expression by stromal cells, it can [31]. It may also be due to the fact that the nuclei
induce osteoclast differentiation in the absence continue to be transcriptionally active [14].
of both accessory cells and exogenous RANKL Indeed, the calcitonin-mediated decrease in
[11, 83]. OPG, moreover, does not block osteo- osteoclast resorptive activity appears due to a
12 Bone Resorption

decrease in transcription by the osteoclast ing the expression of membrane signals neces-
nuclei [14]. sary to induce fusion [37].
The disintegrin eichistatin, derived from
snake venom, inhibits alpha(v)beta3 integrin
Fusion of Mononuclear (avb3) by blocking RGD (arginine-glycine-
Osteoclast Precursors asparagine) sites to which the integrin binds.
Eichistatin binds to osteoclast membranes and
Cell fusion is not a widespread phenomenon. completely inhibits the formation of multinu-
As a rule skeletal myoblasts, syncytial tropho- cleated osteoclasts, even though it cannot do so
blasts, megakaryocytes, and cells of macrophage when introduced in an earlier step of osteoclast
lineage are the only cells that fuse. It is likely differentiation [127]. Eichistatin action is partly
that the fusion processes in these cells and in due to inhibition of migration, a process that
the fusion of spermatocyte with oocyte or of allows precursor cells to co-locate, and partly
enveloped viruses with host cells involve similar to detrimental effects on the fusion process
mechanisms. Cell fusion involves many factors, itself.
including proteins that dissolve discrete areas of The extracellular matrix, upon which the
membrane. An example of the latter is disinte- precursor cells are located, is also important in
grase meltrin-alpha [199], a molecule that is supporting fusion. This is illustrated by the role
also expressed by osteoclast precursors [2]. of ascorbic acid which, when present in the
Other molecular factors are the receptor-ligand culture, causes the fusion of precursors to
partners such as the HIV gp160 envelope glyco- increase [138]. E-cadherin, which binds to cell
protein and the CD4 molecule that are adhesion molecules and is expressed in marrow
expressed in virus-induced cell fusion and in mononuclear cells, is also involved in the fusion
the osteoclast [124]. Syncytin, an aptly named process [116].
protein expressed by trophoblasts, stimulates The macrophage fusion receptor, also called
cell:cell fusion and is also expressed by retro- SHPS-1, is another transmembrane glycopro-
virus [209]. tein that appears to have a role in macrophage
Ketoconazole, long known to inhibit cell fusion leading to multinucleation [50]. The
fusion, also diminishes the fusion of osteoclasts absence of SHIP, a protein that blocks PI-3
in culture [33]. Ketoconazole inhibits HMG- kinase signaling in transgenic animals, leads to
CoA reductase, thus decreasing cholesterol enlarged osteoclasts that may contain upwards
biosynthesis and the production of N-linked of 100 nuclei; clearly when precursors undergo
oligosaccharides necessary for the fusion of excessive fusion, excessive bone resorption
myoblasts in culture [71]. Mannose is an results [174]. The purinergic receptor expressed
oligosaccharide that is expressed on the outer on cells of macrophage lineage is another target
membrane of osteoclast precursors; mannose of anti-fusogenic factors [32].
residues are necessary for the osteoclast fusion The control of osteoclast fusion thus involves
process [92], probably the mannose receptor multiple membrane proteins that regulate cell
enhances cellcell binding [125]. Interleukin-13 migration, cellcell attachment, and intracellu-
increases fusion of macrophages through lar signaling processes. RANKL [72] and 1,25-
upregulation of the mannose receptor [27]. This (OH)2D3 [1] are the major determinants of cell
may be an important mechanism by which fusion and the differentiation pathway.
inammatory cytokines upregulate pathologic
bone loss. In fact, decreasing the oligosaccha-
ride binding partners of the mannose receptor MITF Involvement in
decreases bone loss. Interestingly, cholesterol Late Differentiation
depletion by means of an HMG-CoA reductase
inhibitor like that used to treat hypercholes- Interestingly, the mi/mi microphthalmic
terolemia also decreases the fusion of TRAP- mouse held a clue to late differentiation events
positive mononuclear cells [151]. The ability of well before any of these events were under-
bisphosphonates to inhibit cholesterol synthesis stood. Examination of osteoclasts in these
may in fact contribute to their inhibition of osteopetrotic mice revealed that mi/mi osteo-
osteoclast differentiation, doing so by prevent- clasts were mononuclear and lacked rufed
Osteoclast: Origin and Differentiation 13

borders [176], even though they expressed other


osteoclast enzymes [45, 54]. Other aspects of
the mi/mi phenotype, such as reduced eye size
and pigmentation, involve expression of a gene
that encodes a basic helix-loop-helix-leucine
zipper transcription factor [55], now known
as micropththalmia transcription factor, or
MITF.
MITF plays a role in osteoclastogenesis, by
increasing the expression of TRAP through a
conserved sequence (GGTCATGTGAG) that is
located in the TRAP proximal promoter [108].
It also interacts with the PU.1 transcription
factor [109]. MCSF, another early differentiation
factor, induces phosphorylation of MITF,
thereby triggering recruitment of the transcrip-
tional co-activator, p300 [189]. Cathepsin K is
downstream from these partners, as MITF
upregulates expression of this important osteo-
clast enzyme [126].

Other Factors in Late Differentiation


Other factors that act on late differentiation
continue to be discovered through screening
osteoclast libraries. One such factor cloned from
an immortalized murine osteoclast precursor is
ADAM8, a disintegrin-like meltrin (see above).
ADAM8 increases osteoclast formation during
the later stage by an as yet unknown mecha-
nisms [22].
Figure 1.5 Toulouse-Lautrec: pycnodyostosis phenotype.
Matrix recognition, needed to accomplish
fusion or to identify bone, is a function of cell
surface integrins or adhesion molecules. As
noted above, the alpha(v)b3 integrin (avb3) to be the phenotype exhibited by the great
has been implicated in bone resorption. Mice, French painter, Toulouse-Lautrec (Figure 1.5).
in whom the b3 integrin subunit was deleted, Another unexpected requirement is for the
develop osteosclerosis notwithstanding an ubiquitous intracellular tyrosine kinase c-src,
increased number of multinucleated osteoclasts which is needed to form the rufed borders that
[118]. These b3 subunit null osteoclasts do not seal the resorption bay at the basal surface of the
adequately resorb bone due to a cytoskeletal osteoclast [15, 158].
defect leading to dysfunctions in their ability to
spread and form the actin ring/rufed borders
that are necessary for sealing and bone excava-
tion. For osteoclasts to function, therefore, the Stimulators and Repressors of
entire resorptive apparatus must be expressed.
Any one of multiple osteoclast enzyme de-
Osteoclast Differentiation
ciencies, even if the deciency does not affect
osteoclast recruitment, can lead to osteopetro- Of the many regulators of osteoclast differenti-
sis due to defective bone resorption. For ation, some of the more important are discussed
instance, a defect in cathepsin K results in the briey below, in an effort to provide an overview
curious disease of pycnodyostosis [43], typied of the large number of factors that affect
by osteosclerosis and short stature, and thought osteoclastogenesis.
14 Bone Resorption

Hormones the FGF-2-replete and FGF-2-null cultures. The


RANKL:OPG equilibrium was shifted away
Sex Steroids from resorption due to a 30-fold increase in
osteoprotegerin. This result indicates that
Estrogen deciency has long been known to FGF-2 enhances the inhibitory effect of PTH on
cause a resorptive osteoporosis. While estrogen OPG gene expression. FGF-2 is required in PTH-
may modulate the action of T-lymphocytes and induced hypercalcemia, as evidenced by the
T-lymphocyte expression on RANKL [140, 191], resistance of the FGF-2 null animal to PTH-
it also dampens the RANKL-stimulated signal induced hypercalcemia [134]. Whether this
cascade. It does so by decreasing c-Jun ex- effect is entirely due to FGF-2 mediated bone
pression and its phosphorylation by c-Jun N- remodeling, or whether FGF-2 also promotes
terminal kinase [156]. Androgen deciency also the PTH induced increase in renal calcium
results in bone resorption and osteoporosis excretion, is not known.
through the loss of androgen dampening of
RANK activation of c-Jun [63].
Gonadal steroids also affect osteoclast Insulin-like Growth Factor-I
recruitment by regulating the expression of Insulin-like growth factor-I (IGF-I) is secreted
osteoprotegerin. In ovariectomized rats, both during puberty, inducing long bone growth
RANKL and OPG are upregulated. This sug- [200]. Because of its anabolic effects on bone,
gests that estrogen deciency stimulates the increasing bone density [201], it was thought
entire RANKL system [66]. In men, serum OPG that IGF-I should have salutary effects in osteo-
increases with age, while resorptive markers porosis. Surprisingly, in clinical trials, IGF-I
decrease [166]. In men made acutely hypogo- caused an elevation of bone markers without
nadal, serum OPG increases. This suggests a signicant increases in bone density [39]. This
response to an upregulated resorptive system may be the result of IGF-I action on osteo-
[79]. Alternatively, estrogen appears to upregu- clastogenesis. Addition of IGF-I to osteoclast-
late OPG expression by bone cells [58, 150]. generating systems increased the numbers of
These and other studies suggest that OPG is osteoclasts, as well as the pit area per osteoclast
modied with respect to the bone remodeling [123]. In culture IGF-I increased RANKL
rate [78]. expression and decreased OPG [144]. It is likely
that the pleiotropic effects of IGF-I in bone
allow for bone remodeling, rather than pure
Parathyroid Hormone and Vitamin D anabolism of bone.
Vitamin D is a potent stimulator of osteo-
clastogenesis and bone resorption. It acts by in-
creasing MCSF [145] and RANKL [206] levels
Calcitonin
locally and decreasing OPG [61]. Parathyroid Calcitonin, a 32-amino-acid peptide secreted
hormone, when administered continuously at from the C-cells of the thyroid, has been used
high concentrations, has effects similar to those in hypercalcemia of malignancy to curtail
of vitamin D in terms of MCSF [190] and osteoclastic bone resorption. Osteoclasts ex-
RANKL:OPG [61, 105], leading to increases in press receptors for calcitonin during early dif-
bone resorption. While both PTH and vitamin ferentiation and maintain these throughout
D alter cytokine levels, such as that of IL-6 [46, their life [103]. Thus calcitonin decreases the
47], it is their effect on the RANKL:OPG equi- activity of mature osteoclasts, but also inhibits
librium that is the major factor by means of early differentiation events [168].
which they control osteoclast formation.
The expression of broblast growth factor 2,
which potently induces osteoclast formation, is
Prostaglandins
stimulated by PTH and may serve as a sec- Prostaglandins may have biphasic effects on
ondary mechanism to increase bone resorption. bone remodeling, dependent on the local
In bone marrow cultures made from the FGF-2 microenvironment. Addition of PGE2 to murine
null mouse, PTH was less effective in stimulat- osteoclast generating systems in vitro stimulates
ing osteoclast formation [134]. Interestingly, osteoclast formation [5] and increases RANKL
PTH stimulated RANKL to the same degree in expression [206]. PGE2 does, however, inhibit
Osteoclast: Origin and Differentiation 15

osteoclast formation in cultures of human cells The osteoclast cDNA expression library [169]
[21]. Many of prostaglandins variable effects from which AXII was initially isolated also
may be due to differential or temporal expres- yielded an inhibitor of osteoclast formation
sion of the many prostaglandin receptor types. found to be identical to human legumain, a cys-
teine endopeptidase [23]. Legumain can be
detected in human marrow plasma from normal
Secreted Factors other donors. When human marrow cultures are
than Cytokines treated with an antibody to legumain, osteoclast
formation is enhanced in the absence of 1,25-
Nitric Oxide and other Reactive dihydroxyvitamin D.
Oxygen Species
Nitric oxide (NO) has effects in nearly every Cytokines
tissue in the body, including bone. One of the
earliest reports linking NO to bone remodeling A host of cytokines, including tumor necrosis
showed that inhibition of NOS, the enzyme factor and interleukins-1, -4, -6, -11, -18, have
that synthesizes NO from L-arginine, potenti- both positive and negative effects on osteoclas-
ates bone resorption in ovariectomized rats togenesis. These are dealt with in chapter 5.
[77]. Since then, studies have shown that NO can
slow bone loss in animals and humans [70, 194,
195]. NO alters osteoclast indices in vitro: osteo- Osteoclast-Associated Receptor Ligand
clastic resorption is potentiated during NOS Other factors produced by osteoblasts and
inhibition [77]. This could result from effects on stromal cells have been identied through
osteoclast activity, because NO induces a shape nding binding partners of discrete osteoclast
change in the osteoclasts associated with a receptors. One of the most interesting is the
reduction in bone resorption. Nitric oxide also osteoclast-associated receptor, or OSCAR,
decreases osteoclast recruitment and subse- representing a novel member of the leukocyte
quent bone resorption [107]. The effect of NO receptor complex [80]. OSCAR expression
on osteoclast recruitment is likely through varies from the usual immunoglobulin-like
alteration in the RANKL/OPG equilibrium as surface receptors typical of leukocytes by being
NO has been shown to both decrease RANKL expressed only in pre-osteoclasts and mature
and increase OPG expression in bone stromal osteoclasts. The ligand for OSCAR (OSCAR-L) is
cells [36]. expressed in osteoprogenitor cells. This is a
Other reactive oxygen species induced by developing area of research that may help to link
stress or as byproducts of nitric oxide are active the immune system genes more closely to bone
in cells as well. Hydrogen peroxide has been remodeling.
shown to increase osteoclast differentiation,
either directly or in combination with 1,25-
dihydroxyvitamin D [161]. Mechanical Factors
A major function of the skeletal structure is to
Factors Secreted by Osteoclasts: provide support for locomotion. To adapt to
functional loading, the skeleton has evolved a
Annexin II and Legumain nely tuned response system that recognizes
Annexin II was identied as a candidate gene in mechanical input. Unloading leads to bone
the stimulation of osteoclast formation from a resorption and loading to bone apposition and
mammalian osteoclast cDNA expression library preservation. The cells of bone must thus be
[169]. Puried recombinant AXII induced able to respond to unloading by initiating bone
osteoclast formation in the absence of 1,25- resorption. The results of bone resorption are
dihydroxyvitamin D, probably acting on the seen in the skeletons of paraplegics and of
proliferative stage of osteoclast precursors, and astronauts, and quite readily in situations
enhancing the growth of the CFU-GM [119]. where unloading was generated experimentally
Annexin II appears to stimulate the secretion of [142, 143]. Thus, the recruitment of additional
GM-CSF from T-cells and thus may have a role osteoclasts involved in unloading-associated
in inammatory bone loss. resorption probably occurs via changes in the
16 Bone Resorption

dominant RANKL:OPG equilibrium. Experi- 9. Armstrong A, Tometsko M, Glaccum M, Sutherland C,


mental studies with cells subject to loading Cosman D, Dougall W (2002) A RANK/TRAF6-
dependent signal transduction pathway is essential for
suggest that loading stromal cells decreases osteoclast cytoskeletal organization and resorptive
their expression of RANKL through a pathway function. J Biol Chem 277:4434756.
that involves activation of MAP-kinases [146, 10. Athanasou NA, Quinn J (1990) Immunophenotypic
147]. differences between osteoclasts and macrophage poly-
karyons: immunohistological distinction and implica-
tions for osteoclast ontogeny and function. J Clin
Pathol 43:9971003.
Conclusion 11. Azuma Y, Kaji K, Katogi R, Takeshita S, Kudo A (2000)
Tumor necrosis factor-alpha induces differentiation of
and bone resorption by osteoclasts. J Biol Chem
Resorption of the calcied surface of bone 275:485864.
involves the orchestrated action of a large 12. Biskobing DM, Fan D, Fan X, Rubin J (1997) Induction
variety of enzymes and disposal mechanisms. of carbonic anhydrase II expression in osteoclast prog-
enitors requires physical contact with stromal cells.
Interference with, or overactivity of, any of these Endocrinology 138:48527.
processes, leads to dysregulation of bone 13. Biskobing DM, Fan X, Rubin J (1995) Characterization
remodeling. Thus regulation of both the forma- of MCSF-induced proliferation and subsequent osteo-
tion and action of the osteoclast modulates clast formation in murine marrow culture. J Bone
bone development, bone growth, and bone Miner Res 10:102532.
14. Boissy P, Saltel F, Bouniol C, Jurdic P, Machuca-Gayet I
disease. (2002) Transcriptional activity of nuclei in multinucle-
ated osteoclasts and its modulation by calcitonin.
Endocrinology 143:191321.
References 15. Boyce BF, Yoneda T, Lowe C, Soriano P, Mundy GR
(1992) Requirement of pp60c-src expression for osteo-
clasts to form rufed borders and resorb bone in mice.
1. Abe E, Miyaura C, Tanaka H, Shiina Y, Kuribayashi T, J Clin Invest 90:16227.
Suda S, et al. (1983) 1 alpha,25-dihydroxyvitamin D3 16. Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J,
promotes fusion of mouse alveolar macrophages both Capparelli C, et al. (1998) osteoprotegerin-decient
by a direct mechanism and by a spleen cell-mediated mice develop early onset osteoporosis and arterial cal-
indirect mechanism. Proc Natl Acad Sci USA 80: cication. Genes Dev 12:12608.
55837. 17. Burger EH, Van der Meer JW, van de Gevel JS, Gribnau
2. Abe E, Mocharla H, Yamate T, Taguchi Y, Manolagas SC JC, Thesingh GW, van Furth R (1982) In vitro forma-
(1999) Meltrin-alpha, a fusion protein involved in tion of osteoclasts from long-term cultures of bone
multinucleated giant cell and osteoclast formation. marrow mononuclear phagocytes. J Exp Med 156:
Calcif Tissue Int 64:50815. 160414.
3. Abu-Amer Y, Erdmann J, Alexopoulou L, Kollias G, 18. Burguera B, Hofbauer LC, Thomas T, Gori F, Evans GL,
Ross FP, Teitelbaum SL (2000) Tumor necrosis factor Khosla S, et al. (2001) Leptin reduces ovariectomy-
receptors types 1 and 2 differentially regulate osteo- induced bone loss in rats. Endocrinology 142:3546
clastogenesis. J Biol Chem 275:2730710. 53.
4. Akashi K, Traver D, Miyamoto T, Weissman IL (2000) 19. Cenci S, Weitzmann MN, Roggia C, Namba N, Novack
A clonogenic common myeloid progenitor that gives D, Woodring J, et al. (2000) Estrogen deciency induces
rise to all myeloid lineages. Nature 404:1937. bone loss by enhancing T-cell production of TNF-
5. Akatsu T, Takahashi N, Debari K, Morita I, Murota S, alpha. J Clin Invest 106:122937.
Nagata N, et al. (1989) Prostaglandins promote osteo- 20. Chambers TJ, Owens JM, Hattersley G, Jat PS, Noble
clastlike cell formation by a mechanism involving MD (1993) Generation of osteoclast-inductive and
cyclic adenosine 3,5-monophosphate in mouse bone osteoclastogenic cell lines from the H-2KbtsA58 trans-
marrow cell cultures. J Bone Miner Res 4:2935. genic mouse. Proc Natl Acad Sci USA 90:557882.
6. Akatsu T, Takahashi N, Udagawa N, Sato K, Nagata N, 21. Chenu C, Kurihara N, Mundy GR, Roodman GD (1990)
Moseley JM, et al. (1989) Parathyroid hormone (PTH)- Prostaglandin E2 inhibits formation of osteoclastlike
related protein is a potent stimulator of osteoclast-like cells in long-term human marrow cultures but is not a
multinucleated cell formation to the same extent as mediator of the inhibitory effects of transforming
PTH in mouse marrow cultures. Endocrinology 125: growth factor beta. J Bone Miner Res 5:67781.
207. 22. Choi SJ, Han JH, Roodman GD (2001) ADAM8: a novel
7. Anderson DM, Maraskovsky E, Billingsley WL, Dougall osteoclast stimulating factor. J Bone Miner Res 16:
WC, Tometsko ME, Roux ER, et al. (1997) A homologue 81422.
of the TNF receptor and its ligand enhance T-cell 23. Choi SJ, Reddy SV, Devlin RD, Menaa C, Chung H,
growth and dendritic-cell function. Nature 390:1759. Boyce BF, et al. (1999) Identication of human
8. Arai F, Miyamoto T, Ohneda O, Inada T, Sudo T, Brasel asparaginyl endopeptidase (legumain) as an inhibitor
K, et al. (1999) Commitment and differentiation of of osteoclast formation and bone resorption. J Biol
osteoclast precursor cells by the sequential expression Chem 274:2774753.
of c-Fms and receptor activator of nuclear factor 24. Coccia PF, Krivit W, Cervenka J, Clawson C, Kersey JH,
kappaB (RANK) receptors. J Exp Med 190:174154. Kim TH, et al. (1980) Successful bone-marrow trans-
Osteoclast: Origin and Differentiation 17

plantation for infantile malignant osteopetrosis. N 40. Fuller K, Bayley KE, Chambers TJ (2000) Activin A is
Engl J Med 302:7018. an essential cofactor for osteoclast induction. Biochem
25. Collin-Osdoby P, Rothe L, Anderson F, Nelson M, Biophys Res Commun 268:27.
Maloney W, Osdoby P (2001) RANKL and OPG expres- 41. Fuller K, Lean JM, Bayley KE, Wani MR, Chambers TJ
sion by human microvascular endothelial cells, regula- (2000) A role for TGFbeta(1) in osteoclast differentia-
tion by inammatory cytokines, and role in human tion and survival. J Cell Sci 113 (Pt 13):244553.
osteoclastogenesis. J Biol Chem 23:23. 42. Gao YH, Shinki T, Yuasa T, Kataoka-Enomoto H,
26. Darnay B, Ni J, Moore P, Aggarwal B (1999) Activation Komori T, Suda T, et al. (1998) Potential role of cbfa1,
of NF-kappaB by RANK requires tumor necrosis factor an essential transcriptional factor for osteoblast
receptor-associated factor (TRAF) 6 and NF-kappaB- differentiation, in osteoclastogenesis: regulation of
inducing kinase. Identication of a novel TRAF6 inter- mRNA expression of osteoclast differentiation factor
action motif. J Biol Chem 274:772431. (ODF). Biochem Biophys Res Commun 252:697702.
27. DeFife KM, Jenney CR, McNally AK, Colton E, 43. Gelb BD, Shi GP, Chapman HA, Desnick RJ (1996) Pyc-
Anderson JM (1997) Interleukin-13 induces human nodysostosis, a lysosomal disease caused by cathepsin
monocyte/macrophage fusion and macrophage K deciency. Science 273:12368.
mannose receptor expression. J Immunol 158:338590. 44. Gori F, Hofbauer LC, Dunstan CR, Spelsberg TC, Khosla
28. Dexter TM (1979) Haemopoiesis in long-term bone S, Riggs BL (2000) The expression of osteoprotegerin
marrow cultures. A review. Acta Haematol 62:299 and RANK ligand and the support of osteoclast for-
305. mation by stromal-osteoblast lineage cells is develop-
29. Dexter TM, Allen TD, Lajtha LG, Schoeld R, Lord BI mentally regulated. Endocrinology 141:476876.
(1973) Stimulation of differentiation and proliferation 45. Graves L, 3rd, Jilka RL (1990) Comparison of bone and
of haemopoietic cells in vitro. J Cell Physiol 82:46173. parathyroid hormone as stimulators of osteoclast
30. Emery JG, McDonnell P, Burke MB, Deen KC, Lyn S, development and activity in calvarial cell cultures from
Silverman C, et al. (1998) Osteoprotegerin is a normal and osteopetrotic (mi/mi) mice. J Cell Physiol
receptor for the cytotoxic ligand TRAIL. J Biol Chem 145:1029.
273:143637. 46. Greeneld EM, Horowitz MC, Lavish SA (1996) Stimu-
31. Fallon MD, Teitelbaum SL, Kahn AJ (1983) Multinucle- lation by parathyroid hormone of interleukin-6 and
ation enhances macrophage-mediated bone resorp- leukemia inhibitory factor expression in osteoblasts is
tion. Lab Invest 49:15964. an immediate-early gene response induced by cAMP
32. Falzoni S, Munerati M, Ferrari D, Spisani S, Moretti S, signal transduction. J Biol Chem 271:109849.
Di Virgilio F (1995) The purinergic P2Z receptor of 47. Greeneld EM, Shaw SM, Gornik SA, Banks MA (1995)
human macrophage cells. Characterization and pos- Adenyl cyclase and interleukin 6 are downstream
sible physiological role. J Clin Invest 95:120716. effectors of parathyroid hormone resulting in stimula-
33. Fan X, Biskobing DM, Bain S, Rubin J (1996) Keto- tion of bone resorption. J Clin Invest 96:123844.
conazole and phorbol myristate acetate regulate osteo- 48. Grigoriadis AE, Wang ZQ, Cecchini MG, Hofstetter W,
clast precursor fusion in primary murine marrow Felix R, Fleisch HA, et al. (1994) c-Fos: a key regulator
culture. J Bone Miner Res 11:127480. of osteoclast-macrophage lineage determination and
34. Fan X, Biskobing DM, Fan D, Hofstetter W, Rubin J bone remodeling. Science 266:4438.
(1997) Macrophage colony stimulating factor down- 49. Han JH, Choi SJ, Kurihara N, Koide M, Oba Y, Roodman
regulates MCSF-receptor expression and entry of GD (2001) Macrophage inammatory protein-1alpha
progenitors into the osteoclast lineage. J Bone Miner is an osteoclastogenic factor in myeloma that is inde-
Res 12:138795. pendent of receptor activator of nuclear factor kappaB
35. Fan X, Fan D, Gewant H, Royce C, Nanes M, Rubin ligand. Blood 97:334953.
J (2001) Increasing membrane-bound MCSF does 50. Han X, Sterling H, Chen Y, Saginario C, Brown EJ, Frazier
not enhance OPGL-driven osteoclastogenesis from WA, et al. (2000) CD47, a ligand for the macrophage
marrow cells. Am J Physiol Endocrinol Metab 280: fusion receptor, participates in macrophage multinucle-
E10311. ation. J Biol Chem 275:3798492.
36. Fan X, Zhu L, Roy E, Murphy TC, Hart CM, Rosen C, 51. Hattersley G, Chambers TJ (1989) Calcitonin receptors
Nanes MS, Rubin J (2004) Nitric oxide decreases as markers for osteoclastic differentiation: correlation
RANKL and increases OPG expression in bone stromal between generation of bone-resorptive cells and cells
cells Endocrinology 145:751759. that express calcitonin receptors in mouse bone
37. Fisher JE, Rogers MJ, Halasy JM, Luckman SP, Hughes marrow cultures. Endocrinology 125:160612.
DE, Masarachia PJ, et al. (1999) Alendronate mecha- 52. Hattersley G, Kerby JA, Chambers TJ (1991) Identica-
nism of action: geranylgeraniol, an intermediate in the tion of osteoclast precursors in multilineage hemopoi-
mevalonate pathway, prevents inhibition of osteoclast etic colonies. Endocrinology 128:25962.
formation, bone resorption, and kinase activation in 53. Hattersley G, Owens J, Flanagan AM, Chambers TJ
vitro. Proc Natl Acad Sci USA 96:1338. (1991) Macrophage colony stimulating factor (M-CSF)
38. Fleischmann A, Hafezi F, Elliott C, Reme CE, Ruther U, is essential for osteoclast formation in vitro. Biochem
Wagner EF (2000) Fra-1 replaces c-Fos-dependent Biophys Res Commun 177:52631.
functions in mice. Genes Dev 14:2695700. 54. Helfrich MH, Mieremet RH (1988) A morphologic
39. Friedlander AL, Buttereld GE, Moynihan S, Grillo J, study of osteoclasts isolated from osteopetrotic micro-
Pollack M, Holloway L, et al. (2001) One year of insulin- phthalmic (mi/mi) mouse and human fetal long bones
like growth factor I treatment does not affect bone using an instrument permitting combination of light
density, body composition, or psychological measures and scanning electron microscopy. Bone 9:11319.
in postmenopausal women. J Clin Endocrinol Metab 55. Hodgkinson CA, Moore KJ, Nakayama A, Steingrims-
86:1496503. son E, Copeland NG, Jenkins NA, et al. (1993) Muta-
18 Bone Resorption

tions at the mouse microphthalmia locus are associ- 70. Jamal SA, Browner WS, Bauer DC, Cummings SR
ated with defects in a gene encoding a novel basic- (1998) Intermittent use of nitrates increases bone
helix-loop-helix-zipper protein. Cell 74:395404. mineral density: the study of osteoporotic fractures.
56. Hofbauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, J Bone Miner Res 13:17559.
Spelsberg TC, et al. (1999) Stimulation of osteoprote- 71. Jamieson JC, Wayne S, Belo RS, Wright JA, Spearman
gerin ligand and inhibition of osteoprotegerin pro- MA (1992) The importance of N-linked glycoproteins
duction by glucocorticoids in human osteoblastic and dolichyl phosphate synthesis for fusion of L6
lineage cells: potential paracrine mechanisms of glu- myoblasts. Biochem Cell Biol 70:40812.
cocorticoid-induced. Endocrinology 140:43829. 72. Jimi E, Akiyama S, Tsurukai T, Okahashi N, Kobayashi
57. Hofbauer LC, Heufelder AE (2001) Role of receptor K, Udagawa N, et al. (1999) Osteoclast differentiation
activator of nuclear factor-kappaB ligand and osteo- factor acts as a multifunctional regulator in murine
protegerin in bone cell biology. J Mol Med 79:24353. osteoclast differentiation and function. J Immunol
58. Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spels- 163:43442.
berg TC, Riggs BL (1999) Estrogen stimulates gene 73. Jochum W, David JP, Elliott C, Wutz A, Plenk H, Jr.,
expression and protein production of osteoprotegerin Matsuo K, et al. (2000) Increased bone formation and
in human osteoblastic cells. Endocrinology 140: osteosclerosis in mice overexpressing the transcription
436770. factor Fra-1. Nat Med 6:9804.
59. Horton MA, Pringle JA, Chambers TJ (1985) Identi- 74. Junttila I, Bourette RP, Rohrschneider LR, Silvennoinen
cation of human osteoclasts with monoclonal anti- O (2003) M-CSF induced differentiation of myeloid
bodies. N Engl J Med 312:9234. precursor cells involves activation of PKC-delta and
60. Horton MA, Rimmer EF, Lewis D, Pringle JA, Fuller K, expression of Pkare. J Leukoc Biol 73:2818.
Chambers TJ (1984) Cell surface characterization of 75. Kahn AJ, Simmons DJ (1975) Investigation of cell
the human osteoclast: phenotypic relationship to other lineage in bone using a chimaera of chick and quial
bone marrow-derived cell types. J Pathol 144:28194. embryonic tissue. Nature 258:3257.
61. Horwood N, Elliott J, Martin T, Gillespie M (1998) 76. Kanematsu M, Yoshimura K, Takaoki M, Sato A (2002)
Osteotropic agents regulate the expression of osteo- Vector-averaged gravity regulates gene expression
clast differentiation factor and osteoprotegerin in of receptor activator of NF-kappaB (RANK) ligand
osteoblastic stromal cells. Endocrinology 47436. and osteoprotegerin in bone marrow stromal cells via
62. Hsu H, Lacey DL, Dunstan CR, Solovyev I, Colombero cyclic AMP/protein kinase A pathway. Bone 30:5538.
A, Timms E, et al. (1999) Tumor necrosis factor recep- 77. Kasten TP, Collin-Osdoby P, Patel N, Osdoby P,
tor family member RANK mediates osteoclast differ- Krukowski M, Misko TP, et al. (1994) Potentiation of
entiation and activation induced by osteoprotegerin osteoclast bone-resorption activity by inhibition of
ligand. Proc Natl Acad Sci USA 96:35405. nitric oxide synthase. Proc Natl Acad Sci USA 91:
63. Huber DM, Bendixen AC, Pathrose P, Srivastava S, 356973.
Dienger KM, Shevde NK, et al. (2001) Androgens sup- 78. Khosla S, Arrighi HM, Melton LJ, 3rd, Atkinson EJ,
press osteoclast formation induced by RANKL and OFallon WM, Dunstan C, et al. (2002) Correlates of
macrophage-colony stimulating factor. Endocrinology osteoprotegerin levels in women and men. Osteoporos
142:38008. Int 13:3949.
64. Hughes AE, Ralston SH, Marken J, Bell C, MacPherson 79. Khosla S, Atkinson EJ, Dunstan CR, OFallon WM
H, Wallace RG, et al. (2000) Mutations in TNFRSF11A, (2002) Effect of estrogen versus testosterone on
affecting the signal peptide of RANK, cause familial circulating osteoprotegerin and other cytokine levels
expansile osteolysis. Nat Genet 24:458. in normal elderly men. J Clin Endocrinol Metab 87:
65. Ibbotson KJ, Roodman GD, McManus LM, Mundy GR 15504.
(1984) Identication and characterization of osteo- 80. Kim N, Takami M, Rho J, Josien R, Choi Y (2002) A
clast-like cells and their progenitors in cultures of novel member of the leukocyte receptor complex reg-
feline marrow mononuclear cells. J Cell Biol 99:47180. ulates osteoclast differentiation. J Exp Med 195:2019.
66. Ikeda T, Utsuyama M, Hirokawa K (2001) Expression 81. Kimble RB, Srivastava S, Ross FP, Matayoshi A, Pacici
proles of receptor activator of nuclear factor kappaB R (1996) Estrogen deciency increases the ability of
ligand, receptor activator of nuclear factor kappaB, and stromal cells to support murine osteoclastogenesis via
osteoprotegerin messenger RNA in aged and ovariec- an interleukin-1and tumor necrosis factor-mediated
tomized rat bones. J Bone Miner Res 16:141625. stimulation of macrophage colony-stimulating factor
67. Ishida N, Hayashi K, Hoshijima M, Ogawa T, Koga S, production. J Biol Chem 271:288907.
Miyatake Y, et al. (2002) Large scale gene expression 82. Kitazawa R, Kitazawa S, Maeda S (1999) Promoter
analysis of osteoclastogenesis in vitro and elucidation structure of mouse RANKL/TRANCE/OPGL/ODF
of NFAT2 as a key regulator. J Biol Chem 277:4114756. gene. Biochim Biophys Acta 1445:13441.
68. Itoh K, Udagawa N, Katagiri T, Iemura S, Ueno N, 83. Kobayashi K, Takahashi N, Jimi E, Udagawa N, Takami
Yasuda H, et al. (2001) Bone morphogenetic protein 2 M, Kotake S, et al. (2000) Tumor necrosis factor alpha
stimulates osteoclast differentiation and survival sup- stimulates osteoclast differentiation by a mechanism
ported by receptor activator of nuclear factor-kappaB independent of the ODF/RANKL-RANK interaction.
ligand. Endocrinology 142:365662. J Exp Med 191:27586.
69. Itoh K, Udagawa N, Matsuzaki K, Takami M, Amano H, 84. Kobayashi N, Kadono Y, Naito A, Matsumoto K,
Shinki T, et al. (2000) Importance of membrane- or Yamamoto T, Tanaka S, et al. (2001) Segregation of
matrix-associated forms of M-CSF and RANKL/ODF TRAF6-mediated signaling pathways claries its role
in osteoclastogenesis supported by SaOS-4/3 cells in osteoclastogenesis. EMBO J 20:127180.
expressing recombinant PTH/PTHrP receptors. J Bone 85. Kodaira K, Mizuno A, Yasuda H, Shima N, Murakami
Miner Res 15:176675. A, Ueda M, et al. (1999) Cloning and characterization
Osteoclast: Origin and Differentiation 19

of the gene encoding mouse osteoclast differentiation ment of bone marrow precursors through expression
factor. Gene 230:1217. of membrane-bound TRANCE. Bone 27:2940.
86. Kodama H, Nose M, Niida S, Yamasaki A (1991) Essen- 102. Lee MY, Lottsfeldt JL, Fevold KL (1992) Identication
tial role of macrophage colony-stimulating factor in and characterization of osteoclast progenitors by
the osteoclast differentiation supported by stromal clonal analysis of hematopoietic cells. Blood 80:
cells. J Exp Med 173:12914. 171016.
87. Kolliker A. (1873) Die normale Resorption des 103. Lee SK, Goldring SR, Lorenzo JA (1995) Expression of
Knochengewebes und ihre Bedeutung fr die Entste- the calcitonin receptor in bone marrow cell cultures
hung der typischen Knochenformen, edn. In: Vogel F, and in bone: a specic marker of the differentiated
editor Leipzig. osteoclast that is regulated by calcitonin. Endocrinol-
88. Kondo M, Weissman IL, Akashi K (1997) Identication ogy 136:457281.
of clonogenic common lymphoid progenitors in 104. Lee SK, Lorenzo JA (1999) Parathyroid hormone stim-
mouse bone marrow. Cell 91:66172. ulates TRANCE and inhibits osteoprotegerin messen-
89. Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony ger ribonucleic acid expression in murine bone
S, et al. (1999) Activated T cells regulate bone loss and marrow cultures: correlation with osteoclast-like cell
joint destruction in adjuvant arthritis through osteo- formation. Endocrinology 140:355261.
protegerin ligand. Nature 402:3049. 105. Lee SK, Lorenzo JA (2002) Regulation of receptor acti-
90. Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, vator of nuclear factor-kappa B ligand and osteoprote-
Capparelli C, et al. (1999) OPGL is a key regulator gerin mRNA expression by parathyroid hormone is
of osteoclastogenesis, lymphocyte development and predominantly mediated by the protein kinase a
lymph-node organogenesis. Nature 397:31523. pathway in murine bone marrow cultures. Bone 31:
91. Kukita A, Chenu C, McManus LM, Mundy GR, 2529.
Roodman GD (1990) Atypical multinucleated cells 106. Lomaga MA, Yeh WC, Sarosi I, Duncan GS, Furlonger
form in long-term marrow cultures from patients with C, Ho A, et al. (1999) TRAF6 deciency results in
Pagets disease. J Clin Invest 85:12806. osteopetrosis and defective interleukin-1, CD40, and
92. Kurachi T, Morita I, Oki T, Ueki T, Sakaguchi K, LPS signaling. Genes Dev 13:101524.
Enomoto S, et al. (1994) Expression on outer mem- 107. Lowik C, Nibberin PH, van de Ruit M, Papapoulos SE
branes of mannose residues, which are involved in (1994) Inducible produciton of nitric oxide in
osteoclast formation via cellular fusion events. J Biol osteoblast-like cells and in fetal mouse bone explants
Chem 269:175726. is associated with suppression of osteoclastic bone
93. Kurihara N, Chenu C, Miller M, Civin C, Roodman GD resorption. J Clin Invest 93:146572.
(1990) Identication of committed mononuclear pre- 108. Luchin A, Purdom G, Murphy K, Clark MY, Angel N,
cursors for osteoclast-like cells formed in long term Cassady AI, et al. (2000) The microphthalmia tran-
human marrow cultures. Endocrinology 126:273341. scription factor regulates expression of the tartrate-
94. Kurihara N, Gluck S, Roodman GD (1990) Sequential resistant acid phosphatase gene during terminal
expression of phenotype markers for osteoclasts differentiation of osteoclasts. J Bone Miner Res 15:
during differentiation of precursors for multinucleated 45160.
cells formed in long-term human marrow cultures. 109. Luchin A, Suchting S, Merson T, Rosol TJ, Hume DA,
Endocrinology 127:321521. Cassady AI, et al. (2001) Genetic and physical interac-
95. Kurihara N, Suda T, Miura Y, Nakauchi H, Kodama H, tions between Microphthalmia transcription factor
Hiura K, et al. (1989) Generation of osteoclasts from and PU.1 are necessary for osteoclast gene expression
isolated hematopoietic progenitor cells. Blood 74: and differentiation. J Biol Chem 276:3670310.
1295302. 110. Lum L, Wong BR, Josien R, Becherer JD, Erdjument-
96. Lacey DL, Tan HL, Lu J, Kaufman S, Van G, Qiu W, Bromage H, Schlondorff J, et al. (1999) Evidence for a
et al. (2000) Osteoprotegerin ligand modulates murine role of a tumor necrosis factor-alpha (TNF-alpha)-
osteoclast survival in vitro and in vivo. Am J Pathol converting enzyme-like protease in shedding of
157:43548. TRANCE, a TNF family member involved in osteo-
97. Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, clastogenesis and dendritic cell survival. J Biol Chem
Burgess T, et al. (1998) Osteoprotegerin ligand is a 274:136138.
cytokine that regulates osteoclast differentiation and 111. Ma YL, Cain RL, Halladay DL, Yang X, Zeng Q, Miles
activation. Cell 93:16576. RR, et al. (2001) Catabolic effects of continuous human
98. Lam J, Nelson CA, Ross FP, Teitelbaum SL, Fremont DH pth (138) in vivo is associated with sustained stimu-
(2001) Crystal structure of the TRANCE/RANKL lation of rankl and inhibition of osteoprotegerin and
cytokine reveals determinants of receptor-ligand gene-associated bone formation. Endocrinology 142:
specicity. J Clin Invest 108:9719. 404754.
99. Lam J, Takeshita S, Barker JE, Kanagawa O, Ross FP, 112. MacDonald BR, Mundy GR, Clark S, Wang EA, Kuehl
Teitelbaum SL (2000) TNF-alpha induces osteoclasto- TJ, Stanley ER, et al. (1986) Effects of human
genesis by direct stimulation of macrophages exposed recombinant CSF-GM and highly puried CSF-1 on the
to permissive levels of RANK ligand. J Clin Invest formation of multinucleated cells with osteoclast
106:14818. characteristics in long-term bone marrow cultures. J
100. Lean JM, Fuller K, Chambers TJ (2001) FLT3 ligand can Bone Miner Res 1:22733.
substitute for macrophage colony-stimulating factor in 113. Mansky K, Sankar U, Han J, Ostrowski M (2002)
support of osteoclast differentiation and function. Microphthalmia transcription factor is a target of the
Blood 98:270713. p38 MAPK pathway in response to receptor activator
101. Lean JM, Matsuo K, Fox SW, Fuller K, Gibson FM, of NF-kappa B ligand signaling. J Biol Chem 277:
Draycott G, et al. (2000) Osteoclast lineage commit- 1107783.
20 Bone Resorption

114. Massey HM, Scopes J, Horton MA, Flanagan AM (2001) NF-kappaB and putative signal transducer for the lym-
Transforming growth factor-beta1 (TGF-beta) stimu- photoxin-beta receptor. J Biol Chem 271:146614.
lates the osteoclast-forming potential of peripheral 129. Nakashima T, Kobayashi Y, Yamasaki S, Kawakami A,
blood hematopoietic precursors in a lymphocyte-rich Eguchi K, Sasaki H, et al. (2000) Protein Expression
microenvironment. Bone 28:57782. and Functional Difference of Membrane-Bound and
115. Matsuo K, Owens JM, Tonko M, Elliott C, Chambers TJ, Soluble Receptor Activator of NF-kappaB Ligand:
Wagner EF (2000) Fosl1 is a transcriptional target Modulation of the Expression by Osteotropic Factors
of c-Fos during osteoclast differentiation. Nat Genet and Cytokines. Biochem Biophys Res Commun 275:
24:1847. 768775.
116. Mbalaviele G, Chen H, Boyce BF, Mundy GR, Yoneda T 130. Nakashima T, Kobayashi Y, Yamasaki S, Kawakami A,
(1995) The role of cadherin in the generation of mult- Eguchi K, Sasaki H, et al. (2000) Protein expression and
inucleated osteoclasts from mononuclear precursors functional difference of membrane-bound and soluble
in murine marrow. J Clin Invest 95:275765. receptor activator of NF-kappaB ligand: modulation of
117. McCauley LK, Koh AJ, Beecher CA, Rosol TJ (1997) the expression by osteotropic factors and cytokines.
Proto-oncogene c-fos is transcriptionally regulated by Biochem Biophys Res Commun 275:76875.
parathyroid hormone (PTH) and PTH-related protein 131. Niida S, Kaku M, Amano H, Yoshida H, Kataoka
in a cyclic adenosine monophosphate-dependent H, Nishikawa S, et al. (1999) Vascular endothelial
manner in osteoblastic cells. Endocrinology 138:5427 growth factor can substitute for macrophage colony-
33. stimulating factor in the support of osteoclastic bone
118. McHugh KP, Hodivala-Dilke K, Zheng MH, Namba N, resorption. J Exp Med 190:2938.
Lam J, Novack D, et al. (2000) Mice lacking beta3 132. Nutt SL, Heavey B, Rolink AG, Busslinger M (1999)
integrins are osteosclerotic because of dysfunctional Commitment to the B-lymphoid lineage depends on
osteoclasts. J Clin Invest 105:43340. the transcription factor Pax5. Nature 401:55662.
119. Menaa C, Devlin RD, Reddy SV, Gazitt Y, Choi SJ, 133. OBrien CA, Kern B, Gubrij I, Karsenty G, Manolagas
Roodman GD (1999) Annexin II increases osteoclast SC (2002) Cbfa1 does not regulate RANKL gene activ-
formation by stimulating the proliferation of osteo- ity in stromal/osteoblastic cells. Bone 30:45362.
clast precursors in human marrow cultures. J Clin 134. Okada Y, Montero A, Zhang X, Sobue T, Lorenzo J,
Invest 103:160513. Doetschman T, et al. (2003) Impaired osteoclast for-
120. Menaa C, Reddy SV, Kurihara N, Maeda H, Anderson mation in bone marrow cultures of Fgf2 null mice in
D, Cundy T, et al. (2000) Enhanced RANK ligand response to parathyroid hormone. J Biol Chem 278:
expression and responsivity of bone marrow cells in 2125866.
Pagets disease of bone. J Clin Invest 105:18338. 135. Osawa M, Hanada K, Hamada H, Nakauchi H (1996)
121. Min H, Morony S, Sarosi I, Dunstan CR, Capparelli C, Long-term lymphohematopoietic reconstitution by a
Scully S, et al. (2000) Osteoprotegerin reverses osteo- single CD34-low/negative hematopoietic stem cell.
porosis by inhibiting endosteal osteoclasts and pre- Science 273:2425.
vents vascular calcication by blocking a process 136. Perkins S, Kling S (1995) Local concentrations of MCSF
resembling osteoclastogenesis. J Exp Med 192:46374. mediate osteoclastic differentiation. Amer J Physiol
122. Miyauchi A, Kuroki Y, Fukase M, Fujita T, Chihara K, 269:E102430.
Shiozawa S (1994) Persistent expression of proto- 137. Povolny BT, Lee MY (1993) The role of recombinant
oncogene c-fos stimulates osteoclast differentiation. human M-CSF, IL-3, GM-CSF and calcitriol in clonal
Biochem Biophys Res Commun 205:154755. development of osteoclast precursors in primate bone
123. Mochizuki H, Hakeda Y, Wakatsuki N, Usui N, Akashi marrow. Exp Hematol 21:5327.
S, Sato T, et al. (1992) Insulin-like growth factor-I 138. Ragab AA, Lavish SA, Banks MA, Goldberg VM,
supports formation and activation of osteoclasts. Greeneld EM (1998) Osteoclast differentiation
Endocrinology 131:107580. requires ascorbic acid. J Bone Miner Res 13:9707.
124. Mori K, Nishimura M, Tsurudome M, Ito M, Nishio M, 139. Ragab AA, Nalepka JL, Bi Y, Greeneld EM (2002)
Kawano M, et al. (2003) The functional interaction Cytokines synergistically induce osteoclast differenti-
between CD98 and CD147 in regulation of virus- ation: support by immortalized or normal calvarial
induced cell fusion and osteoclast formation. Med cells. Am J Physiol Cell Physiol 283:C67987.
Microbiol Immunol (Berl). 140. Roggia C, Gao Y, Cenci S, Weitzmann MN, Toraldo G,
125. Morishima S, Morita I, Tokushima T, Kawashima H, Isaia G, et al. (2001) Up-regulation of TNF-producing
Miyasaka M, Omura K, et al. (2003) Expression and role T cells in the bone marrow: a key mechanism by which
of mannose receptor/terminal high-mannose type estrogen deciency induces bone loss in vivo. Proc Natl
oligosaccharide on osteoclast precursors during osteo- Acad Sci USA 98:139605.
clast formation. J Endocrinol 176:28592. 141. Roodman GD, Ibbotson KJ, MacDonald BR, Kuehl TJ,
126. Motyckova G, Weilbaecher KN, Horstmann M, Rieman Mundy GR (1985) 1,25-Dihydroxyvitamin D3 causes
DJ, Fisher DZ, Fisher DE (2001) Linking osteopetrosis formation of multinucleated cells with several osteo-
and pycnodysostosis: regulation of cathepsin K expres- clast characteristics in cultures of primate marrow.
sion by the microphthalmia transcription factor Proc Natl Acad Sci USA 82:821317.
family. Proc Natl Acad Sci USA 98:5798803. 142. Rubin C, Gross T, Qin YX, Fritton S, Guilak F, McLeod
127. Nakamura I, Tanaka H, Rodan GA, Duong LT (1998) K (1996) Differentiation of the bone-tissue remodeling
Echistatin inhibits the migration of murine prefusion response to axial and torsional loading in the turkey
osteoclasts and the formation of multinucleated osteo- ulna. J Bone Joint Surg Am 78:152333.
clast-like cells. Endocrinology 139:518293. 143. Rubin C, Lanyon L (1984) Regulation of bone forma-
128. Nakano H, Oshima H, Chung W, Williams-Abbott L, tion by applied dynamic loads. J Bone Joint Surgery
Ware C, Yagita H, et al. (1996) TRAF5, an activator of 66A:397402.
Osteoclast: Origin and Differentiation 21

144. Rubin J, Ackert-Bicknell C, Zhu L, Fan X, Murphy T, 162. Suda T, Suda J, Kajigaya S, Nagata S, Asano S, Saito M,
Nanes M, et al. (2002) Insulin Like Growth Factor-I et al (1987) Effects of recombinant murine granulocyte
(IGF-I) regulates osteoprotegerin (OPG) and RANK colony-stimulating factor on granulocyte-macrophage
Ligand (RANKL) in vitro and osteoprotogerin in vivo. and blast colony formation Exp Hematol 15:95865.
Journal of Clinical Endocrinology and Metabolism 163. Suda T, Takahashi N, Martin J (1992) Modulation of
87:42739. osteoclast differentiation. Endocrine Rev 13:6680.
145. Rubin J, Fan X, Thornton D, Bryant R, Biskobing D 164. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT,
(1996) Regulation of murine osteoblast macrophage Martin TJ (1999) Modulation of osteoclast differentia-
colony-stimulating factor production by 1,25(OH)2D3. tion and function by the new members of the tumor
Calcied Tissue 59:2916. necrosis factor receptor and ligand families. Endocr
146. Rubin J, Murphy T, Fan X, Goldschmidt M, Taylor W Rev 20:34557.
(2002) Mechanical strain inhibits RANKL expression 165. Suda T, Udagawa N, Nakamura I, Miyaura C, Takahashi
through activation of ERK1/2 in bone marrow stromal N (1995) Modulation of osteoclast differentiation by
cells. J Bone Miner Res 17:145260. local factors. Bone 17:87S91S.
147. Rubin J, Murphy T, Nanes MS, Fan X (2000) Mechani- 166. Szulc P, Hofbauer LC, Heufelder AE, Roth S, Delmas PD
cal strain inhibits expression of osteoclast differentia- (2001) Osteoprotegerin serum levels in men: correla-
tion factor by murine stromal cells. Am J Physiol Cell tion with age, estrogen, and testosterone status. J Clin
Physiol 278:C112632. Endocrinol Metab 86:31625.
148. Rubin J, Titus L, Nanes MS (1992) cAMP promotion of 167. Takahashi N, Udagawa N, Akatsu T, Tanaka H,
osteoclast-like cell development from mouse bone Shionome M, Suda T (1991) Role of colony-stimulating
marrow cells requires a permissive action of 1,25- factors in osteoclast development. J Bone Miner Res
(OH)2D3. J Bone Miner Res 7:61117. 6:97785.
149. Ruther U, Komitowski D, Schubert FR, Wagner EF 168. Takahashi N, Yamana H, Yoshiki S, Roodman GD,
(1989) c-fos expression induces bone tumors in trans- Mundy GR, Jones SJ, et al. (1988) Osteoclast-like cell
genic mice. Oncogene 4:8615. formation and its regulation by osteotropic hormones
150. Saika M, Inoue D, Kido S, Matsumoto T (2001) 17beta- in mouse bone marrow cultures. Endocrinology 122:
estradiol stimulates expression of osteoprotegerin by a 137382.
mouse stromal cell line, ST-2, via estrogen receptor- 169. Takahashi S, Reddy SV, Chirgwin JM, Devlin R, Haipek
alpha. Endocrinology 142:220512. C, Anderson J, et al. (1994) Cloning and identication
151. Sato T, Morita I, Murota S (1998) Involvement of cho- of annexin II as an autocrine/paracrine factor that
lesterol in osteoclast-like cell formation via cellular increases osteoclast formation and bone resorption.
fusion. Bone 23:13540. J Biol Chem 269:28696701.
152. Sells Galvin RJ, Gatlin CL, Horn JW, Fuson TR (1999) 170. Takai H, Kanematsu M, Yano K, Tsuda E, Higashio K,
TGF-beta enhances osteoclast differentiation in Ikeda K, et al. (1998) Transforming growth factor-
hematopoietic cell cultures stimulated with RANKL beta stimulates the production of osteoprotegerin/
and M-CSF. Biochem Biophys Res Commun 265:2339. osteoclastogenesis inhibitory factor by bone marrow
153. Sherr CJ (1990) Colony-stimulating factor-1 receptor. stromal cells. J Biol Chem 273:270916.
Blood 75:112. 171. Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M,
154. Sherr CJ (1990) The colony-stimulating factor 1 recep- Yoshida H, et al. (2002) Induction and activation of
tor: pleiotropy of signal-response coupling. Lym- the transcription factor NFATc1 (NFAT2) integrate
phokine Res 9:5438. RANKL signaling in terminal differentiation of osteo-
155. Sherr CJ (1991) Mitogenic response to colony- clasts. Dev Cell 3:889901.
stimulating factor 1. Trends Genet 7:398402. 172. Takayanagi H, Kim S, Matsuo K, Suzuki H, Suzuki T,
156. Shevde NK, Bendixen AC, Dienger KM, Pike JW (2000) Sato K, et al. (2002) RANKL maintains bone home-
Estrogens suppress RANK ligand-induced osteoclast ostasis through c-Fos-dependent induction of inter-
differentiation via a stromal cell independent mecha- feron-beta. Nature 416:7449.
nism involving c-Jun repression. Proc Natl Acad Sci 173. Takayanagi H, Ogasawara K, Hida S, Chiba T, Murata S,
USA 97:782934. Sato K, et al. (2000) T-cell-mediated regulation of
157. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang osteoclastogenesis by signalling cross-talk between
MS, Luthy R, et al. (1997) Osteoprotegerin: a novel RANKL and IFN-gamma. Nature 408:6005.
secreted protein involved in the regulation of bone 174. Takeshita S, Namba N, Zhao JJ, Jiang Y, Genant HK,
density. Cell 89:30919. Silva MJ, et al. (2002) SHIP-decient mice are severely
158. Soriano P, Montgomery C, Geske R, Bradley A (1991) osteoporotic due to increased numbers of hyper-
Targeted disruption of the c-src proto-oncogene leads resorptive osteoclasts. Nat Med 8:9439.
to osteopetrosis in mice. Cell 64:693702. 175. Tanaka S, Takahashi N, Udagawa N, Tamura T, Akatsu
159. Spangrude GJ, Heimfeld S, Weissman IL (1988) Puri- T, Stanley E, et al. (1993) Macrophage colony stimulat-
cation and characterization of mouse hematopoietic ing factor is indispensable for both proliferation and
stem cells. Science 241:5862. differentiation of osteoclast progenitors. J Clin Invest
160. Stanley ER, ed.: Thomson A (1994) Colony stimulat- 92:25763.
ing factor-1 (Macrophage colony stimulating factor). 176. Thesingh CW, Scherft JP (1985) Fusion disability of
The Cytokine Handbook. 387418, Academic Press, embryonic osteoclast precursor cells and macrophages
London. in the microphthalmic osteopetrotic mouse. Bone
161. Steinbeck MJ, Kim JK, Trudeau MJ, Hauschka PV, 6:4352.
Karnovsky MJ (1998) Involvement of hydrogen per- 177. Thirunavukkarasu K, Halladay D, Miles R, Yang X,
oxide in the differentiation of clonal HD-11EM cells Galvin R, Chandrasekhar S, et al. (2000) The
into osteoclast-like cells J Cell Physiol 176:57487. osteoblast-specic transcription factor Cbfa1 con-
22 Bone Resorption

tributes to the expression of osteoprotegerin, a potent Podgornik MN, McAlister WH, et al. (2002) Osteopro-
inhibitor of osteoclast differentiation and function. J tegerin deciency and juvenile Pagets disease. N Engl
Biol Chem 276:2516372. J Med 347:17584.
178. Thomas GP, Baker SU, Eisman JA, Gardiner EM (2001) 193. Wiktor-Jedrzejczak W, Bartocci A, Ferrante AJ, Ahmed-
Changing RANKL/OPG mRNA expression in differen- Ansari A, Sell K, Pollard J, et al. (1990) Total absence
tiating murine primary osteoblasts. J Endocrinol 170: of colony-stimulating factor 1 in the macrophage-
45160. decient osteopetrotic (op/op) mouse. Proc Natl Acad
179. Tondravi MM, McKercher SR, Anderson K, Erdmann Sci USA 87:482832.
JM, Quiroz M, Maki R, et al. (1997) Osteopetrosis in 194. Wimalawansa S, Chapa T, Fang L, Yallampalli C,
mice lacking haematopoietic transcription factor PU.1. Simmons D (2000) Frequency-dependent effect of
Nature 386:814. nitric oxide donor nitroglycerin on bone. J Bone Miner
180. Toraldo G, Roggia C, Qian WP, Pacici R, Weitzmann Res 15:111925.
MN (2003) IL-7 induces bone loss in vivo by induction 195. Wimalawansa SJ (2000) Nitroglycerin therapy is as ef-
of receptor activator of nuclear factor kappa B ligand cacious as standard estrogen replacement therapy
and tumor necrosis factor alpha from T cells. Proc Natl (Premarin) in prevention of oophorectomy-induced
Acad Sci USA 100:12530. bone loss: a human pilot clinical study. J Bone Miner
181. Udagawa N, Takahashi N, Jimi E, Matsuzaki K, Tsurukai Res 15:22404.
T, Itoh K, et al. (1999) Osteoblasts/stromal cells 196. Wong BR, Besser D, Kim N, Arron JR, Vologodskaia M,
stimulate osteoclast activation through expression of Hanafusa H, et al. (1999) TRANCE, a TNF family
osteoclast differentiation factor/RANKL but not member, activates Akt/PKB through a signaling
macrophage colony-stimulating factor: receptor acti- complex involving TRAF6 and c-Src. Mol Cell
vator of NF-kappaB ligand. Bone 25:51723. 4:10419.
182. Udagawa N, Takahashi N, Katagiri T, Tamura T, Wada 197. Wong BR, Josien R, Lee SY, Sauter B, Li HL, Steinman
S, Findlay DM, et al. (1995) Interleukin (IL)-6 induc- RM, et al. (1997) TRANCE (tumor necrosis factor
tion of osteoclast differentiation depends on IL-6 [TNF]-related activation-induced cytokine), a new
receptors expressed on osteoblastic cells but not on TNF family member predominantly expressed in T
osteoclast progenitors. J Exp Med 182:14618. cells, is a dendritic cell-specic survival factor. J Exp
183. Udagawa N, Takahashi N, Yasuda H, Mizuno A, Itoh K, Med 186:207580.
Ueno Y, et al. (2000) Osteoprotegerin produced by 198. Wong BR, Josien R, Lee SY, Vologodskaia M, Steinman
osteoblasts is an important regulator in osteoclast RM, Choi Y (1998) The TRAF family of signal trans-
development and function. Endocrinology 141:3478 ducers mediates NF-kappaB activation by the
84. TRANCE receptor. J Biol Chem 273:283559.
184. Underwood J (1984) From where comes the osteoclast? 199. Yagami-Hiromasa T, Sato T, Kurisaki T, Kamijo K,
J Pathol 144:2256. Nabeshima Y, Fujisawa-Sehara A (1995) A metallopro-
185. van de Wijngaert FP, Tas MC, Burger EH (1987) Char- tease-disintegrin participating in myoblast fusion.
acteristics of osteoclast precursor-like cells grown Nature 377:6526.
from mouse bone marrow. Bone Miner 3:11123. 200. Yakar S, Rosen CJ (2003) From mouse to man: reden-
186. Van Wesenbeeck L, Odgren PR, MacKay CA, DAngelo ing the role of insulin-like growth factor-I in the acqui-
M, Safadi FF, Popoff SN, et al. (2002) The osteopetrotic sition of bone mass. Exp Biol Med (Maywood) 228:
mutation toothless (tl) is a loss-of-function frameshift 24552.
mutation in the rat Csf1 gene: Evidence of a crucial role 201. Yakar S, Rosen CJ, Beamer WG, Ackert-Bicknell CL,
for CSF-1 in osteoclastogenesis and endochondral Wu Y, Liu JL, et al. (2002) Circulating levels of IGF-1
ossication. Proc Natl Acad Sci USA 99:143038. directly regulate bone growth and density. J Clin Invest
187. Walker DG (1975) Bone resorption restored in 110:77181.
osteopetrotic mice by transplants of normal bone 202. Yamane T, Kunisada T, Yamazaki H, Nakano T, Orkin
marrow and spleen cells. Science 190:7845. SH, Hayashi SI (2000) Sequential requirements for
188. Wani MR, Fuller K, Kim NS, Choi Y, Chambers T (1999) SCL/tal-1, GATA-2, macrophage colony-stimulating
Prostaglandin E2 cooperates with TRANCE in osteo- factor, and osteoclast differentiation factor/osteoprote-
clast induction from hemopoietic precursors: syner- gerin ligand in osteoclast development. Exp Hematol
gistic activation of differentiation, cell spreading, and 28:83340.
fusion. Endocrinology 140:192735. 203. Yamashita T, Asano K, Takahashi N, Akatsu T, Udagawa
189. Weilbaecher KN, Motyckova G, Huber WE, Takemoto N, Sasaki T, et al. (1990) Cloning of an osteoblastic cell
CM, Hemesath TJ, Xu Y, et al. (2001) Linkage of M-CSF line involved in the formation of osteoclast-like cells.
signaling to Mitf, TFE3, and the osteoclast defect in J Cell Physiol 145:58795.
Mitf(mi/mi) mice. Mol Cell 8:74958. 204. Yao GQ, Sun B, Hammond EE, Spencer EN, Horowitz
190. Weir E, Lowik C, Paliwal I, Insogna K (1996) Colony MC, Insogna KL, et al. (1998) The cell-surface form of
stimulating factor-1 plays a role in osteoclast forma- colony-stimulating factor-1 is regulated by osteotropic
tion and function in bone resorption induced by agents and supports formation of multinucleated
parathyroid hormone and parathyroid hormone- osteoclast- like cells. J Biol Chem 273:411928.
related protein. J Bone Miner Res 11:147481. 205. Yao GQ, Wu JJ, Sun BH, Troiano N, Mitnick MA,
191. Weitzmann MN, Cenci S, Rifas L, Haug J, Dipersio J, Insogna K (2003) The cell surface form of colony-
Pacici R (2001) T cell activation induces human stimulating factor-1 is biologically active in bone in
osteoclast formation via receptor activator of nuclear vivo. Endocrinology 144:367782.
factor kappaB ligand-dependent and -independent 206. Yasuda H, Shima N, Nakagawa N, Yamaguchi K,
mechanisms. J Bone Miner Res 16:32837. Kinosaki M, Mochizuki S, et al. (1998) Osteoclast dif-
192. Whyte MP, Obrecht SE, Finnegan PM, Jones JL, ferentiation factor is a ligand for osteoprotegerin/
Osteoclast: Origin and Differentiation 23

osteoclastogenesis-inhibitory factor and is identical osteopetrosis is in the coding region of the


to TRANCE/RANKL. Proc Natl Acad Sci USA 95:3597 macrophage colony stimulating factor gene. Nature
602. 345:4424.
207. Yeh W, Shahinian A, Speiser D, Kraunus J, Billia F, 209. Yu C, Shen K, Lin M, Chen P, Lin C, Chang GD, et al.
Wakeham A, et al. (1997) Early lethality, functional NF- (2002) GCMa regulates the syncytin-mediated tro-
kappaB activation, and increased sensitivity to TNF- phoblastic fusion. J Biol Chem 277:500628.
induced cell death in TRAF2-decient mice. Immunity 210. Zhang J, Dai J, Qi Y, Lin DL, Smith P, Strayhorn C,
7:71525. et al. (2001) Osteoprotegerin inhibits prostate cancer-
208. Yoshida H, Hayashi S, Kunisada T, Ogawa M, Nishikawa induced osteoclastogenesis and prevents prostate
S, Okamura H, et al. (1990) The murine mutation tumor growth in the bone. J Clin Invest 107:123544.
2.
Osteolytic Enzymes of Osteoclasts
Merry Jo Oursler

resorbed. It has therefore been of great in-


Introduction terest to discover the identity and nature of the
enzymes that execute bone resorption in the
Ninety percent of the protein in bone is type I hope of targeting these enzymes to regulate
collagen in the form of a cross-linked triple rates of bone loss. The osteolytic roles of TRAP,
helical structure, making it more resistant cathepsins, and MMPs are discussed in the
to proteolysis and enzymatic cleavage. The remainder of this chapter.
stability that this structure confers means that
specic proteases are required for effective
degradation [112, 113]. Bone also contains min-
erals in the form of hydroxyapatite crystals. Tartrate-Resistant Acid
Thus, for bone resorption to occur both the
mineral and the protein components of this
Phosphatase (TRAP)
matrix must be removed. Osteoclasts are unique
in that they create an external acidic hemivac- TRAP, expressed mainly in osteoclasts and
uole adjacent to the bone surface as an early step related monohistiocytic cells, has the capacity
in bone resorption [4]. This compartment is to hydrolyze phosphoproteins, ATP and other
created when osteoclasts attach to bone. After nucleotide triphosphates, and arylphosphates
attachment, osteoclasts transport lysosomes to [73].Although it has been known for many years
the portion of the plasma membrane that is that osteoclasts express this membrane enzyme,
juxtaposed to the area of the bone that is to its role in osteoclast activity has remained
be resorbed. The vesicles fuse with the plasma elusive. The porcine homolog, uteroferrin, may
membrane and thereby increase the plasma have as its function the delivery of iron that is
membrane surface area in this restricted region. secreted by the endometrium in utero [29].
Lysosome membranes also contain membrane However, there is no indication that the enzyme
tartrate-resistant acid phosphatase (TRAP) and lls this potential role in osteoclasts. Although
high levels of a hydrogen pump. Both of these there is only limited sequence similarity
enzymes are retained in the plasma membrane between the kidney bean purple acid phos-
after fusion. Lysosome contents are discharged phatase and TRAP, the fact that the conforma-
into this compartment as the vesicles fuse with tion of what is thought to be the active site of
the plasma membrane and this is followed by TRAP resembles that of the known catalytic
hydrogen pump-mediated acidication of the domain of the bean phosphatase strengthens
hemivacuole. The combined action of enzymes the inference that the presumed site in TRAP is
from the lysosomes (such as TRAP and cathep- in fact the active site [111]. The crystal structure
sins), the matrix metalloproteases (MMPs), and of TRAP also revealed a protease sensitive
the reduction in pH together cause bone to be surface loop near the active site [111]. This
24
Osteolytic Enzymes of Osteoclasts 25

explains the need for proteolytic activation of considered a pivotal marker for osteoclasts,
the enzyme. attempts have been made to correlate serum
Targeted TRAP disruption in mice resulted TRAP levels with resorption rates in vivo. This
in altered epiphyseal growth plate develop- has met with little success. Attempts have
ment [55]. This is consistent with a disruption recently been made to measure type 5b TRAP,
in chondro/osteoclast activity. In these animals, whose expression is thought to be restricted
moreover, early onset osteopetrosis was ob- osteoclasts [60].
served. Since osteoclast differentiation levels
seemed normal, the osteopetrosis probably
resulted from reduced osteoclast activity. Given
that osteoclasts in these mice contain high
Cathepsins
numbers of cytoplasmic vesicles near the rufed
border, the question arose whether TRAP in the Many studies have linked these lysosomal cys-
lysosomal vesicle membrane was involved in teine proteases to osteoclast-mediated bone
transport to or fusion with the plasma mem- resorption. Much of the evidence indicates that
brane. Osteoclasts from these mice secreted cysteine protease inhibition can block bone
normal levels of cathepsin K, leading to the resorption in vitro and in vivo [17, 19, 20, 22, 36,
conclusion that the accumulating vesicles were 46, 47, 53, 69, 72, 87, 90, 95, 99]. This includes
not lysosomal vesicles that fail to merge with studies where the activities of cathepsin B and
the plasma membrane [55]. Microphthalmia- cathepsin L were inhibited and resorption was
associated transcription factor (MITF) and PU.1 slowed or stopped [22, 46, 47, 69, 87, 90, 95, 99].
are two transcription factors that synergistically In some studies, however, inhibition of either
activate TRAP gene expression [79]. Mice that protease had only a modest or no effect on
are heterozygous for mutant MITF and a PU.1 resorption [52, 64, 95, 110, 111, 118]. This has
null allele exhibit early-onset osteopetrosis with led to a search for alternate proteases respon-
osteoclasts of normal size and number [79]. The sible for osteoclast-mediated bone resorption.
above studies support the concept that TRAP is Cathepsin K (also referred to as O or X) was
involved in osteoclast activity, but its precise rst identied by differential screening of a
role has remained unresolved. Recent data rabbit osteoclast cDNA library and both human
document that cathepsin K is a physiologi- and mouse homologs have subsequently been
cal activator of TRAP and that TRAP can cloned [61, 92, 109]. Mice with cathepsin K
dephosphorylate osteopontin [3]. Osteopontin knocked out exhibited osteopetrosis, abnormal
is involved in stimulating osteoclast activity by joint morphology, increased bone volume,
promoting adhesion, migration, and resorption greater trabecular thickness, and more trabecu-
[13, 14, 30, 39, 57, 59, 82, 93, 96]. Razzouk et al. lae [48]. Differentiated osteoclasts were found in
[93] have investigated the importance of osteo- association with demineralized bone. This sug-
pontin phosphorylation in these roles and their gests that osteoclasts decient in cathepsin K
data support that osteopontin phosphorylation can demineralize the matrix but cannot degrade
may be involved in promoting resorption, but it. In other studies of cathepsin K null mice,
not attachment or actin ring formation. Thus, osteopetrosis was observed even though the
TRAP-mediated osteopontin dephosphoryla- osteoclast numbers were normal, suggesting
tion would repress bone resorption. impaired osteoclast activity [97, 98]. In addition,
TRAP is expressed in osteoclasts, some tissue the resorption surface was broadly demineral-
macrophages, dendritic cells, parenchymal ized, but there was undigested collagen present.
liver cells, glomerular mesangial kidney cells, The absence of cathepsin K and the presence
and pancreatic acinar cells [115]. In many cases, of undigested collagen suggest that cathepsin K
there are different RNAs expressed by these dif- is involved in collagen degradation. Cultured
ferent cell types as the 5 untranslated regions of osteoclasts from these mice also had impaired
the mRNAs differ due to altered rst exons. resorption activity [98]. Studies of human
Walsh et al. [115] have identied an osteoclast- cathepsin K using giant cell tumors of the
specic transcription initiation site and further bone and bone tissues show that cathepsin K
documented that there are four different pro- was highly expressed in osteoclasts, with
moter regions that are restricted in a tissue- and no detectable cathepsin B or L expression [28].
cell-specic manner. Since TRAP has long been From these studies it appears that other tissues
26 Bone Resorption

do not express cathepsin K. However, the failure lagen nonhelical regions or the destabilized
to detect cathepsins B and L may simply be due helical region once collagenase has cleaved the
to an insufciently sensitive assay, inasmuch as structure [94]. Unlike other cathepsins, cathep-
in other reports these enzymes are detected in sin K cleaves both helical and telopeptide
osteoclasts [22, 46, 47, 69, 87, 90, 95, 99]. Indeed, regions of collagen [43]. Thus, typical of cathep-
although the above study suggested that cathep- sins, cathepsin K can cleave outside of the
sin K might be exclusively expressed by osteo- helical region. In addition, it can also act inside
clasts, it is clear from other data that cathepsin the helical region, i.e., the region that is usually
K expression is not restricted to osteoclasts attacked by the MMPs and neutral elastase [2, 6,
[1012, 15, 25, 26, 4951, 68, 74, 78, 83, 84, 91, 92, 65, 75, 76, 88]. This dual ability to target both
107]. collagen regions is unique among the mam-
The human disease pycnodysostosis is a lyso- malian collagenases and is reminiscent of bac-
some disease that manifests itself as an inher- terial collagenase. A conrmation of the critical
ited sclerosing skeletal dysplasia. Gelb et al. role played by cathepsin K in collagen digestion
[44] have shown that patients with this disease in humans comes from a study in which bro-
have nonsense, missense, or stop codon muta- blasts in pycnodysostotic embryos accumulate
tions in their cathepsin K genes. Similar to undigested collagen in lysosomal vacuoles [37].
mouse models lacking cathepsin K, pyc- Thus, cathepsin K can degrade the stable colla-
nodysostotic osteoclasts can demineralize bone gen triple helical structure and is therefore
matrix but cannot degrade the matrix. These likely to be a reasonable target for pharmaco-
data support that cathepsin K is involved in logic intervention to block bone resorption
human osteoclast-mediated bone resorption. In (reviewed in [117]). As noted above, another
mice, Rantakokko et al. [92] observed cathepsin role for cathepsin K in bone resorption may be
K in both osteoclasts and hypertrophic chon- to activate TRAP, causing osteopontin dephos-
drocytes. This observation adds support to the phorylation and thereby inhibiting bone
hypothesis that cathepsin K plays a role in resorption.
matrix degradation as well. Cathepsin K inhibition by antisense or small
An interesting study by Dodds et al. [27] has molecules has been used to assess its role in
examined the patterns of inactive and active osteoclast-mediated bone resorption. Pharma-
cathepsin K protein in osteoclasts. In osteoclasts cological inhibitors of cathepsin K that span the
that are not involved in bone resorption, most active site inhibit resorption both in vivo and in
of the cathepsin K was present as an inactive vitro [71, 106, 110]. Blocking cathepsin K with
zymogen. In osteoclasts that were located closer antisense repressed bone resorption as well
to the bone matrix, active cathepsin K appeared [62]. In an in vivo study using SB-357114 to
restricted to the osteoclast area closest to the block bone resorption, the inhibitor blocked
bone. When osteoclasts were actively resorbing both cathepsin K and cathepsin L activity [106].
bone, they contained only active cathepsin K Since these osteoclasts expressed much more
that was localized at the bone surface in the cathepsin K than cathepsin L, cathepsin K may
rufed membrane. Thus, local factors present in be the likely inhibition target, but this has not
the immediate vicinity of the bone seem to be been directly tested. Independent of the molec-
important in stimulating cathepsin K activation. ular target of this inhibitor, resorption was
Early studies of the ability of cathepsin K blocked, raising hope that targeted disruption
to degrade bone proteins suggested that it was of cathepsins may become an effective anti-
not very effective against collagen or resorptive therapy.
bronectin, but readily degraded osteonectin This returns us to the issue of the conicting
[9]. It has since been documented that optimal data of the potential roles of different cathepsins
cathepsin K activity toward collagen requires a in osteoclast-mediated bone resorption. An
complex of the cathepsin molecule and soluble interesting study by Furuyama et al. [42] may
glycosaminoglycans [58]. Intriguingly, Hou et help resolve this issue. Using calvarial organ cul-
al. [58] have shown that cathepsin K cleaved tures, they were able to detect trace levels of
aggrecan aggregates at two specic sites, thereby cathepsin L in untreated osteoclasts. When the
generating the soluble glycosaminoglycans cultures were treated with 1,25-dihydroxyvita-
required for formation of the active complex min D3, parathyroid hormone, interleukin-1a,
themselves. Most cathepsins typically target col- interleukin-6, or tumor necrosis factor-a all
Osteolytic Enzymes of Osteoclasts 27

known to stimulate bone loss the cathepsin L extracellular matrix proteins [104]. Other non-
levels increased. In contrast, cathepsin K was matrix targets also exist for each MMP [104].
abundant under basal conditions and its con- In bone, osteoblasts are a major source of
centration was unchanged following treatment MMP production. However, MMP13 in bone is
with these stimulatory agents. Under basal con- produced by osteocytes or mononuclear cells
ditions, resorption was blocked when cathep- located near osteoclasts [23, 41, 102, 103]. Osteo-
sin K was inhibited, but not when cathepsin L clasts also produce some MMPs. Lin et al.
was inhibited. However, when the calvaria were [77] found that rat osteoclasts expressed MMP1
treated with stimulatory factors, inhibition of mRNA. Further, rabbit osteoclasts express
either cathepsin K or cathepsin L partially in- MMP9 mRNA and MMP1 and 9 mRNAs were
hibited resorption, with inhibition becoming present in human neonatal rib osteoclasts [7,
additive when both inhibitors were used. These 108]. Intriguingly, MMP1 and MMP9 mRNAs
ndings demonstrate that cathepsin K is impor- also were seen in a subset of osteoclasts situated
tant in basal bone resorption. Cathepsin L, on at resorption sites, and MMP9 mRNA was
the other hand, may be involved in bone loss expressed in human giant cell tumors with the
under pathological conditions when its expres- degree of expression correlating with the sever-
sion is elevated. ity of the osteolysis [8, 70]. Moreover, osteoclast
MMP1 and 9 production is regulated by IL-1, a
cytokine that promotes bone resorption in vivo
Matrix Metalloproteases [54]. These data suggest a role for MMPs 1 and
9 in osteoclast-mediated bone degradation.
(MMPs) All MMPs require zinc for activity and,
compared to MMP1, MMP3 has a deeper S1-
Studies of patients with pycnodysostosis have specicity pocket, which is distinctly different
shown that, for the typical bone phenotype to from the active site of MMP1 [24]. This could
become evident, there must be complete loss of explain why it has proven possible to develop
cathepsin K activity [44]. However, even in cases specic inhibitors that differentiate between
with complete loss, whether in patients or mice, these two enzymes. X-ray structure compar-
the calvaria appear normal. It would therefore isons of MMPs 1 and 3 indicate there are two
seem that some bone resorption is taking place clusters of regions of differences between these
[16]. The collagenolytic mechanisms in these two MMPs in regions that form the entrances to
cases remain unknown, but may involve over- the active sites [24]. Differences in substrate
expression of other cathepsins, perhaps in con- specicity may be attributed to these structural
junction with MMPs. differences [24].
Individual MMPs are known by a variety of Given that MMPs can destroy tissues, their
names and their substrates are as varied as their enzymatic activity must be tightly regulated
names. We will discuss MMP1, 3, 9, 12, 13, and [104]. MMP activity can be controlled by mod-
14 and their potential roles in osteoclast- ulating the gene expression level, by stimulating
mediated bone resorption. MMP1 also is known or inhibiting proenzyme activation, or with the
as collagenase-I. Its matrix substrates include aid of inhibitory molecules such as the tissue
intact collagen types I, II, III, VII, VIII, X [104]. inhibitors of metalloproteinases (TIMPs) [105].
MMP3 also is known as stromelysin and its Expression stimulators include the membrane-
substrates include osteopontin [1], decorin, anchored reversion-inducing cysteine-rich
proteoglycan, bronectin, laminin, and type IV protein with Kazal motifs (RECK) and alpha2-
collagen, but not interstitial type I collagen macroglobulin [81, 85, 86]. MMPs are synthe-
[116]. MMP9 also is known as gelatinase B and sized with a pro-domain that blocks activity by
its substrates include aggrecans, denatured type means of an unpaired cysteine in the carboxy
I collagen [40], and collagens IV, V, VIII, X, and terminus. X-ray analysis has shown that, by
XIV [104]. MMP12 also is known as macrophage inserting the pro-domains of MMP3 and 9 into
metalloelastase. Its substrate is elastin and it their respective clefts, activity is blocked [5, 31].
has no collagen substrates [104]. MMP13 is also The unpaired cysteine acts as a fourth inacti-
known as collagenase-3 and MMP14 is also vating ligand for the zinc atom that is present in
known as MTI-MMP. Their substrates include the active site. Activation can be by either pro-
type I collagen, other collagens, and additional teolysis, so as to remove the pro-domain, or by
28 Bone Resorption

a conformational change that releases the zinc family members. Bone resorption rates were
atom from the cysteine. In that situation, the normal in mice lacking MMP9, MMP12, and
thiol group is replaced by water and the enzyme MMP14 [16]. However, mice lacking MMP13
is thus enabled to remove the pro-domain. In had reduced bone resorption rates. This nding
most cases, MMP activation takes place outside again supports a role for MMP13 in bone
of the cell. Given that MMPs reside in bone resorption. Although there was no apparent
as latent enzymes, MMP family members not impact of MMP9 knock-out on bone resorption,
synthesized by osteoclasts can be activated by the bones from these mice had lengthened
osteoclastic proteolytic activity and then par- growth plates [114]. Since introduction of
ticipate in bone degradation. Inactivation normal marrow restored development to
mechanisms include expression of TIMPs, normal, it was apparent that the defect was in
which interact with the active site of MMPs, the chondroclasts [114]. MMP9 knockout mice
and thus block activity [45]. In normal human exhibited delayed osteoclast recruitment [32]. In
bone, TIMP1 is highly expressed in osteoclasts patients with vanishing bone syndrome, there is
derived from neonatal bone, but was absent in severe bone degradation, mainly in the hands
osteoclasts from pathological tissues (osteo- and the feet; this is due to the loss of the MMP2
phytic and heterotopic bone both having gene [80]. The reasons that loss of an MMP
poorly organized bone formation) [7]. TIMP1 causes bone destruction are unknown, but
expression may therefore be important in the possibilities include a compensatory overpro-
regulation of normal bone remodeling in the duction of other MMPs or a loss of coupling
course of development. between osteoclasts and osteoblasts. Mice
Selective MMP inhibition studies have shown lacking MMP2 do not exhibit the same pheno-
that the functional importance of a given MMP type. This is another indication why reliance on
varies with the type of bone [38]. Moreover, the mouse genetics may mislead in understanding
presence of inammatory cytokines may alter human disease.
MMP levels. Hill et al. [54] have documented MMPs also facilitate other aspects of osteo-
that MMP9 inhibition did not inhibit basal clast function. Inhibition of MMPs blocked
osteoclast activity in vitro, but did inhibit IL-1- osteoclast migration through collagen gel in an
stimulated calvarial explant bone resorption. in vitro assay [16]. Osteoclasts decient in
Consequently, MMP9 may play a role in inam- MMPs 9, 12, and 14 have been examined for
matory cytokine-stimulated bone loss. Thus the their migratory patterns [16]. MMP12-decient
specic function of MMPs in bone resorption osteoclasts migrated at normal rates, whereas
seems to depend on the bone and/or the type of osteoclasts decient in MMP9 or MMP14 had
resorption stimulus. slower migration patterns. The MMPs can be
Inhibition of either cathepsin or MMP de- found in podosomes, the osteoclast-bone
creased bone resorption, but inhibiting both was attachment structure, and at the leading edges
not additive [34, 36]. Even though cathepsins of normal migrating osteoclasts [63, 100].
and MMPs thus appear to have similar functions Further supporting a MMP role in migration,
and may be able to substitute for each other, MMP inhibition increased the lifespan of a
MMPs act later in the resorption process than podosome; this in turn will decrease migra-
cathepsins do [38]. In view of the fact that these tion[100]. Another function of MMPs may be to
two enzyme classes act on different resorption stimulate osteoclast recruitment [16].
steps, their actions may not be additive. One hypothesis is that MMP13-generated col-
MMP13, synthesized by osteocytes but not by lagen fragments initiate bone resorption by
osteoclasts, has been detected in the resorption activating osteoclasts [56]. In addition, MMP9
pits of cathepsin K null mice [35]. This obser- activity released transforming growth factor-b
vation raises the possibility that osteocytes may from bone matrix, where transforming growth
participate in osteoclast-mediated bone resorp- factor- is stored in signicant amounts [66].
tion by synthesizing and secreting MMP13. TGF- causes cell retraction, thus leading to
Everts et al. [35] have proposed that the role greater bone surface exposure, increasing
of MMP13 is to mop up after cathepsin K- osteoclast precursor attraction to the bone
mediated bone resorption is completed. surface [66]. MMP14 causes membrane receptor
Studies of mice lacking a given MMP provide activator of NFkappaB ligand (RANKL) release,
insight into specic functions of the various thereby perhaps inuencing osteoclast differen-
Osteolytic Enzymes of Osteoclasts 29

tiation and/or activation [67, 101]. Thus, MMPs 3. Andersson G, Ek-Rylander B, Hollberg K, Ljusberg-
most likely coordinate matrix degradation and Sjolander J, Lang P, Norgard M, et al. (2003) TRACP
as an osteopontin phosphatase. J Bone Miner Res 18:
recruitment and differentiation of osteoclasts 191215.
and chondroclasts and may be an important 4. Baron R, Neff L, Louvard D, Courtoy PJ (1985) Cell-
component of bone metabolism [89]. mediated extracellular acidication and bone resorp-
tion: Evidence for a low pH in resorbing lacunae and
localization of a 100-kDa lysosomal membrane protein
at the osteoclast rufed border. J Cell Biol 101:2210
Conclusions 22.
5. Becker JW, Marcy AI, Rokosz LL, Axel MG, Burbaum
JJ, Fitzgerald PM, et al. (1995) Stromelysin-1: three-
In light of the above information, cooperation dimensional structure of the inhibited catalytic
domain and of the C-truncated proenzyme. Protein Sci
among proteases is likely to be important in 4:196676.
osteoclast-mediated bone resorption [18, 19, 21, 6. Birkedal-Hansen H, Moore WG, Bodden MK, Windsor
33, 36, 112, 113]. Although much more now is LJ, Birkedal-Hansen B, DeCarlo A, et al. (1993) Matrix
known about the enzymatic mechanisms by metalloproteinases: a review. Crit Rev Oral Biol Med
which bone is resorbed, it is equally clear that 4:197250.
7. Bord S, Horner A, Beeton CA, Hembry RM, Compston
many gaps in our knowledge remain. The role JE (1999) Tissue inhibitor of matrix metalloproteinase-
of TRAP in bone resorption remains unre- 1 (TIMP-1) distribution in normal and pathological
solved, as do the respective roles of cathepsins human bone. Bone 24:22935.
and MMPs. Furthermore, the mechanisms by 8. Bord S, Horner A, Hembry RM, Reynolds JJ, Compston
JE (1997) Distribution of matrix metalloproteinases
which resorption occurs in the course of bone and their inhibitor, TIMP-1, in developing human
development are likely to differ from those osteophytic bone. J Anat 191(Pt 1):3948.
involved in bone turnover in the process of 9. Bossard MJ, Tomaszek TA, Thompson SK, Amegadzie
pathological bone loss. Changes in bone mass BY, Hanning CR, Jones C, et al. (1996) Proteolytic
such as those that occur during menopause or activity of human osteoclast cathepsin K. Expression,
purication, activation, and substrate identication.
as a result of anti-inammatory therapies may J Biol Chem 271:1251724.
involve a different set of enzymes. Similarly, 10. Brubaker KD, Vessella RL, True LD, Thomas R, Corey
bone loss induced by inammatory cytokines E (2003) Cathepsin K mRNA and protein expression in
may involve mechanisms that differ from those prostate cancer progression. J Bone Miner Res 18:222
30.
in other conditions of bone resorption. It seems 11. Buhling F, Reisenauer A, Gerber A, Kruger S, Weber E,
reasonable to hope that future studies of oste- Bromme D, et al. (2001) Cathepsin K a marker of
olytic enzymes will improve our knowledge of macrophage differentiation? J Pathol 195:37582.
this important aspect of bone metabolism. 12. Buhling F, Waldburg N, Gerber A, Hackel C, Kruger S,
Reinhold D, et al. (2000) Cathepsin K expression in
human lung. Adv Exp Med Biol 477:2816.
13. Chellaiah MA, Hruska KA (2003) The integrin
Acknowledgments alpha(v)beta(3) and CD44 regulate the actions of
osteopontin on osteoclast motility. Calcif Tissue Int
72:197205.
14. Chellaiah MA, Kizer N, Biswas R, Alvarez U, Strauss-
Dr. Oursler is supported by the Department of Schoenberger J, Rifas L, et al. (2003) Osteopontin
the Army Grant DAMD17-00-1-0346 and the deciency produces osteoclast dysfunction due to
National Institutes of Health Grant DE14680. reduced CD44 surface expression. Mol Biol Cell 14:
17389.
15. Chiellini C, Costa M, Novelli SE, Amri EZ, Benzi L,
Bertacca A, et al. (2003) Identication of cathepsin K
References as a novel marker of adiposity in white adipose tissue.
J Cell Physiol 195:30921.
16. Delaisse JM, Andersen TL, Engsig MT, Henriksen K,
1. Agnihotri R, Crawford HC, Haro H, Matrisian LM, Troen T, Blavier L (2003) Matrix metalloproteinases
Havrda MC, Liaw L (2001) Osteopontin, a novel sub- (MMP) and cathepsin K contribute differently to
strate for matrix metalloproteinase-3 (stromelysin-1) osteoclastic activities. Microsc Res Tech 61:50413.
and matrix metalloproteinase-7 (matrilysin). J Biol 17. Delaisse JM, Boyde A, Maconnachie E, Ali NN, Sear CH,
Chem 276:282617. Eeckhout Y, et al. (1987) The effects of inhibitors of
2. Aimes RT, Quigley JP (1995) Matrix metalloproteinase- cysteine-proteinases and collagenase on the resorptive
2 is an interstitial collagenase. Inhibitor-free enzyme activity of isolated osteoclasts. Bone 8:30513.
catalyzes the cleavage of collagen brils and soluble 18. Delaisse JM, Eeckhout Y, Sear C, Galloway A,
native type I collagen generating the specic 3/4- and McCullagh K, Vaes G (1985) A new synthetic inhibitor
1/4-length fragments. J Biol Chem 270:58726. of mammalian tissue collagenase inhibits bone resorp-
30 Bone Resorption

tion in culture. Biochem Biophys Res Commun 133: play distinct roles in the subosteoclastic resorption
48390. zone. J Bone Miner Res 13:142030.
19. Delaisse JM, Eeckhout Y, Vaes G (1980) Inhibition of 35. Everts V, Delaisse JM, Korper W, Jansen DC, Tigchelaar-
bone resorption in culture by inhibitors of thiol pro- Gutter W, Saftig P, et al. (2002) The bone lining cell: its
teinases. Biochem J 192:3658. role in cleaning Howships lacunae and initiating bone
20. Delaisse JM, Eeckhout Y, Vaes G (1984) In vivo and in formation. J Bone Miner Res 17:7790.
vitro evidence for the involvement of cysteine pro- 36. Everts V, Delaisse JM, Korper W, Niehof A, Vaes G,
teinases in bone resorption. Biochem Biophys Res Beertsen W (1992) Degradation of collagen in the
Commun 125:4417. bone-resorbing compartment underlying the osteo-
21. Delaisse JM, Eeckhout Y, Vaes G (1985) Bisphospho- clast involves both cysteine-proteinases and matrix
nates and bone resorption: effects on collagenase and metalloproteinases. J Cell Physiol 150:22131.
lysosomal enzyme excretion. Life Sci 37:22916. 37. Everts V, Hou WS, Rialland X, Tigchelaar W, Saftig P,
22. Delaisse JM, Ledent P, Vaes G (1991) Collagenolytic Bromme D, et al. (2003) Cathepsin K Deciency in
cysteine proteinases of bone tissue. Cathepsin B, Pycnodysostosis Results in Accumulation of Non-
(pro)cathepsin L and a cathepsin L-like 70 kDa pro- Digested Phagocytosed Collagen in Fibroblasts. Calcif
teinase. Biochem J 279(Pt 1):16774. Tissue Int 73:38086.
23. Dew G, Murphy G, Stanton H, Vallon R, Angel P, 38. Everts V, Korper W, Jansen DC, Steinfort J, Lammerse
Reynolds JJ, et al. (2000) Localisation of matrix metal- I, Heera S, et al. (1999) Functional heterogeneity of
loproteinases and TIMP-2 in resorbing mouse bone. osteoclasts: matrix metalloproteinases participate in
Cell Tissue Res 299:38594. osteoclastic resorption of calvarial bone but not in
24. Dhanaraj V, Ye QZ, Johnson LL, Hupe DJ, Ortwine DF, resorption of long bone. Faseb J 13:121930.
Dunbar JB, Jr., et al. (1996) X-ray structure of a hydrox- 39. Faccio R, Grano M, Colucci S, Zallone AZ, Quaranta V,
amate inhibitor complex of stromelysin catalytic Pelletier AJ (1998) Activation of alphav beta3 integrin
domain and its comparison with members of the zinc on human osteoclast-like cells stimulates adhesion
metalloproteinase superfamily. Structure 4:37586. and migration in response to osteopontin. Biochem
25. Diaz A, Willis AC, Sim RB (2000) Expression of the pro- Biophys Res Commun 249:5225.
teinase specialized in bone resorption, cathepsin K, in 40. Fosang AJ, Last K, Knauper V, Neame PJ, Murphy G,
granulomatous inammation. Mol Med 6:64859. Hardingham TE, et al. (1993) Fibroblast and neutrophil
26. Diaz A, Willis AC, Sim RB (2002) Cathepsin K expres- collagenases cleave at two sites in the cartilage aggre-
sion in epithelioid and multinucleated giant cells. J can interglobular domain. Biochem J 295(Pt 1):2736.
Pathol 197:690; author reply 691. 41. Fuller K, Chambers TJ (1995) Localisation of mRNA
27. Dodds RA, James IE, Rieman D, Ahern R, Hwang SM, for collagenase in osteocytic, bone surface and chon-
Connor JR, et al. (2001) Human osteoclast cathepsin K drocytic cells but not osteoclasts. J Cell Sci 108(Pt 6):
is processed intracellularly prior to attachment and 222130.
bone resorption. J Bone Miner Res 16:47886. 42. Furuyama N, Fujisawa Y (2000) Distinct roles of
28. Drake FH, Dodds RA, James IE, Connor JR, Debouck cathepsin K and cathepsin L in osteoclastic bone
C, Richardson S, et al. (1996) Cathepsin K, but not resorption. Endocr Res 26:189204.
cathepsins B, L, or S is abundantly expressed in human 43. Garnero P, Borel O, Byrjalsen I, Ferreras M, Drake FH,
osteoclasts. Journal of Biological Chemistry 271: McQueney MS, et al. (1998) The collagenolytic activity
1251116. of cathepsin K is unique among mammalian pro-
29. Ducsay CA, Buhi WC, Bazer FW, Roberts RM, Combs teinases. J Biol Chem 273:3234752.
GE (1984) Role of uteroferrin in placental iron trans- 44. Gelb BD, Shi GP, Chapman HA, Desnick RJ (1996) Pyc-
port: effect of maternal iron treatment on fetal iron nodysostosis, a lysosomal disease caused by cathepsin
and uteroferrin content and neonatal hemoglobin. J K deciency. Science 273:12368.
Anim Sci 59:13038. 45. Gomis-Ruth FX, Maskos K, Betz M, Bergner A, Huber
30. Ek-Rylander B, Flores M, Wendel M, Heinegard D, R, Suzuki K, et al. (1997) Mechanism of inhibition of
Andersson G (1994) Dephosphorylation of osteopon- the human matrix metalloproteinase stromelysin-1 by
tin and bone sialoprotein by osteoclastic tartrate- TIMP-1. Nature 389:7781.
resistant acid phosphatase. Modulation of osteoclast 46. Goto T, Kiyoshima T, Moroi R, Tsukuba T, Nishimura
adhesion in vitro. J Biol Chem 269:148536. Y, Himeno M, et al. (1994) Localization of cathepsins
31. Elkins PA, Ho YS, Smith WW, Janson CA, DAlessio B, D, and L in the rat osteoclast by immuno-light and
KJ, McQueney MS, et al. (2002) Structure of the C- -electron microscopy. Histochemistry 101:3340.
terminally truncated human ProMMP9, a gelatin- 47. Goto T, Tsukuba T, Ayasaka N, Yamamoto K, Tanaka T
binding matrix metalloproteinase. Acta Crystallogr D (1992) Immunocytochemical localization of cathepsin
Biol Crystallogr 58:118292. D in the rat osteoclast. Histochemistry 97:1318.
32. Engsig MT, Chen QJ, Vu TH, Pedersen AC, Therkidsen 48. Gowen M, Lazner F, Dodds R, Kapadia R, Feild J,
B, Lund LR, et al. (2000) Matrix metalloproteinase 9 Tavaria M, et al. (1999) Cathepsin K knockout mice
and vascular endothelial growth factor are essential develop osteopetrosis due to a decit in matrix degra-
for osteoclast recruitment into developing long bones. dation but not demineralization. J Bone Miner Res 14:
J Cell Biol 151:87989. 165463.
33. Etherington DJ, Birkedahl-Hansen H (1987) The inu- 49. Haeckel C, Krueger S, Buehling F, Broemme D, Franke
ence of dissolved calcium salts on the degradation of K, Schuetze A, et al. (1999) Expression of cathepsin K
hard-tissue collagens by lysosomal cathepsins. Coll in the human embryo and fetus. Dev Dyn 216:8995.
Relat Res 7:18599. 50. Haeckel C, Krueger S, Kuester D, Ostertag H, Samii M,
34. Everts V, Delaisse JM, Korper W, Beertsen W (1998) Buehling F, et al. (2000) Expression of cathepsin K in
Cysteine proteinases and matrix metalloproteinases chordoma. Hum Pathol 31:83440.
Osteolytic Enzymes of Osteoclasts 31

51. Hansen T, Unger RE, Gaumann A, Hundorf I, Maurer induced osteoblast elongation regulates osteoclastic
J, Kirkpatrick CJ, et al. (2001) Expression of matrix- bone resorption through a p38 mitogen-activated
degrading cysteine proteinase cathepsin K in protein kinase- and matrix metalloproteinase-
cholesteatoma. Mod Pathol 14:122631. dependent pathway. J Biol Chem 276:393508.
52. Hayman AR, Cox TM (1994) Purple acid phosphatase 67. Khosla S (2001) Minireview: the OPG/RANKL/RANK
of the human macrophage and osteoclast. Characteri- system. Endocrinology 142:50505.
zation, molecular properties, and crystallization of 68. Konttinen YT, Mandelin J, Li TF, Salo J, Lassus J,
the recombinant di-iron-oxo protein secreted by Liljestrom M, et al. (2002) Acidic cysteine endopro-
baculovirus-infected insect cells. J Biol Chem teinase cathepsin K in the degeneration of the super-
269:1294300. cial articular hyaline cartilage in osteoarthritis.
53. Hill PA, Buttle DJ, Jones SJ, Boyde A, Murata M, Arthritis Rheum 46:95360.
Reynolds JJ, et al. (1994) Inhibition of bone resorption 69. Kremer M, Judd J, Rifkin B, Auszmann J, Oursler MJ
by selective inactivators of cysteine proteinases. J Cell (1995) Estrogen Modulation of Osteoclast Lysosomal
Biochem 56:11830. Enzyme Secretion. J Cell Biochem 57:27179.
54. Hill PA, Murphy G, Docherty AJ, Hembry RM, Millican 70. Kumta SM, Huang L, Cheng YY, Chow LT, Lee KM,
TA, Reynolds JJ, et al. (1994) The effects of selective Zheng MH (2003) Expression of VEGF and MMP-9 in
inhibitors of matrix metalloproteinases (MMPs) on giant cell tumor of bone and other osteolytic lesions.
bone resorption and the identication of MMPs and Life Sci 73:142736.
TIMP-1 in isolated osteoclasts. J Cell Sci 107(Pt 11): 71. Lark MW, Stroup GB, James IE, Dodds RA, Hwang SM,
305564. Blake SM, et al. (2002) A potent small molecule, non-
55. Hollberg K, Hultenby K, Hayman A, Cox T, Andersson peptide inhibitor of cathepsin K (SB 331750) prevents
G (2002) Osteoclasts from mice decient in tartrate- bone matrix resorption in the ovariectomized rat.
resistant acid phosphatase have altered rufed borders Bone 30:74653.
and disturbed intracellular vesicular transport. Exp 72. Lerner UH, Grubb A (1992) Human cystatin C, a cys-
Cell Res 279:22738. teine proteinase inhibitor, inhibits bone resorption in
56. Holliday LS, Welgus HG, Fliszar CJ,Veith GM, Jeffrey JJ, vitro stimulated by parathyroid hormone and parathy-
Gluck SL (1997) Initiation of osteoclast bone resorp- roid hormone-related peptide of malignancy. J Bone
tion by interstitial collagenase. J Biol Chem 272: Miner Res 7:43340.
220538. 73. Li CY,Yam LT, Lam KW (1970) Acid phosphatase isoen-
57. Horton MA, Nesbit MA, Helfrich MH (1995) Interac- zyme in human leukocytes in normal and pathologic
tion of osteopontin with osteoclast integrins. Ann N Y conditions. J Histochem Cytochem 18:47381.
Acad Sci 760:190200. 74. Li YP, Chen W (1999) Characterization of mouse
58. Hou WS, Li Z, Buttner FH, Bartnik E, Bromme D (2003) cathepsin K gene, the gene promoter, and the gene
Cleavage site specicity of cathepsin K toward carti- expression. J Bone Miner Res 14:48799.
lage proteoglycans and protease complex formation. 75. Li Z, Hou WS, Bromme D (2000) Collagenolytic activ-
Biol Chem 384:8917. ity of cathepsin K is specically modulated by carti-
59. Hultenby K, Reinholt FP, Heinegard D, Andersson G, lage-resident chondroitin sulfates. Biochemistry 39:
Marks SC, Jr. (1995) Osteopontin: a ligand for the 52936.
alpha v beta 3 integrin of the osteoclast clear zone 76. Li Z, Hou WS, Escalante-Torres CR, Gelb BD, Bromme
in osteopetrotic (ia/ia) rats. Ann N Y Acad Sci 760: D (2002) Collagenase activity of cathepsin K depends
31518. on complex formation with chondroitin sulfate. J Biol
60. Igarashi Y, Lee MY, Matsuzaki S (2002) Acid phos- Chem 277:2866976.
phatases as markers of bone metabolism. J Chro- 77. Lin SK, Kok SH, Kuo MY, Wang TJ, Wang JT, Yeh FT, et
matogr B Analyt Technol Biomed Life Sci 781:34558. al. (2002) Sequential expressions of MMP-1, TIMP-1,
61. Inaoka T, Bilbe G, Ishibashi O, Tezuka K, Kumegawa M, IL-6, and COX-2 genes in induced periapical lesions in
Kokubo T (1995) Molecular cloning of human cDNA rats. Eur J Oral Sci 110:24653.
for cathepsin K: novel cysteine proteinase predomi- 78. Littlewood-Evans AJ, Bilbe G, Bowler WB, Farley D,
nantly expressed in bone. Biochem Biophys Res Wlodarski B, Kokubo T, et al. (1997) The osteoclast-
Commun 206:8996. associated protease cathepsin K is expressed in human
62. Inui T, Ishibashi O, Inaoka T, Origane Y, Kumegawa M, breast carcinoma. Cancer Res 57:538690.
Kokubo T, et al. (1997) Cathepsin K antisense oligo- 79. Luchin A, Suchting S, Merson T, Rosol TJ, Hume DA,
deoxynucleotide inhibits osteoclastic bone resorption. Cassady AI, et al. (2001) Genetic and physical interac-
J Biol Chem 272:810912. tions between Microphthalmia transcription factor
63. Irie K, Tsuruga E, Sakakura Y, Muto T, Yajima T (2001) and PU.1 are necessary for osteoclast gene expression
Immunohistochemical localization of membrane type and differentiation. J Biol Chem 276:3670310.
1-matrix metalloproteinase (MT1-MMP) in osteoclasts 80. Martignetti JA, Aqeel AA, Sewairi WA, Boumah CE,
in vivo. Tissue Cell 33:47882. Kambouris M, Mayouf SA, et al. (2001) Mutation of the
64. James IE, Marquis RW, Blake SM, Hwang SM, Gress CJ, matrix metalloproteinase 2 gene (MMP2) causes a
Ru Y, et al. (2001) Potent and selective cathepsin L multicentric osteolysis and arthritis syndrome. Nat
inhibitors do not inhibit human osteoclast resorption Genet 28:2615.
in vitro. J Biol Chem 276:1150711. 81. Migita K, Eguchi K, Tominaga M, Origuchi T, Kawabe
65. Kaenah W, Buttle DJ, Burnett D, Hollander AP (1998) Y, Nagataki S (1997) Beta 2-microglobulin induces
Cleavage of native type I collagen by human neutrophil stromelysin production by human synovial broblasts.
elastase. Biochem J 330(Pt 2):897902. Biochem Biophys Res Commun 239:6215.
66. Karsdal MA, Fjording MS, Foged NT, Delaisse JM, 82. Miyauchi A, Alvarez J, Greeneld EM, Teti A, Grano M,
Lochter A (2001) Transforming growth factor-beta- Colucci S, et al. (1993) Binding of osteopontin to the
32 Bone Resorption

osteoclast integrin alpha v beta 3. Osteoporos Int. 3 98. Saftig P, Hunziker E, Wehmeyer O, Jones S, Boyde A,
Suppl 1:1325. Rommerskirch W, et al. (1998) Impaired osteoclastic
83. Muir P, Hayashi K, Manley PA, Colopy SA, Hao Z (2002) bone resorption leads to osteopetrosis in cathepsin-K-
Evaluation of tartrate-resistant acid phosphatase and decient mice. Proc Natl Acad Sci USA 95:134538.
cathepsin K in ruptured cranial cruciate ligaments in 99. Sasaki T, Ueno-Matsuda E (1993) Cysteine-proteinase
dogs. Am J Vet Res 63:127984. localization in osteoclasts: an immunocytochemical
84. Nakase T, Takeuchi E, Sugamoto K, Kaneko M, Tomita study. Cell Tissue Res 271:1779.
T, Myoui A, et al. (2000) Involvement of multinucleated 100. Sato T, del Carmen Ovejero M, Hou P, Heegaard AM,
giant cells synthesizing cathepsin K in calcied ten- Kumegawa M, Foged NT, et al. (1997) Identication of
dinitis of the rotator cuff tendons. Rheumatology the membrane-type matrix metalloproteinase MT1-
(Oxford) 39:10747. MMP in osteoclasts. J Cell Sci 110(Pt 5):58996.
85. Noda M, Oh J, Takahashi R, Kondo S, Kitayama H, 101. Schlondorff J, Lum L, Blobel CP (2001) Biochemical
Takahashi C (2003) RECK: a novel suppressor of malig- and pharmacological criteria dene two shedding
nancy linking oncogenic signaling to extracellular activities for TRANCE/OPGL that are distinct from the
matrix remodeling. Cancer Metastasis Rev 22:16775. tumor necrosis factor alpha convertase. J Biol Chem
86. Oh J, Takahashi R, Kondo S, Mizoguchi A, Adachi E, 276:1466574.
Sasahara RM, et al. (2001) The membrane-anchored 102. Shimizu H, Sakamoto M, Sakamoto S (1990) Bone
MMP inhibitor RECK is a key regulator of extracellu- resorption by isolated osteoclasts in living versus devi-
lar matrix integrity and angiogenesis. Cell 107:789 talized bone: differences in mode and extent and the
800. effects of human recombinant tissue inhibitor of met-
87. Ohsawa Y, Nitatori T, Higuchi S, Kominami E, alloproteinases. J Bone Miner Res 5:41118.
Uchiyama Y (1993) Lysosomal cysteine and aspartic 103. Sodek J, Overall CM (1992) Matrix metalloproteinases
proteinases, acid phosphatase, and an endogenous cys- in periodontal tissue remodelling. Matrix Suppl 1:352
teine proteinase inhibitor, cystatin-beta, in rat osteo- 62.
clasts. J Histochem Cytochem 41:107583. 104. Somerville RP, Oblander SA, Apte SS (2003) Matrix
88. Ohuchi E, Imai K, Fujii Y, Sato H, Seiki M, Okada Y metalloproteinases: old dogs with new tricks. Genome
(1997) Membrane type 1 matrix metalloproteinase Biol 4:216.
digests interstitial collagens and other extracellular 105. Stamenkovic I (2003) Extracellular matrix remodel-
matrix macromolecules. J Biol Chem 272:244651. ling: the role of matrix metalloproteinases. J Pathol
89. Ortega N, Behonick D, Stickens D, Werb Z (2003) How 200:44864.
proteases regulate bone morphogenesis. Ann N Y Acad 106. Stroup GB, Lark MW, Veber DF, Bhattacharyya A, Blake
Sci 995:10916. S, Dare LC, et al. (2001) Potent and selective inhibition
90. Page AE, Warburton MJ, Chambers TJ, Pringle JA, of human cathepsin K leads to inhibition of bone
Hayman AR (1992) Human osteoclastomas contain resorption in vivo in a nonhuman primate. J Bone
multiple forms of cathepsin B. Biochim Biophys Acta Miner Res 16:173946.
1116:5766. 107. Tepel C, Bromme D, Herzog V, Brix K (2000) Cathepsin
91. Punturieri A, Filippov S, Allen E, Caras I, Murray R, K in thyroid epithelial cells: sequence, localization and
Reddy V, et al. (2000) Regulation of elastinolytic possible function in extracellular proteolysis of thy-
cysteine proteinase activity in normal and cathepsin roglobulin. J Cell Sci 113 Pt 24:448798.
K-decient human macrophages. J Exp Med 192:789 108. Tezuka K, Nemoto K, Tezuka Y, Sato T, Ikeda Y, Kobori
99. M, et al. (1994) Identication of matrix metallopro-
92. Rantakokko J, Aro HT, Savontaus M, Vuorio E (1996) teinase 9 in rabbit osteoclasts. J Biol Chem 269:15006
Mouse cathepsin K: cDNA cloning and predominant 9.
expression of the gene in osteoclasts, and in some 109. Tezuka K, Tezuka Y, Maejima A, Sato T, Nemoto K,
hypertrophying chondrocytes during mouse develop- Kamioka H, et al. (1994) Molecular cloning of a possi-
ment. FEBS Lett 393:30713. ble cysteine proteinase predominantly expressed in
93. Razzouk S, Brunn JC, Qin C, Tye CE, Goldberg HA, osteoclasts. J Biol Chem 269:11069.
Butler WT (2002) Osteopontin posttranslational mod- 110. Thompson SK, Halbert SM, Bossard MJ, Tomaszek
ications, possibly phosphorylation, are required for in TA, Levy MA, Zhao B, et al. (1997) Design of potent
vitro bone resorption but not osteoclast adhesion. and selective human cathepsin K inhibitors that
Bone 30:407. span the active site. Proc Natl Acad Sci USA 94:14249
94. Reynolds JJ (1970) Degradation processes in bone and 54.
cartilage. Calcif Tissue Res Suppl 1:5256. 111. Uppenberg J, Lindqvist F, Svensson C, Ek-Rylander B,
95. Rifkin BR, Vernillo AT, Kleckner AP, Auszmann JM, Andersson G (1999) Crystal structure of a mammalian
Rosenberg LR, Zimmerman M (1991) Cathepsin B and purple acid phosphatase. J Mol Biol 290:20111.
L activities in isolated osteoclasts. Biochem Biophys 112. Vaes G (1988) Cellular biology and biochemical mech-
Res Commun 179:6369. anism of bone resorption. A review of recent develop-
96. Ross FP, Chappel J, Alvarez JI, Sander D, Butler WT, ments on the formation, activation, and mode of action
Farach-Carson MC, et al. (1993) Interactions between of osteoclasts. Clin Orthop 23971.
the bone matrix proteins osteopontin and bone sialo- 113. Vaes G, Delaisse JM, Eeckhout Y (1992) Relative roles
protein and the osteoclast integrin alpha v beta 3 of collagenase and lysosomal cysteine-proteinases in
potentiate bone resorption. J Biol Chem 268:99017. bone resorption. Matrix Suppl 1:3838.
97. Saftig P, Hunziker E, Everts V, Jones S, Boyde A, 114. Vu TH, Shipley JM, Bergers G, Berger JE, Helms JA,
Wehmeyer O, et al. (2000) Functions of cathepsin K in Hanahan D, et al. (1998) MMP-9/gelatinase B is a key
bone resorption. Lessons from cathepsin K decient regulator of growth plate angiogenesis and apoptosis
mice. Adv Exp Med Biol 477:293303. of hypertrophic chondrocytes. Cell 93:41122.
Osteolytic Enzymes of Osteoclasts 33

115. Walsh NC, Cahill M, Carninci P, Kawai J, Okazaki Y, and tumorigenic cells. Proc Natl Acad Sci USA 84:
Hayashizaki Y, et al. (2003) Multiple tissue-specic 67259.
promoters control expression of the murine tartrate- 117. Yamashita DS, Dodds RA (2000) Cathepsin K and the
resistant acid phosphatase gene. Gene 307:11123. design of inhibitors of cathepsin K. Curr Pharm Des
116. Wilhelm SM, Collier IE, Kronberger A, Eisen AZ, 6:124.
Marmer BL, Grant GA, et al. (1987) Human skin 118. Zaidi M, Troen B, Moonga BS, Abe E (2001) Cathepsin
broblast stromelysin: structure, glycosylation, sub- K, osteoclastic resorption, and osteoporosis therapy.
strate specicity, and differential expression in normal J Bone Miner Res 16:17479.
3.
Regulation of Osteoclast Activity
Roland Baron and William C. Horne

The activation of RANK by RANKL is required


Introduction for osteoclast differentiation and also regulates
osteoclast activity, but the stimulation of
The maintenance of normal bone mass during myelomonocytic precursors also requires M-
adult life depends on a balance between CSF to induce the formation of osteoclasts.
osteoblastic bone formation and osteoclastic [138, 161].
bone destruction. Bone resorption is primarily Because Chapter 1 is devoted to the regula-
carried out by osteoclasts. The overall rate of tion of osteoclast differentiation, the aim of this
osteoclastic bone resorption is regulated at two chapter is to focus specically on the regulation
main levels: 1) determining the number of of osteoclast activity.
osteoclasts through the regulation of the osteo-
clast precursor pool and their rate of differenti-
ation, i.e., regulating osteoclastogenesis (see
Chapter 1); and 2) determining the bone resorb- The Molecular Mechanisms
ing activity of individual osteoclasts through
the regulation of their key functional features.
of Bone Resorption:
In both cases, it now is apparent that activa- Identification of the
tion of RANK (receptor activator of NFkB) sig-
naling by its extracellular ligand RANKL
Main Effectors
(RANK ligand) plays a central role, as does
osteoprotegerin (OPG), a decoy RANK. Indeed, The understanding of the mechanisms by which
many of the previously known regulators of the activity of the osteoclast is regulated re-
bone resorption, such as parathyroid hormone quires a clear comprehension of the mecha-
(PTH), 1,25-(OH)2D3, or several interleukins, nisms of bone resorption, thereby identifying
affect osteoclast differentiation and/or function the various levers through which the cell can
through their impact on RANKL or OPG pro- effectively be activated or inhibited. In other
duction by stromal cells or osteoblasts and not, words, we need to understand the effectors of
as initially thought, by a direct action on the bone resorption in order to understand the
osteoclast or its precursors [16]. RANKL binds mechanisms by which they are regulated.
to its receptor RANK, which is expressed at the The main functional features (Figure 3.1) that
surface of osteoclast progenitors as well as dene the activity of an osteoclast are 1) its
in mature osteoclasts, and activates several adhesion to the bone surface; 2) its ability to
intracellular signaling pathways. The specic synthesize and directionally secrete enzymes;
individual role of each is currently being unrav- 3) its ability to acidify the subosteoclastic bone
eled. OPG modulates RANK activation by resorbing compartment; 4) its ability efciently
competing with RANK for binding to RANKL. to internalize the material generated by the
34
Regulation of Osteoclast Activity 35

Acidification
Gene Expression

Vesicular Traffic

Carbonic
Internalization
Anhydrase II Adhesion and Migration

HCO3 HCO3+ H+ clear


Cl Cl H+
Lysosomes zone
ClC-7 Secretion

integrins integrins
H+ ATPase
Cl
Cl H+
Secreted Enzymes
+
H+ Cl H Cathepsin K
MMP-9
H+ TRAP

Bone Matrix
B

Figure 3.1 (A) The osteoclast contains multiple nuclei, an endoplasmic reticulum where lysosomal enzymes are synthesized, and
prominent Golgi stacks around each nucleus. The cell is attached to bone matrix (bottom) and forms a separate compartment under-
neath itself, limited by the sealing zone. The plasma membrane of the cell facing this compartment is extensively folded and forms
the ruffled border. Vesicles in the cytoplasm transport enzymes toward the bone matrix and internalize partially digested bone
matrix. (B) Schematic representation of a polarized active osteoclast including the main molecular effectors of the process of bone
resorption (see text)
36 Bone Resorption

Figure 3.2 Steps in the attachment, resorption, and migration of the osteoclast: dynamics of podosomes: podosomes, or points
of adhesion, are turning over every 26 minutes, allowing plasticity of the cell attachment and cell migration. A dense ring of
podosomes, or reorganized actin, forms the sealing zone around resorption pits.

extracellular degradation of the bone matrix;


and 5) its ability to migrate efciently toward
and along the bone surfaces.
Adhesion and migration depend essentially
on adhesion receptors and their ability to regu-
late the cytoskeleton, particularly the rapid
assembly and disassembly of cytoskeletal pro-
teins at sites of adhesion (podosomes, Figure
3.2). Secretion depends essentially on vesicular
transport, the directional ssion-fusion of
transport vesicles from the endoplasmic reticu-
lum to the Golgi and from the Golgi to the secre-
tory pole of the cell, i.e. the rufed border
(Figure 3.3). Internalization also depends upon
vesicular transport, but in a retrograde manner, Figure 3.3 The main vesicular traffic pathways in an osteo-
i.e., from the rufed border to endosomes and clast: secretion via exocytosis and internalization via endocyto-
lysosomes or through the cell to the basal pole sis and transcytosis are effected by different membrane
in transcytosis (Figure 3.3). Finally, acidica- domains.
tion depends essentially on the generation of
protons by the enzyme carbonic anhydrase and
their transport across the plasma membrane by
the apical vacuolar proton ATPase (V-ATPase), vitronectin receptor knockout mice [116], Src
in parallel with the extrusion of chloride ions and Pyk2 [143] as well as gelsolin [25] -decient
through the ClC-7 chloride channel as well as mice. Altered synthesis of key enzymes such as
homeostatic ion transport at the basolateral cathepsin K lead to osteopetrosis in mice [140]
membrane (Figure 3.4). and occur in humans aficted by pycnodysos-
The importance of these ve key functional tosis [57].A defect in vesicular trafc is the most
effectors (Figure 3.1B) in the regulation of likely cause of osteopetrosis in the gray-lethal
osteoclastic activity is readily demonstrated by mutation mouse [20, 139]. Finally, mutations in
the fact that specic genetic alterations of these the V-ATPase [54, 92, 105, 149], the ClC-7 chlo-
molecules or pathways all lead to altered bone ride channel [91], or the enzyme carbonic anhy-
resorption and osteopetrosis. Defects in adhe- drase II [159], essential components of the
sion receptor signaling or cytoskeletal proteins acidication process, lead to osteopetrosis in
and their associated tyrosine kinases, affecting mice and in humans. These examples illustrate
cell adhesion and cell migration, are observed in unequivocally the importance of these various
Regulation of Osteoclast Activity 37

Figure 3.4 The osteoclast ion transport


systems in the apical ruffled-border and the
basolateral membranes allow efficient extrusion
of protons via the ruffled-border vacuolar ATPase
in parallel with a chloride conductance. Basolat-
eral transport systems avoid changes in pH
and/or in membrane polarization of the cell. Car-
bonic anhydrase generates protons and mito-
chondriae generate ATP.

pathways in the regulation of the activity of the bone resorption. The attachment of the cell to
osteoclast, thereby dening and validating novel the matrix is performed via integrin receptors
targets for therapeutic intervention. Genetic (mostly avb3, avb5, and a2b1), which bind to spe-
mutations aside from those affecting osteoclast cic sequences in matrix proteins, and activate
activity decrease bone resporption, whether in a specic signaling cascade requiring several
humans or in mutant or genetically altered now identied signaling molecules to ensure
mice, through alterations in the differentiation adhesion and cell motility (c-Src, Pyk2, Cbl,
of osteoclast lineage rather than through mod- Gelsolin). The plasma membrane in the rufed
ulation of the activity of the mature osteoclast. border area contains proteins that also are
These key functional effectors dene the found at the limiting membrane of endosomes
most prominent morphological features of the and related organelles, and a specic type of
osteoclast (Figure 3.1). Besides its multinucle- electrogenic proton ATPase as well as a specic
ation, these are deep foldings of the plasma chloride channel (ClC-7) responsible for acidi-
membrane in the area facing the bone matrix, cation of the extracellular bone resorbing
i.e., the rufed border, which is formed as compartment. The basolateral plasma mem-
a result of directional insertion of vesicles brane (Figures 3.4 and 3.5) of the osteoclast is
required for active secretion of protons and highly and specically enriched in Na,K+-
lysosomal enzymes toward the bone surface, as ATPase (sodium-potassium pump), HCO3/Cl-
well as directional endocytic activity from the exchangers, and Na+/H+ exchangers, as well as
bone resorbing compartment [8, 9, 181]. This several ion channels. This membrane also
central area of the interface between the cell and expresses RANK, the receptor for RANK ligand,
the bone surface is surrounded by a ring of and the M-CSF receptor, both of which are
contractile proteins (sealing zone) that serve responsible for osteoclast differentiation, as well
to attach the cell to the bone surface while as the calcitonin receptor, which is capable of
ensuring its continued ability to migrate. This rapidly inactivating the osteoclast.
attachment ring seals off the subosteoclastic Lysosomal enzymes (for example, tartrate
bone-resorbing compartment, sandwiched be- resistant acid phosphatase, cathepsin K) are
tween the rufed border plasma membrane actively synthesized by the osteoclast and are
and the surface of the bone matrix undergoing found in the endoplasmic reticulum, as well as
38 Bone Resorption

Figure 3.5 The main receptors regulating osteoclast activity target the cytoskeleton, affecting adhesion and migration, vesicular
traffic, secretion and internalization, ion transport, acidification, or on a longer-term basis gene expression, affecting all aspects
of osteoclast function.

in the Golgi apparatus. The osteoclast typically process that involves the transporting activity of
contains multiple copies of the Golgi complex, vacuolar proton pumps [12, 180]. Recent genetic
organized as rings surrounding each nucleus evidence demonstrates that the electrogenic
[8], and many readily identiable transport proton-pump ATPase is related to, but different
vesicles moving directionally toward the rufed from, the pump of kidney-tubule-acidifying
border membrane [8, 9]. The enzymes are cells or lysosomes [54, 105, 149]. The protons are
secreted, via the rufed border, into the extra- provided to the pumps by carbonic anhydrase,
cellular bone-resorbing compartment where an enzyme that is highly concentrated in the
they reach a sufciently high extracellular con- cytosol of the osteoclast. ATP and CO2 are pro-
centration that allows extracellular degradation vided by the mitochondria, which are charac-
of the bone matrix, within the sealed compart- teristically found in very high numbers in the
ment. The transport and targeting of these osteoclast. Apical chloride channels (ClC-7) are
secreted enzymes at the apical pole of the osteo- also found in the rufed border membrane [92],
clast involves mannose-6-phosphate receptors where they serve to prevent the hyperpolariza-
[8]. Furthermore, the cell secretes several tion created by the massive extrusion of posi-
metalloproteases, including collagenase and tively charged protons by the V-ATPase. The
gelatinase [37]. basolateral membrane actively exchanges bicar-
The osteoclast acidies the extracellular bonate for chloride, thereby avoiding an alka-
compartment [9] by secreting protons across linization of the cytosol and providing chloride
the rufed-border membrane (Figure 3.4), a for the rufed-border channels [67, 185]. The
Regulation of Osteoclast Activity 39

basolateral sodium pumps [10] might be upon migration of the osteoclast away from the
involved in secondary active transport of resorption lacuna, and may play an essential
calcium and/or protons in association with a role in the coupling to bone formation during
Na+/Ca2+ exchanger and/or a Na+/H+ antiport. the remodeling cycle.
This cell thus appears to function in a manner The various components of the osteoclast
similar to that of kidney tubule or gastric pari- bone resorbing machinery are therefore the
etal cells, which also acidify their respective levers (Figure 3.1B) by which the endocrine,
lumens. paracrine, or endocrine ligands found in the
The extracellular bone-resorbing compart- osteoclast microenvironment exert their posi-
ment is the functional equivalent of a lysosome, tive or negative regulatory action on the bone-
with (i) a low pH, (ii) lysosomal enzymes, and resorbing capacity of individual osteoclasts.
(iii) the substrate, i.e. the bone matrix. First, the Indeed, some molecules such as RANKL, OPG,
hydroxyapatite crystals are mobilized by diges- and M-CSF inuence both differentiation and
tion of their link to collagen (the noncollage- activity of osteoclasts, whereas others, such as
nous proteins) and dissolved by the acid calcitonin or integrin ligands, appear to regulate
environment. Then, the residual collagen bers only osteoclast activity.
are digested by the specic action of cathepsin
K at low pH [13], and possibly by the activation
of latent collagenase [37, 190]. During active Regulation of Osteoclast
resorption, the degraded bone matrix is
processed extracellularly, after which some Activity and Bone Resorption
products are endocytosed into the osteoclast
and degraded within secondary lysosomes, and Integrin Signaling (Figure 3.6)
others are transported through the cell by trans-
cytosis and secreted through the basal mem- Integrins are a superfamily of heterodimeric (a
brane [130, 141]. The degraded products from and b) transmembrane receptors which medi-
the bone matrix probably also are released into ate cell-matrix interactions. Integrin-mediated
the local microenvironment during periods of adhesion and signaling regulate a variety of cell
relapse of the sealing zone, possibly induced by processes [30, 146]. In addition to cell adhesion,
a calcium sensor that responds to the rise of integrin receptor assembly also organizes extra-
extracellular calcium in the bone-resorbing cellular matrices, modulates cell shape changes,
compartment (mM). Furthermore, degradation and participates in cell spreading and motility
products are left behind at the bone surface [186, 189].

Outside-inavb3mediated Inside-out Signaling by Cbl


signaling
A B Vn
Vn
Figure 3.6 Integrin signaling con- (-)
trols adhesion and migration: (A) av b3 av b3
Binding of the vitronectin receptor to
extracellular matrix proteins activates
calcium entry into the osteoclast. This
Ca2+ (-)
leads to Pyk2 autophosphorylation, pY402 Pyk2
recruitment of Src and Cbl to (+) (+)
pY402 Pyk2
podosomes and Cbl (as well as several
cytoskeletal proteins) becoming phos- Src SH3 SH2 K
phorylated on tyrosine residues. (B) pY416 Src SH3 SH2 K
Phosphorylation of Cbl leads to Src (+) pY416 (-)
kinase down-regulation and to loss of Cbl PPP PTB
integrin adhesion to the substratum, P Cbl PPP PTB
allowing cell migration. P
40 Bone Resorption

Upon ligand binding, integrins undergo Additional in vivo studies demonstrated that
receptor clustering, leading to the formation of echistatin or RGD-peptidomimetics inhibit
adhesion contacts where these receptors are bone resorption in ovariectomized rodents by
linked to intracellular cytoskeletal complexes, blocking the function of avb3 integrins [47, 194].
including bundles of actin laments and signal- Moreover, infusion of anti-rat b3 integrin
ing molecules. Indeed, the short cytoplasmic subunit antibody blocked the effect of PTH on
domains of the a and b integrin subunits can serum calcium in TPTX rats [31]. Further com-
recruit a variety of cytoskeletal and signaling pelling evidence for the role of avb3 integrin in
molecules. Integrin-mediated signaling changes osteoclast function was recently provided by the
phosphoinositide metabolism, raises intra- targeted disruption of b3 integrin subunit in
cellular calcium, and induces tyrosine or serine mice, which induces late onset osteopetrosis, 3
phosphorylation of signaling molecules [58, to 6 months after birth [116].
146]. Recognition of extracellular matrix compo-
Adhesion of osteoclasts to the bone surface nents by osteoclasts is an important step in ini-
involves the interaction of integrins with ex- tiating osteoclast function. Several studies have
tracellular matrix proteins within bone. Bone demonstrated that integrin-mediated cell adhe-
consists mainly of type I collagen (>90%) and sion to vitronectin, bronectin or collagen
of noncollagenous proteins interacting with a induces cell spreading and actin rearrangement
mineral phase of hydroxyapatite. Osteoclasts in osteoclasts. avb3 integrin plays a role in the
express very high levels of the integrin avb3 adhesion and spreading of osteoclasts on bone
[75]. Mammalian osteoclasts also express lower [97, 129]. Expression of avb3 integrin is detected
levels of the collagen/laminin receptor a2b1 and in mononucleated osteoclast precursors, where
the vitronectin/bronectin receptor avb1. Rat it mediates the adhesion and migration neces-
osteoclasts adhere in an avb3-dependent sary for their fusion during osteoclast differen-
manner to extracellular matrix proteins con- tiation in culture [126]. Furthermore, avb3
taining arginine-glycine-aspartic acid (RGD) integrin plays a role in the regulation of two
sequences, including vitronectin, osteopontin, processes required for effective osteoclastic
bone sialoprotein, and a cryptic RGD-site in bone resorption: cell migration and mainte-
denatured collagen type I [51]. More recently, nance of the sealing zone [123].
it was reported that rat osteoclasts adhere to Targeted disruption of b3 integrin subunit
native collagen type I using a2b1 integrin, sur- results in an osteopetrotic phenotype [116].
prisingly in an RGD-dependent manner [72]. Osteoclasts isolated from these mice fail to
Moreover, osteoclastic bone resorption is spread, do not form actin rings, have abnormal
partially inhibited by both anti-a2 and anti-b1 rufed membranes, and exhibit reduced bone
antibodies [124]. The rst evidence that avb3 resorption activity in vitro. Thus, avb3 plays a
plays an important role in osteoclast function pivotal role in the resorptive process, through its
was obtained when a monoclonal antibody functions in cell adhesion-dependent signal
raised against osteoclasts inhibited bone transduction and in cell motility. Indeed, avb3
resorption in vitro [21]; the antigen was later transmits bone matrix-derived signals, ulti-
identied to be the avb3 integrin [35]. Further- mately activating intracellular events that regu-
more, osteoclastic bone resorption in vitro can late cytoskeletal organization essential for
be inhibited by RGD-containing peptides and osteoclast migration and polarization. Recent
disintegrins or by blocking antibodies to avb3 research interests have been focused on identi-
[76, 87, 144]. fying the hierarchy of the avb3-regulated
Inhibition of avb3 integrins can also block cascade of signaling and structural proteins
bone resorption in vivo. In the thyroparathy- required for this cytoskeletal organization. One
roidectomized (TPTX) model, co-infusion of of the earliest events initiated by integrin-
disintegrins such as echistatin or kinstrin and ligand engagement is elevation of intracellular
PTH completely blocked the PTH-induced calcium. In rat, mouse, and human osteoclasts,
increase in serum calcium [50, 87]. Echistatin RGD-containing peptides trigger a transient
was also shown to inhibit bone loss in hyper- increase in intracellular calcium that seems to
parathyroid mice maintained on a low calcium be mobilized from intracellular stores [135,
diet. In the bones of these animals, echistatin co- 205]. This event is independent of the presence
localizes with avb3 integrins in osteoclasts [111]. or activation of c-Src [143].
Regulation of Osteoclast Activity 41

surface of resorbing osteoclasts of avb3 integrins


A Role for Tyrosine Kinases and their downstream effectors, including Pyk2,
p130Cas and paxillin [96]. Thus, c-Src might
c-Src Regulates avb3 Integrin- not be important for the initial recruitment of
mediated Cytoskeletal Organization avb3-dependent downstream effectors, but may
mediate the turnover of the integrin-associated
and Cell Motility complex of signaling and cytoskeletal mole-
cules. Recently, this hypothesis has been sup-
Src kinases play important roles in cell adhesion ported strongly by the nding that indeed
and migration, in cell cycle control, and in cell Src-/- osteoclasts demonstrate signicant
proliferation and differentiation [175]. More- decreases in their ability to migrate over a sub-
over, novel roles for Src kinases in the control of strate, at least in vitro [143]. It is conceivable
cell survival and angiogenesis have recently therefore, that most of the decreased bone
emerged [147]. In bone, the tyrosine kinase c- resorption in Src-/- osteoclasts is attributable to
Src was found to be essential for osteoclast low cell motility.
polarization, resorbing activity, and/or motility.
Targeted disruption of c-Src in mice induces
osteopetrosis due to loss of osteoclast function, Pyk2 and c-Src Co-regulate avb3
and occurs without a reduction in osteoclast Integrin-mediated Signals
number [160]. Morphologically, osteoclasts in
Src-/- mice are not able to form rufed borders, Pyk2 was recently identied as a major adhe-
but do form sealing zones on bone surfaces [15]. sion-dependent tyrosine kinase in osteoclasts,
Src is highly expressed in osteoclasts, along with both in vivo and in vitro [42] (Figure 3.6). Pyk2
at least four other Src family members (Hck, is a member of the focal adhesion kinase (FAK)
Fyn, Lyn, and Yes) [74, 108]. Hck partly com- family that is highly expressed in cells of the
pensates for the absence of Src, since the double central nervous system and in haematopoietic
mutant Src-/-/Hck-/- mouse is more severely lineage [146]. Pyk2 and FAK share about 45%
osteopetrotic than the Src-/- mouse, despite the overall amino acid identity and have a high
fact that the single mutant Hck-/- mouse is not degree of sequence conservation surrounding
obviously osteopetrotic [108]. On the other binding sites of SH2- and SH3-domain-contain-
hand, transgenic expression of kinase-decient ing proteins. Although the presence of FAK has
Src in Src-/- mice partially rescued osteoclast been reported in osteoclasts [11, 168], Pyk2 is
function, indicating that full Src tyrosine kinase expressed at much higher levels than FAK in
activity is not absolutely required for mediating osteoclasts [42]. Ligation of avb3 integrins either
osteoclast function. Its role as an adaptor by ligand binding or by antibody-mediated clus-
protein to recruit downstream signaling mole- tering results in an increase in Pyk2 tyrosine
cules [148] is also essential to its function in phosphorylation [42]. Moreover, Pyk2 co-
osteoclasts [143]. localizes with F-actin, paxillin, and vinculin in
c-Src is associated with the plasma mem- podosomes and in sealing zones of resorbing
brane and multiple intracellular organelles [74, osteoclasts on bone [42]. These observations
167]. It is concentrated in the actin ring and the strongly implicate Pyk2 as a downstream effec-
cell periphery [143], regions that are involved in tor in the avb3-dependent signaling, mediating
osteoclast attachment and migration. This sug- the cytoskeletal organization associated with
gests that the absence of Src might compromise osteoclast adhesion, spreading, migration, and
these aspects of osteoclast function. Indeed, sealing zone formation.
Src-/- osteoclasts generated in vitro do not More recently, it has been shown that osteo-
resorb bone and exhibit profound defects in cell clasts infected with adenovirus expressing Pyk2
adhesion and spreading on vitronectin, sug- antisense oligonucleotides have signicantly
gesting that c-Src plays an important role in the reduced levels of Pyk2 [43]. Like Src-/- osteo-
adhesion-dependent cytoskeletal organization clasts, these cells do not resorb bone and exhibit
in osteoclasts [96, 122]. Although avb3 integrin defects in cell adhesion, spreading, and sealing
expression and ligand binding afnity are not zone organization, suggesting that Pyk2 is also
altered in Src-/- osteoclasts, the lack of c-Src important for cytoskeletal organization in
results in pronounced aggregation in the basal osteoclasts [43]. Consistent with this, mice that
42 Bone Resorption

lack Pyk2 develop osteopetrosis at an age only Src andPyk2, but also c-Cbl [143] (Figure
similar to b3-/- mice [158]. Furthermore, 3.6). The product of the proto-oncogene c-Cbl is
mice that lack both Src and Pyk2 are more a 120 kDa adaptor protein which is tyrosine-
severely osteopetrotic than either of the single phosphorylated in response to the activation of
mutants [158], indicating that these two pro- various signaling pathways, including M-CSF
teins might interact during normal osteoclast stimulation in monocytes and v-Src transfor-
functioning. mation, EGF, or PDGF activation in broblasts
Tyrosine phosphorylation and kinase activity [168]. Tanaka and colleagues reported that the
of Pyk2 have been reported to be markedly level of tyrosine phosphorylation of c-Cbl
reduced in pre-osteoclasts derived from Src-/- immunoprecipitated from Src-/- osteoclasts is
mice [42]. This contrasts ndings reported by markedly reduced, compared with levels in
Sanjay et al. [143], who reported that, upon wild-type osteoclasts [167]. Furthermore,
adhesion, Src-/- osteoclasts demonstrated levels c-Src is found to associate and co-localize with
of Pyk2 phosphorylation comparable to those c-Cbl in the membranes of intracellular vesicles
found in wild-type osteoclasts. The reasons for in osteoclasts [167]. c-Cbl is a negative regula-
this discrepancy are not known, but it should be tor of several receptor and non-receptor tyro-
noted that the cells used in these two studies sine kinases [142, 174]. Consistent with this
represent different stages of differentiation. function, overexpression of Cbl results in
Moreover, in adherent osteoclasts, Pyk2 is decreased Src kinase activity. Interestingly,
tightly associated with c-Src via its SH2- inhibitory effects are seen with any fragment of
domain. An increase in intracellular calcium Cbl that contains the N-terminal phosphotyro-
that results from the engagement or cross- sine-binding (PTB) domain, regardless of
linking of the avb3 integrin may activate Pyk2 whether the Src-binding proline-rich region
by inducing autophosphorylation of Tyr402. is present [143]. The inhibition of Src kinase
This phosphorylated residue binds to the Src activity, and the binding to Src of v-Cbl (which
SH2-domain [40], displacing the inhibitory Src lacks the proline-rich domain of Cbl and there-
phosphotyrosine 527 and activating Src, leading fore will not bind to the Src SH3-domain)
to further recruitment and activation of Src- requires the presence of phosphorylated Src
mediated downstream signals.At the same time, Tyr416, the autophosphorylation site of Src
activation of Pyk2 following integrin-ligand on the activation loop of the kinase domain
engagement results in the recruitment of [143].
cytoskeletal proteins in a manner similar to FAK In HEK293 cells, overexpression of the Cbl
[146]. The N-terminal domain of FAK was constructs inhibits cell adhesion in parallel with
shown to interact with the cytoplasmic domain decreases in Src kinase activity. Thus, c-Cbl and
of the b integrin subunit. Besides binding to Src, fragments of Cbl that contain the PTB domain
the autophosphorylation sites in FAK and Pyk2 reduce cell adhesion, while the C-terminal half
can bind to SH2-domains of phospholipase C-g of Cbl, which contains the Src SH3-binding
[122, 146, 202]. Furthermore, the C-terminal proline-rich region, but not the PTB domain,
domain of FAK and Pyk2 contain binding sites does not [143]. Interestingly, the absence of
for Grb-2, p130Cas, and paxillin [146]. The asso- either c-Src or c-Cbl decreases both cell motil-
ciation and activation of c-Src and Pyk2 could ity (the rapid extension and retraction of lamel-
initiate a cascade of activation and recruitment lipodia) and directional migration of authentic
of additional signaling and structural molecules osteoclasts isolated from Src-/- or Cbl-/- mice,
to the avb3 integrin-associated protein complex and freshly plated osteoclasts from Src-/- mice
that together modulate the actin cytoskeleton in fail to disassemble podosomes efciently as
osteoclasts. the cell spreads [143], suggesting that the
Pyk2/Src/Cbl complex is required for the dis-
assembly of attachment structures. Increased
Association of Pyk2 with Src and stability of podosomes could well explain the
c-Cbl Downstream of Integrins decreased motility of the Src-/- and Cbl-/-
osteoclasts.
Engagement of avb3 integrin induces the for- Another recently described function of c-Cbl
mation of a signaling complex that contains not could contribute to the regulation and stability
Regulation of Osteoclast Activity 43

of podosomes in osteoclasts. It has been recently avb3 Integrin-Dependent Activation


shown that Cbl acts as a ubiquitin ligase, tar-
geting ubiquitin conjugating enzymes to the of PI3-Kinase
kinases that Cbl binds and down-regulates, with
In avian osteoclasts, avb3 is associated with the
the result that the kinases are ubiquitinated and
signaling molecule PI3-kinase and c-Src [78].
degraded by the proteosome [82, 101, 199]. Since
Interaction of avb3 with osteopontin resulted in
active, but not inactive, Src is ubiquitinated and
increased PI3-kinase activity and association
degraded at this location [65], it could be that
with Triton-insoluble gelsolin [24]. In murine
once Cbl is bound to the Pyk2/Src complex,
osteoclasts formed in culture, PI3-kinase was
it recruits the ubiquitinating system to the
found to translocate into the cytoskeleton upon
podosome. This would lead to the ubiquitina-
osteoclast attachment to the bone surface [98].
tion and degradation of the other components
In addition, potent inhibitors of PI3-kinase,
of the complex, and thereby participating in the
such as wortmannin, inhibit mammalian osteo-
turnover of podosome components that are
clastic bone resorption in vitro and in vivo
required to ensure cell motility.
[125].
Gelsolin, an actin-binding protein, regulates
Recruitment of Other the length of F-actin in vitro, and thus cell shape
and motility [32]. More recently, gelsolin-
Cytoskeletal Proteins decient mice were generated that express mild
defects during hemostasis, inammation, and
Another integrin-dependent signaling adaptor
possibly skin remodeling [187]. Gelsolin-/- mice
recently characterized in osteoclasts is p130Cas
have normal tooth eruption and bone develop-
(Cas, Crk-associated substrate). In a number of
ment, with only modestly thickened calcied
different cell types, p130Cas has been shown to
cartilage trabeculae attributed to delayed carti-
serve as a substrate for either FAK, Pyk2, Src
lage resorption along with a mild osteopetrosis
kinase, or Abl [146]. In osteoclasts, p130Cas can
[25]. However, osteoclasts isolated from gel-
directly associate with the proline-rich motifs in
solin-/- mice lack podosomes and actin rings,
the C-terminal domains of FAK and Pyk2 via its
and display reduced cell motility [25].
SH3-domain. Similar to Pyk2, p130Cas localizes
In conclusion, there is little doubt that the
to the actin ring formed in osteoclasts on glass,
integrins that are present in the osteoclast
to the sealing zone on bone, and is highly tyro-
membrane and particularly in the avb3 vit-
sine phosphorylated upon osteoclast adhesion
ronectin receptor play critical roles in osteoclast
to extracellular matrix proteins [95, 121]. In
biology. This involves not only adhesion, but
adherent osteoclasts, p130Cas constitutively asso-
also the regulation of outside-in signaling,
ciates with Pyk2, suggesting that this adapter
which ensures the proper organization of the
molecule participates in the integrin-PYK2-
cytoskeleton, and of inside-out signaling,
signaling pathway [95].
which ensures modulation of the afnity of the
Much less is known about the signals that are
receptors for their substrate. These two regula-
generated by phosphorylation of the p130Cas
tory modes ensure the assembly and disassem-
adaptor protein. In broblasts, integrin-
bly of the attachment structures (podosomes),
stimulated tyrosine phosphorylation of p130Cas
a cyclic process necessary for efcient cell
promotes binding to the SH2-domain of either
motility.
Crk [182] or Nck [145]. More recently, expres-
sion of SH3-domain of p130Cas inhibited FAK-
mediated cell motility [17], while overexpression A Role for Tyrosine Phosphatases
of Crk was shown to promote cell migration in
a Rac-dependent and Ras-independent manner Reversible phosphorylation of tyrosine residues
[88]. Moreover, the downstream components of in proteins is a central regulator of cellular func-
this Rac-mediated migration pathway appear to tions, and is controlled by the opposing actions
involve phosphatidyl inositol 3-kinase (PI3- of protein tyrosine kinases (PTKs) and tyrosine
kinase) [150]. In osteoclasts, the PI3-kinase phosphatases (PTPs) [3, 80, 103, 178].
dependent target was found to be the cytoskele- Tyrosine phosphorylation plays a major role
tal-associated protein gelsolin [24]. in regulating bone resorption, as discussed
44 Bone Resorption

above, and one may consequently expect that (Hcphme/Hcphme; abbreviation me/me) results
dysregulation of PTPs also will produce bone in a frame shift and premature truncation of
abnormalities. Indeed, aberrant tyrosine phos- the mRNA, producing an essentially SHP-1 null
phorylation also can be caused by changes in phenotype [153]. me/me animals die at about
PTP activity. Inhibition of the receptor-type three weeks of age, a result of inammatory lung
phosphatase PTP-OC in cultured rabbit osteo- disease [154]. Another mutation results in
clasts reduces their resorption activity [162, proteins that retain approximately 25% of the
191], while inhibition of OST-PTP abrogates wild-type activity and the mutant mice are
differentiation of cultured osteoblasts [28, 114, consequently viable and designated mev/mev
115]. In addition, as discussed below, mice [153].
lacking SHP-1 are osteopenic as a consequence Given the importance of tyrosine phosphory-
of increased osteoclast function [4, 179], indi- lation in pathways such as M-CSF receptor acti-
cating that this PTP is a negative regulator of vation [68], integrins [143], and Src-dependent
osteoclasts. Mice lacking PTPe show a decrease signaling [175], it was predicted that SHP-1
in osteoclast activity, indicating that it can be a would play a signicant role in the modulation
positive regulator. of signaling in osteoclasts. This prediction was
borne out by studies of the formation and func-
tion of osteoclasts using the milder mev/mev
SHP1 Down-regulates mutant mouse [4, 179]. The mev/mev mouse is
Osteoclast Function osteopenic, exhibiting decreased trabecular
thickness with increased bone resorption
Many signaling pathways in osteoclasts cru- indices, particularly an increased number of
cially depend on tyrosine phosphorylation, and osteoclasts. Impaired osteoblast function also
defective signaling by one or another tyrosine may contribute to the phenotype, since the
kinase is implicated in several of the currently osteoid volume, mineralizing surface and
known osteopetrotic mice (i.e., the M-CSF osteoblast surface are decreased compared to
receptor, c-Src). The normal function of these controls. In vivo and in vitro data reveal that
signaling mechanisms requires that the tyrosine both osteoclast differentiation and osteoclast
phosphorylation of signaling effectors be activity are increased in the absence of normal
limited, both in terms of time and amplitude, by SHP-1, suggesting that this tyrosine phos-
dephosphorylation catalyzed by protein tyro- phatase is a negative regulator of osteoclastoge-
sine phosphatases (PTPs), and, in fact, several nesis and, possibly, of osteoclast resorbing
PTPs have recently been identied to play activity. The increased osteoclastogenesis is
important roles in the modulation of osteoclast attributable to effects of the mev/mev mutation
signaling. in the hematopoietic precursor cells, because
One such PTP is the Src homology 2-domain- the osteoclastogenic potency of stromal cells is
containing phosphatase 1 (SHP-1) [1] (also not affected. The fact that increased osteoclas-
known as PTP1c [151], SHPTP-1 [136], HCP togenesis is intrinsic to the hematopoietic
[198] or PTPN6 [137]) that functions as a lineage excludes the possibility that it could be
cytosolic protein-tyrosine phosphatase (PTP) a result of increased RANKL or of a decrease in
and is predominantly expressed in hematopoi- OPG production by cells of the stromal lineage
etic cells [113, 137, 151, 198]. In various cell or osteoblasts [94, 157].
types, SHP-1 associates with tyrosine phospho- Thus, SHP-1 apparently down-regulates sig-
rylated receptors, including those for EGF [151, naling in response to factor(s) that activate
177], interleukin (IL)-3, IL-4 and IL-13 [69, 197], differentiation and possibly bone resorption.
c-Kit [196], erythropoietin [89], platelet-derived Indeed, M-CSF signaling, Src and NF-kB activ-
growth factor [200], and M-CSF [26], raising the ity have all been shown to be affected by
possibility that SHP-1 might terminate signals reduced SHP-1 activity. In me/me macrophages,
generating from activation of these receptors. where SHP-1 is totally inactive, the M-CSF
Spontaneous homozygous mutations of the receptor is hyperphosphorylated. At least some
gene encoding SHP-1 (Hcph [153]) give rise to of its associated proteins are hyperactivated
the motheaten phenotype in mice, character- when the cells are treated with M-CSF [26] this
ized by immunodefeciency, systemic autoimmu- indicates that SHP-1 is involved in the dephos-
nity and premature death [62]. One mutation phorylation of the M-CSF receptor and suggests
Regulation of Osteoclast Activity 45

that SHP-1 is a critical negative regulator of M- young (<12 weeks) female mice, but markedly
CSF signaling. The increase in osteoclast forma- less so in adult female or in male mice, suggest-
tion in mev/mev mice could thus be explained as ing that lack of PTPe may affect sex hormone-
a result of the failure of SHP-1 to down-regulate related signaling pathways in osteoclasts from
M-CSF receptor signaling mechanisms. Src affected mice. At the cellular level, cultures of
activity is regulated by its tyrosine phosphory- osteoclast-like cells from PTPe-decient mice
lation state and the absence of SHP-1 in me/me failed to polarize and exhibited abnormalities in
thymocytes leads to the hyperactivation of Src the structure and localization of podosomes. Src
family kinases [107]. A similar Src-related effect activity, Pyk2 phosphorylation, and response to
could be responsible for increased osteoclastic RANKL treatment were, however, normal in
activity. A third possibility is that the lack of osteoclasts from female mice lacking PTPe, sug-
SHP-1 activity promotes the activation of NF-kB gesting these signaling pathways are not gener-
downstream of RANK in osteoclast precursors, ally affected. These results establish that PTPe is
as it does in both mev/mev T and B cells [86]. required for optimal structure and organization
of podosomes in osteoclasts in the context of
PTPe Activates Osteoclast Function cell polarization, and is essential for the optimal
function of these cells in vitro and in vivo.
The protein tyrosine phosphatase epsilon
(PTPe) subfamily contains four distinct pro-
teins, all products of a single gene. The two SHIP: An Inositol Phosphatase that
major forms of PTPe are the receptor-type Down-regulates Osteoclast Function
(RPTPe) and non-receptor type (cyt-PTPe) iso-
forms [45, 46, 93, 127, 169]. Two other forms of In a recent study, Takeshita et al. [166] identied
PTPe are p67 and p65 PTPe; these are shorter SH2-containing inositol-5-phosphatase (SHIP)
molecules, whose production is regulated at the as an important regular of osteoclast function.
levels of translation and post-translational pro- SHIP is a protein that becomes tyrosine phos-
cessing [60, 61]. The four PTPe forms differ only phorylated and associated with Shc after acti-
at their amino termini, resulting in their unique vation of membrane receptors for a number
subcellular localization patterns and distinct of hematopoietic cytokines. SHIP has an N-
physiological functions. RPTPe assists Neu- terminal SH2-domain, a central inositol poly-
induced mouse mammary tumor cells maintain phosphate-5-phosphatase catalytic domain, and
their transformed phenotype, most likely by two NPXY sequences that bind phosphotyro-
dephosphorylating and activating Src in vivo sine binding domains when phosphorylated
[44, 46, 59]. The same form of PTPe also can and a proline-rich C terminus. SHIP specically
down-regulate insulin receptor signaling in recognizes and cleaves the 5-phosphate group
cultured cells [2, 120]. In some systems PTPe can from phosphatidylinositol-3,4,5-trisphosphate
down-regulate mitogenic signaling, as exempli- (PIP3), the major product of PI3-K activity, and
ed by its inhibition of mitogen-activated from inositol-1,3,4,5-tetrakisphosphate.
protein kinase (ERK1/2) activity [176, 183] and Signals promoting osteoclast precursor sur-
inhibition of Janus kinase-signal transducer vival, differentiation, and function that are gen-
and activator of transcription (JAK-STAT) sig- erated after engagement of M-CSF R, RANK or
naling in M1 leukemia cells [170172]. PTPe integrin receptors lead to activation of PI3-K.
also has been shown to be important for proper The ability of SHIP to dephosphorylate PIP3
function of macrophages [163]. suggested this 5-phosphatase also may inhibit
In a recent study [29], mice genetically osteoclast recruitment and function, a hypoth-
lacking PTPe were shown to have an altera- esis tested by by examining mice with a
tion in bone remodeling, affecting primarily homozygous deletion of SHIP [166]. The
females. Cyt-PTPe is strongly expressed in animals did not survive after 14 weeks due to a
osteoclasts, and young female mice genetically myeloproliferative disorder, an observation that
lacking PTPe exhibit increased trabecular bone is consistent with SHIP functioning as a nega-
mass. This phenotype is attributable to reduced tive regulator of cytokine signaling. More rele-
bone resorption by osteoclasts, which function vant to this chapter, however, the deletion of
abnormally both in vitro and in vivo. Reduced SHIP markedly increased the sensitivity of
bone resorption is most readily detected in marrow macrophages to M-CSF and RANKL,
46 Bone Resorption

enhancing the generation of large and hyper-


resorptive osteoclasts in a cell autonomous
manner. As a consequence of this hyperactivity
AC + C1a-CTR
of the SHIP-/- osteoclasts, the mice developed a - Gs
severe form of osteoporosis. cAMP
Gi
Signaling from the Calcitonin PKA - Gq
Receptor
Because of its potent inhibitory effects on osteo-
PKC DAG PLC
Erk1/2
clast activity, calcitonin has long been recog- IP3
nized as a potential therapeutic agent for the Ca2+
treatment of diseases that are characterized by A
increased bone resorption including osteoporo-
sis, Pagets disease, and late-stage malignancies
(Figure 3.7). Signaling mechanisms down-
stream of the calcitonin receptor have, there-
fore, been of great interest. A comprehensive
discussion of calcitonin-induced signaling Gq ECM
PLC
appears elsewhere in this volume; hence we Gi
focus here on what is known of the interaction Integrin
of calcitonin-activated signaling events with PKC , Ca2+
attachment-related signaling. F-actin,
In situ, calcitonin reduces contact of osteo-
clasts with the bone surface and alters osteoclast Src
morphology [73, 84]. Cultured in vitro, calci- MEK
tonin-treated osteoclasts retract and become P-
FAK
less mobile [22, 23, 201]. These effects suggest P-HEF1 pax
that some of the key targets of calcitonin sig- -P
naling are involved in cell attachment and Erk1/2
B
cytoskeletal function, possibly in conjunction
with the integrins and/or their signaling Figure 3.7 Signal transduction from the calcitonin receptor
function. involves three different G proteins (A) and activation of PKA and
The calcitonin receptor, a G protein-coupled PKC and PLC. (B) Cross-talk between the CTR and integrin
receptor that has been cloned from several signaling via Gi and Gq regulates the cytoskeleton.
species and cell types, couples to multiple het-
erotrimeric G proteins (Gs, Gi/o, and Gq) [19, 27,
52, 155, 156]. For technical reasons, much of
the recent characterization of signaling down-
stream of the calcitonin receptor has been nisms that involve the bg subunits of pertussis
conducted in cells other than osteoclasts, toxin-sensitive Gi, as well as pertussis toxin-
particularly cell lines that express recombinant insensitive signaling via phospholipase C, PKC
calcitonin receptor. Because it couples to multi- and elevated intracellular calcium, ([Ca2+]i).
ple G proteins, the proximal signaling mecha- It has been shown in the same cell line that
nisms that are activated by calcitonin include calcitonin induces the tyrosine phosphoryla-
many classical GPCR-activated effectors, such as tion and association of components of cellular
adenylyl cyclase and protein kinase A (PKA); adhesion complexes, including FAK, paxillin,
phospholipases C, D, and A2; and protein kinase and HEF1, a member of the p130Cas family [204].
C (PKC). In HEK 293 cells that express the rabbit Interestingly, while these cells express both
calcitonin receptor, calcitonin induces the phos- HEF1 and p130Cas, only HEF1 is phosphorylated
phorylation and activation of the ERK1/2 and associates with FAK and paxillin. This
pathway via extracellular signal-regulated response to calcitonin is independent of adeny-
kinases, Erk1 and Erk2 [27, 128], via mecha- lyl cyclase/PKA and of pertussis toxin-sensitive
Regulation of Osteoclast Activity 47

mechanisms and appears to be mediated by the kinases: IKK, JNK, p38m Erk and Src, as well as
pertussis toxin-insensitive PKC/[Ca2+]i signal- c-fos and NFAT activation. [Six main pathways
ing pathway. The relevance of the FAK/paxillin/ are activated downstream of RANK: TRAF6-
HEF1 phosphorylation to integrins and the Src, Akt, NFkB, JNK, p38, and c-fos-IFN b.]
actin cytoskeleton is demonstrated by its TRAF 2, 5, and 6 all have been shown to bind to
requirement for integrin attachment to the sub- RANK, and their individual docking sites have
stratum and its sensitivity to agents (cytocha- been mapped, yet only TRAF 6 inactivation
lasin D, latrunculin A) that disrupt actin results in osteopetrosis due to loss of osteoclast
laments [203]. The calcitonin-induced tyrosine activity [90, 94, 106] and deletion of the mem-
phosphorylation of paxillin and HEF1 is brane-proximal TRAF6 binding site specically
enhanced by the overexpression of c-Src and prevents the restoration of osteoclastogenic
strongly inhibited by the overexpression of a potential in RANK-/- derived hematopoietic
dominant negative kinase-dead Src, indicating precursors [6].
that c-Src is required at some point in the cou- TRAF acts as a key adaptor to assemble sig-
pling mechanism. Interestingly, the dominant naling complexes that direct osteoclast-specic
negative Src has little effect on the calcitonin- gene expression leading to differentiation and
induced phosphorylation of Erk1/2, in contrast activation. The two best studied pathways are
to what has been reported for some other G those leading to the activation of NFkB and
protein coupled receptors [38, 39], indicating AP-1 transcription factors, both of which are
that some aspects of calcitonin-induced signal- required for osteoclast formation [53, 64, 192].
ing may be signicantly different from other Activation of these transcription factors is
better studied G protein-coupled receptors. induced by phosphorylation cascades mediated
These or similar events could well play impor- by protein kinases, including IKK1/2 (NFkB),
tant roles in mediating the calcitonin-induced and JNK1 (AP-1) [34, 53, 64, 85, 192]. Coupling
changes in cell adhesion and motility in of RANK to IKK and JNK involves the ERK1/2-
osteoclasts. related TGFb-inducible kinase, TAK1, along
with the TRAF-binding adapter protein TAB2,
both of which are present in activated receptor
Signaling by the M-CSF Receptor complexes [100, 119], as shown by the inhibition
(c-Fms) and RANK of RANKL-mediated activation of both IKK1/2
and JNK2 by dominant-negative mutant forms
These two receptors and their respective of TAK1 [193]. The ERK1/2-related kinase
ligands, M-CSF and RANKL, have been demon- MKK7 also is required for JNK activation in
strated to be both necessary and, when co- osteoclasts.
activated, sufcient to induce the differentiation The stress-activated protein kinase p38,
of osteoclasts precursors and to regulate the apparently activated by RANK-induced phos-
activity and survival of the mature osteoclasts phorylation by MKK [104, 112], also is involved
[94, 138, 161]. Deletion of the genes encoding in mediating key signals from RANK, coupling
RANK, RANKL, or M-CSF lead to a block to the downstream activation of the transcrip-
in osteoclast differentiation, producing an tional regulator mi/Mitf and the expression of
osteopetrotic phenotype [161, 173]. TRAP and cathepsin K [110]. ERK1/2 pathway
activation via the extracellular signal-regulated
Signaling by RANK (Figure 3.8) kinase 1 (Erk1) is also activated by RANK sig-
naling, apparently via activation of MEK1 [77,
A number of the RANK-induced signaling path- 184]. MEK activation of Erk appears to play an
ways in osteoclasts ultimately induce the expres- important role in protecting osteoclasts from
sion of several genes, including TRAP, cathepsin apoptosis [118].
K, the calcitonin receptor, the aVb3 integrin, and Src binding to TRAF-6 allows RANK-
the transcriptional regulators mi/Mitf, myc and mediated signaling to activate rst PI3-K and
NFAT2. [41, 102]. Signaling is initiated by the consequently the Akt/PKB [188], which induce
rapid binding of TRAFs to specic domains in cell survival, cytoskeletal rearrangements, and
the cytoplasmic tail of RANK [33, 56, 79]. TRAF motility. In dendritic cells, activated RANK
binding, in turn, induces at least ve distinct recruits TRAF-6, c-Cbl, and PI3-K in a Src
phosphorylation cascades mediated by protein kinase-dependent manner [7]. TRAF-6, in turn,
48 Bone Resorption

RANKL

RANK
Src / Cbl
TRAF6

TAB2

IRAK
TAK1

NIK MKK7
MEK1
PI3-kinase

JNK1 Erk

c-Fos NFATc1 NF-kB c-Jun Elk Akt

Transcriptional Activity Motility/cytoskeletal


Survival

Figure 3.8 RANKL/RANK signaling activates several pathways that regulate both transcriptional activity and the cytoskeleton, as
well as osteoclast survival. One such pathway involves Src and Cbl.

enhances Src kinase activity, leading to phos- of the Erk and Src pathways also can negatively
phorylation of downstream signaling mole- regulate osteoclastogenesis [77, 152]. Other
cules, including c-Cbl [188]. RANKL-induced mechanisms switch off the RANK-signaling
activation of Akt/PKB is defective in Cbl-b-/- pathway once it has been activated. For example,
dendritic cells (but perhaps not in Cbl-b-/- RANKL induces the expression and secretion of
osteoclast like cells) [7] , implicating Cbl-b as a interferon b, which is secreted and acts in an
positive modulator of RANK signaling. autocrine fashion to down regulate the expres-
RANK expression and signaling appears to be sion of c-fos, a critical factor involved in osteo-
tightly controlled, with several mechanisms clast development [71, 164], while interferon
reported to exert positive or negative control of g promotes the degradation of TRAF 6, and
the pathway, thereby enhancing or inhibiting inhibits in vitro osteoclastogenesis [165].
RANKL-induced osteoclastogenesis and subse-
quent activation. IL-1, M-CSF, TNFa, PGE2, and
TGFb all potentiate osteoclastogenesis in vitro, Signaling by c-Fms, the M-CSF
and can stimulate bone resorption in vivo. In Receptor
contrast, inhibition of MEK1 and mTOR, its
downstream target, lead to increased osteoclas- Activation of c-Fms by its ligand, M-CSF, pro-
togenesis in vitro. This suggests that activation motes the proliferation and survival of cells of
Regulation of Osteoclast Activity 49

the macrophage/monocyte lineage, including tion, cell spreading, and increased migration in
osteoclasts and their precursors [48, 68]. In osteoclasts [81]. M-CSF also induces relocaliza-
addition, M-CSF is required for osteoclast dif- tion of PI3-K to the cell membrane [63, 132] and
ferentiation, at least in part by inducing the decreases bone resorption [55, 70]. Of possible
expression of RANK in osteoclast precursors, relevance to our observation of elevated RANK
priming the cells to differentiate in response to on the surface of Cbl-b-/- osteoclast like cells,
RANKL [5]. Following M-CSF-induced dimer- activating c-Fms increases the levels of RANK
ization, c-Fms autophosphorylates several on the cell surface, and in addition, up-regulates
tyrosines on its cytoplasmic tail, thereby pro- other components of RANK signaling pathways
viding binding sites for several SH2-containing [5, 109, 195].
proteins (c-Src, Y559; STAT 1, Y706; PI3-K, Y721; The effect of M-CSF on the spreading of both
Grb 2, Y697, and Y921) that are recruited to the macrophages and mature osteoclasts is brought
c-Fms-associated signaling complex. c-Cbl has about by cytoskeletal rearrangements, involv-
been shown to interact with c-Fms via the phos- ing Src-dependent phosphorylation [81] and
phorylated tyrosine Y973. c-Cbl appears to play translocation of PI3-kinase from the cytoplasm
both positive and negative signaling roles to the cell membrane [63]. Upon ligand binding,
downstream of the activated c-Fms. Once c-Fms phosphorylates a number of proteins,
bound to c-Fms, it couples the activated recep- including c-Cbl and ERK1/2, leading to regula-
tor to several downstream signaling effectors, tion of cell cycle progression by cyclins.
including PI3-K [131] and Akt/PKB, the Feng et al. have recently shown that of all the
ERK1/2s, and the JAK/STAT system (Figure 3.9). autophosphorylated tyrosines on the cytoplas-
The activation of Erk 1,2 may protect osteo- mic domain of the activated c-Fms, only Y559
clasts from apoptosis [117]. At the same time, it (the Src binding site) and Y807, which is the
targets the ubiquitination machinery to the most strongly phosphorylated tyrosine whose
activated receptor, promoting c-Fms ubiquiti- binding partners have not yet been identied,
nation and rapid endocytosis and attenuating are required for osteoclast differentiation [49].
macrophage proliferation [99]. Interestingly, Y807-dependent signaling mecha-
We previously have shown that M-CSF nisms are not required for normal bone resorp-
induces Src-dependent cytoskeletal reorganiza- tion by mature osteoclasts.
M-CSF stimulates motility and cytoplasmic
spreading in mature osteoclasts and both Src
and c-Cbl play important roles in these
M-CSF
processes. In WT osteoclasts, M-CSF treatment
induces rapid cytoplasmic spreading, with
redistribution of F-actin from a well-delineated
M-CSFR / c-Fms central attachment ring to the periphery of the
Src cell. It simultaneously induces the phosphoryla-
Cbl tion of several cellular proteins in osteoclast-
PI3-K like cells, including c-Fms and c-Src, and
Tyk2
concurrently increases Src kinase activity three-
ERK fold. More recent and unpublished studies in
our laboratory have shown that treatment of
RAW cells with M-CSF results in rapid phos-
JAK/STAT AKT phorylation of both c-Fms and c-Cbl, consistent
with other reports of the role of c-Cbl in regu-
Cell spreading and Migration lating c-Fms signaling. In other unpublished
experiments, a role of c-Cbl in M-CSF-mediated
signaling was demonstrated further by the
Figure 3.9 MCSF signaling activates and subsequent phos- decreased migration of c-Cbl-/- osteoclast like
phorylation of several proteins, including Cbl. Cbl, promotes cells through a semipermeable membrane in
ubiquitination and endocytosis of c-Fms. PI3-kinase interacts response to M-CSF in a Boyden chamber assay.
with the receptor resulting in the activation of AKT leading to
cytoskeletal reorganization and cell spreading.
50 Bone Resorption

DAP12 /TREM2, Co-receptors for of typical osteoclast markers such as integrin


avb3 and calcitonin receptor. These cells demon-
the Immune System, Act as Key strate decreased activity in dentin resorption
Co-receptors for RANK in the assays, potentially reecting inefcient bone
Regulation of Osteoclast resorption in vivo.
Consistent with these results, defective devel-
Function [18, 83] opment and resorptive function of osteoclasts
have been recently observed in DAP12-decient
Rare TREM-2- and DAP12-decient individuals mice [83]. Thus, DAP12/TREM-2 may guide
recently have been identied [133, 134]. These myeloid cell differentiation by modulating
individuals are affected by a genetic syndrome responsiveness to certain cytokines or reinforc-
characterized by bone cysts and presenile ing signaling pathways triggered by cytokine
dementia called Nasu-Hakola disease, or poly- receptors or integrins [42, 121123, 143]. The
cystic lipomembranous osteodysplasia with TREM-2/DAP12 signaling pathway also may
sclerosing leukoencephalopathy [66]. DAP12- play an important role in promoting actin poly-
decient (DAP12-/-) mice develop an increased merization, organization of the cytoskeleton,
bone mass (osteopetrosis) and a reduction and rufed border trafcking via recruitment of
of myelin (hypomyelinosis) accentuated in protein tyrosine kinases, activation of phospho-
the thalamus. In vitro osteoclast induction lipase C1, and calcium mobilization. These func-
from DAP12-/- bone marrow cells yielded tions are critical for promoting cell fusion, as
immature cells with attenuated bone resorption well as transforming osteoclast precursors into
activity. a polarized cell type capable of resorbing bone
In the immune system, DAP12 is expressed as in a highly regulated fashion.
a disulde-linked homodimer that associates
with various immunoreceptors, many of whose
ligands are unknown, including TREM2 [14, 36].
TREMs are members of the immunoglobulin
superfamily that promote myeloid cell activa- Conclusions
tion by associating with DAP12. DAP12 harbors
an immunoreceptor tyrosine-based activation The last decade has brought signicant progress
motif (ITAM) also found in other immunore- to our understanding of the mechanisms by
ceptor adapters such as the Fc receptor common which osteoclasts resorb bone and of the regu-
chain. Tyrosine phosphorylation of ITAM of lation of these activities. As discussed in this
these immunoreceptor adapters by Src-family chapter, the key elements of function can be
kinase is the primary step for generation of an summarized, beyond differentiation, as follows:
activation signaling motif that functions as 1) Adhesion and migration, via adhesion recep-
docking site for the protein tyrosine kinases Syk tors and their ability to regulate the rapid
and Zap70. These kinases mediate a cascade of assembly and disassembly of cytoskeletal pro-
phosphorylation events that ultimately activate teins at sites of adhesion (podosomes). 2) Secre-
the cell. Within the TREM receptor family, tion of proteolytic enzymes, via vesicular
TREM-2 is selectively expressed on monocyte- transport to the secretory pole of the cell, i.e.,
derived dendritic cells, promoting their migra- the rufed border. 3) Internalization also via
tion and activation in vitro, suggesting that it vesicular transport, but in a retrograde manner,
may have a role in antigen presentation and T- i.e., from the rufed border to lysosomes or
cell stimulation in vivo. This observation sug- transcytosis. 4) Acidication via the apical vac-
gests that the TREM-2DAP12 complex may uolar proton ATPase and the ClC-7 chloride
regulate the function of an unexpectedly vast channel at the rufed border membrane as well
array of myeloid cells, including osteoclasts. as homeostatic ion transport at the basolateral
Indeed, TREM-2decient individuals have a membrane. The key regulatory pathways identi-
striking defect in osteoclast development, which ed to date involve signaling from the adhesion
results in impaired bone resorption in vitro. receptors, particularly the integrins, signaling
TREM-2decient osteoclast precursors fail to from the RANK and M-CSF receptors and sig-
fuse into multinucleated cells, do not develop naling from the calcitonin receptor. These
podosomes, and show reduced or no expression pathways converge upon both transcriptional
Regulation of Osteoclast Activity 51

regulation of key osteoclastic genes and upon 8. Baron R, Neff L, Brown W, Courtoy PJ, Louvard D,
the regulation of vesicular trafc, the cytoskele- Farquhar MG (1988) Polarized secretion of lysosomal
enzymes: co-distribution of cation- independent
ton and ion transport systems. Most of these mannose-6-phosphate receptors and lysosomal
signaling cascades involve sequential phospho- enzymes along the osteoclast exocytic pathway. J Cell
rylation and dephosphorylation events, both Biol 106:186372.
on tyrosines and on serine-threonines. All 9. Baron R, Neff L, Louvard D, Courtoy PJ (1985) Cell-
mediated extracellular acidication and bone resorp-
components of the regulatory pathways that tion: evidence for a low pH in resorbing lacunae and
modulate the activity of the osteoclast and bone localization of a 100-kD lysosomal membrane protein
resorption constitute valid targets for drug dis- at the osteoclast rufed border. J Cell Biol 101:
covery or explain the mechanisms by which 221022.
existing drugs affect the cells. 10. Baron R, Neff L, Roy C, Boisvert A, Caplan M (1986)
Evidence for a high and specic concentration of
(Na+,K+)ATPase in the plasma membrane of the osteo-
clast. Cell 46:31120.
11. Berry V, Rathod H, Pulman LB, Datta HK (1994)
Acknowledgments Immunouorescent evidence for the abundance of
focal adhesion kinase in the human and avian osteo-
clasts and its down regulation by calcitonin. J
The authors are supported by NIH grants Endocrinol 141:R1115.
DE-04724, AR-42927, and AR-48218 to R.B. and 12. Blair HC, Teitelbaum SL, Ghiselli R, Gluck S (1989)
AR-49879 to W.C.H. The authors are indebted Osteoclastic bone resorption by a polarized vacuolar
to Archana Sanjay, Cecile Itsztein, Olivier proton pump. Science 245:8557.
13. Bossard MJ, Tomaszek TA, Thompson SK, Amegadzie
Destaing, and Lynn Neff for help with some BY, Hanning CR, Jones C, et al. (1996) Proteolytic activ-
gures. ity of human osteoclast cathepsin K. Expression,
purication, activation, and substrate identication.
J Biol Chem 271:1251724.
14. Bouchon A, Hernandez-Munain C, Cella M, Colonna M
References (2001) A DAP12-mediated pathway regulates expres-
sion of CC chemokine receptor 7 and maturation of
human dendritic cells. J Exp Med 194:111122.
1. Adachi M, Fischer EH, Ihle J, Imai K, Jirik F, Neel B, et 15. Boyce BF, Yoneda T, Lowe C, Soriano P, Mundy GR
al. (1996) Mammalian SH2-containing protein tyrosine (1992) Requirement of pp60c-src expression for osteo-
phosphatases. Cell. 85:15. clasts to form rufed borders and resorb bone in mice.
2. Andersen JN, Elson A, Lammers R, Romer J, Clausen J Clin Invest 90:16227.
JT, Moller KB, et al. (2001) Comparative study of 16. Boyle WJ, Simonet WS, Lacey DL (2003) Osteoclast dif-
protein tyrosine phosphatase-e isoforms: membrane ferentiation and activation. Nature 423:33742.
localization confers specicity in cellular signalling. 17. Cary LA, Han DC, Polte TR, Hanks SK, Guan JL (1998)
Biochem J 354:58190. Identication of p130Cas as a mediator of focal adhe-
3. Andersen JN, Mortensen OH, Peters GH, Drake PG, sion kinase-promoted cell migration. J Cell Biol
Iversen LF, Olsen OH, et al. (2001) Structural and evo- 140:21121.
lutionary relationships among protein tyrosine phos- 18. Cella M, Buonsanti C, Strader C, Kondo T, Salmaggi A,
phatase domains. Mol Cell Biol 21:711736. Colonna M (2003) Impaired differentiation of osteo-
4. Aoki K, DiDomenico E, Sims NA, Mukhopadhyay K, clasts in TREM-2-decient individuals. J Exp Med
Neff L, Houghton A, et al. (1999) The tyrosine phos- 198:64551.
phatase SHP-1 is a negative regulator of osteoclasto- 19. Chabre O, Conklin BR, Lin HY, Lodish HF, Wilson E,
geneisis and osteoclast resorbing activity: increased Ives HE, et al. (1992) A recombinant calcitonin recep-
resorption and osteopenia in mev/mev mutant mice. tor independently stimulates 3,5-cyclic adenosine
Bone 25:2617. monophosphate and Ca2+/inositol phosphate signaling
5. Arai F, Miyamoto T, Ohneda O, Inada T, Sudo T, Brasel pathways. Mol Endocrinol 6:5516.
K, et al. (1999) Commitment and differentiation of 20. Chalhoub N, Benachenhou N, Rajapurohitam V, Pata
osteoclast precursor cells by the sequential expression M, Ferron M, Frattini A, et al. (2003) Grey-lethal muta-
of c-Fms and receptor activator of nuclear factor kB tion induces severe malignant autosomal recessive
(RANK) receptors. J Exp Med 190:174154. osteopetrosis in mouse and human. Nat Med 9:
6. Armstrong AP, Tometsko ME, Glaccum M, Sutherland 399406.
CL, Cosman D, Dougall WC (2002) A RANK/TRAF6- 21. Chambers TJ, Fuller K, Darby JA, Pringle JA, Horton
dependent signal transduction pathway is essential for MA (1986) Monoclonal antibodies against osteoclasts
osteoclast cytoskeletal organization and resorptive inhibit bone resorption in vitro. Bone Miner 1:127
function. J Biol Chem 277:4434756. 35.
7. Arron JR, Vologodskaia M, Wong BR, Naramura M, 22. Chambers TJ, Magnus CJ (1982) Calcitonin alters
Kim N, Gu H, et al. (2001) A positive regulatory role for behaviour of isolated osteoclasts. J Pathol 136:2739.
Cbl family proteins in tumor necrosis factor-related 23. Chambers TJ, Revell PA, Fuller K, Athanasou NA (1984)
activation-induced cytokine (TRANCE) and CD40L- Resorption of bone by isolated rabbit osteoclasts. J Cell
mediated akt activation. J Biol Chem 276:3001117. Sci 66:38399.
52 Bone Resorption

24. Chellaiah M, Fitzgerald C, Alvarez U, Hruska K (1998) 39. Della Rocca GJ, van Biesen T, Daaka Y, Luttrell DK,
c-Src is required for stimulation of gelsolin-associated Luttrell LM, Lefkowitz RJ (1997) Ras-dependent
phosphatidylinositol 3-kinase. J Biol Chem 273:11908 mitogen-activated protein kinase activation by G
16. protein-coupled receptors. Convergence of Gi- and Gq-
25. Chellaiah M, Kizer N, Silva M, Alvarez U, Kwiatkowski mediated pathways on calcium/calmodulin, Pyk2, and
D, Hruska KA (2000) Gelsolin deciency blocks Src kinase. J Biol Chem 272:1912532.
podosome assembly and produces increased bone 40. Dikic I, Tokiwa G, Lev S, Courtneidge SA, Schlessinger
mass and strength. J Cell Biol 148:66578. J (1996) A role for Pyk2 and Src in linking G-protein-
26. Chen HE, Chang S, Trub T, Neel BG (1996) Regulation coupled receptors with MAP kinase activation. Nature
of colony-stimulating factor 1 receptor signaling by 383:54750.
the SH2 domain-containing tyrosine phosphatase 41. Dougall WC, Glaccum M, Charrier K, Rohrbach K,
SHPTP1. Mol Cell Biol 16:368597. Brasel K, De Smedt T, et al. (1999) RANK is essential
27. Chen Y, Shyu J-F, Santhanagopal A, Inoue D, David J-P, for osteoclast and lymph node development. Genes &
Dixon SJ, et al. (1998) The calcitonin receptor stimu- Dev 13:241224.
lates Shc tyrosine phosphorylation and Erk1/2 activa- 42. Duong LT, Lakkakorpi PT, Nakamura I, Machwate M,
tion. Involvement of Gi, protein kinase C, and calcium. Nagy RM, Rodan GA (1998) PYK2 in osteoclasts is an
J Biol Chem 273:1980916. adhesion kinase, localized in the sealing zone, acti-
28. Chengalvala MV, Bapat AR, Hurlburt WW, Kostek B, vated by ligation of avb3 integrin, and phosphorylated
Gonder DS, Mastroeni RA, et al. (2001) Biochemical by Src kinase. J Clin Invest 102:88192.
characterization of osteo-testicular protein tyrosine 43. Duong LT, Nakamura I, Lakkakorpi PT, Lipfert L, Bett
phosphatase and its functional signicance in rat AJ, Rodan GA (2001) Inhibition of osteoclast function
primary osteoblasts. Biochemistry 40:81421. by adenovirus expressing antisense protein-tyrosine
29. Chiusaroli R, Knobler H, Luxenburg C, Sanjay A, kinase 2. J Biol Chem 276:748492.
Granot-Attas S, Tiran Z, et al. (2004) Tyrosine phos- 44. Elson A (1999) Protein tyrosine phosphatase e
phatase epsilon is a positive regulator of osteoclast increases the risk of mammary hyperplasia and
function in vitro and in vivo. Mol Biol Cell 15:23444. mammary tumors in transgenic mice. Oncogene 18:
30. Clark EA, Brugge JS (1995) Integrins and signal 753542.
transduction pathways: the road taken. Science 268: 45. Elson A, Leder P (1995) Identication of a cytoplasmic,
2339. phorbol ester-inducible isoform of protein tyrosine
31. Crippes BA, Engleman VW, Settle SL, Delarco J, phosphatase e. Proc Natl Acad Sci USA 92:122359.
Ornberg RL, Helfrich MH, et al. (1996) Antibody to b3 46. Elson A, Leder P (1995) Protein-tyrosine phosphatase
integrin inhibits osteoclast-mediated bone resorption e. An isoform specically expressed in mouse mam-
in the thyroparathyroidectomized rat. Endocrinology mary tumors initiated by v-Ha-ras or neu. J Biol Chem
137:91824. 270:2611622.
32. Cunningham CC, Stossel TP, Kwiatkowski DJ (1991) 47. Engleman VW, Nickols GA, Ross FP, Horton MA,
Enhanced motility in NIH 3T3 broblasts that overex- Griggs DW, Settle SL, et al. (1997) A peptidomimetic
press gelsolin. Science 251:12336. antagonist of the avb3 integrin inhibits bone resorp-
33. Darnay BG, Haridas V, Ni J, Moore PA, Aggarwal BB tion in vitro and prevents osteoporosis in vivo. J Clin
(1998) Characterization of the intracellular domain of Invest 99:2284892.
receptor activator of NF-kB (RANK). Interaction with 48. Fantl WJ, Johnson DE, Williams LT (1993) Signalling
tumor necrosis factor receptor-associated factors and by receptor tyrosine kinases. Annu Rev Biochem 62:
activation of NF-kB and c-Jun N-terminal kinase. J Biol 45381.
Chem 273:205515. 49. Feng X, Takeshita S, Namba N, Wei S, Teitelbaum SL,
34. David J-P, Sabapathy K, Hoffmann O, Idarraga MH, Ross FP (2002) Tyrosines 559 and 807 in the cytoplas-
Wagner EF (2002) JNK1 modulates osteoclastogenesis mic tail of the macrophage colony-stimulating factor
through both c-Jun phosphorylation-dependent and receptor play distinct roles in osteoclast differentiation
-independent mechanisms. J Cell Sci 115:431725. and function. Endocrinology 143:486874.
35. Davies J, Warwick J, Totty N, Philp R, Helfrich M, 50. Fisher JE, Cauleld MP, Sato M, Quartuccio HA, Gould
Horton M (1989) The osteoclast functional antigen, RJ, Garsky VM, et al. (1993) Inhibition of osteoclastic
implicated in the regulation of bone resorption, is bio- bone resorption in vivo by echistatin, an arginyl-
chemically related to the vitronectin receptor. J Cell glycyl-aspartyl (RGD)-containing protein. Endo-
Biol 109:181726. crinology 132:141113.
36. Daws MR, Lanier LL, Seaman WE, Ryan JC (2001) 51. Flores ME, Heinegard D, Reinholt FP, Andersson G
Cloning and characterization of a novel mouse (1996) Bone sialoprotein coated on glass and plastic
myeloid DAP12-associated receptor family. Eur J surfaces is recognized by different b3 integrins. Exp
Immunol 31:78391. Cell Res 227:406.
37. Delaisse JM, Eeckhout Y, Neff L, Francois-Gillet C, 52. Force T, Bonventre JV, Flannery MR, Gorn AH, Yamin
Henriet P, Su Y, et al. (1993) (Pro)collagenase (matrix M, Goldring SR (1992) A cloned porcine renal calci-
metalloproteinase-1) is present in rodent osteoclasts tonin receptor couples to adenylyl cyclase and phos-
and in the underlying bone-resorbing compartment. J pholipase C. Am J Physiol 262:F111015.
Cell Sci 106:107182. 53. Franzoso G, Carlson L, Xing L, Poljak L, Shores EW,
38. Della Rocca GJ, Maudsley S, Daaka Y, Lefkowitz RJ, Brown KD, et al. (1997) Requirement for NF-kB in
Luttrell LM (1999) Pleiotropic coupling of G protein- osteoclast and B-cell development. Genes Dev 11:
coupled receptors to the mitogen- activated protein 348296.
kinase cascade. Role of focal adhesions and receptor 54. Frattini A, Orchard PJ, Sobacchi C, Giliani S, Abinun M,
tyrosine kinases. J Biol Chem 274:1397884. Mattsson JP, et al. (2000) Defects in TCIRG1 subunit of
Regulation of Osteoclast Activity 53

the vacuolar proton pump are responsible for a subset naling (SOCS). The possible counteracting role of
of human autosomal recessive osteopetrosis. Nat Genet SOCSs in IFN-b-inhibited osteoclast formation. J Biol
25:3436. Chem 277:278806.
55. Fuller K, Owens JM, Jagger CJ, Wilson A, Moss R, 72. Helfrich MH, Nesbitt SA, Lakkakorpi PT, Barnes MJ,
Chambers TJ (1993) Macrophage colony-stimulating Bodary SC, Shankar G, et al. (1996) b1 integrins and
factor stimulates survival and chemotactic behavior in osteoclast function: involvement in collagen recogni-
isolated osteoclasts. J Exp Med 178:173344. tion and bone resorption. Bone 19:31728.
56. Galibert L, Tometsko ME, Anderson DM, Cosman D, 73. Holtrop ME, Raisz LG, Simmons HA (1974) The effects
Dougall WC (1998) The involvement of multiple tumor of parathyroid hormone, colchicine, and calcitonin on
necrosis factor receptor (TNFR)-associated factors in the ultrastructure and the activity of osteoclasts in
the signaling mechanisms of receptor activator of NF- organ culture. J Cell Biol 60:34655.
kB, a member of the TNFR superfamily. J Biol Chem 74. Horne WC, Neff L, Chatterjee D, Lomri A, Levy JB,
273:341207. Baron R (1992) Osteoclasts express high levels of
57. Gelb BD, Shi G-P, Chapman HA, Desnick RJ (1996) Pyc- pp60c-src in association with intracellular membranes.
nodysostosis, a lysosomal disease caused by cathepsin J Cell Biol 119:100313.
K deciency. Science 273:12368. 75. Horton MA (1997) The avb3 integrin vitronectin
58. Giancotti FG, Ruoslahti E (1999) Integrin signaling. receptor. Int J Biochem Cell Biol 29:7215.
Science 285:102833. 76. Horton MA, Taylor ML, Arnett TR, Helfrich MH (1991)
59. Gil-Henn H, Elson A (2003) Tyrosine phosphatase-e Arg-Gly-Asp (RGD) peptides and the anti-vitronectin
activates Src and supports the transformed phenotype receptor antibody 23C6 inhibit dentine resorption and
of Neu-induced mammary tumor cells. J Biol Chem cell spreading by osteoclasts. Exp Cell Res 195:368
278:1557986. 75.
60. Gil-Henn H, Volohonsky G, Elson A (2001) Regulation 77. Hotokezaka H, Sakai E, Kanaoka K, Saito K, Matsuo K,
of protein-tyrosine phosphatases a and e by calpain- Kitaura H, et al. (2002) U0126 and PD98059, specic
mediated proteolytic cleavage. J Biol Chem 276: inhibitors of MEK, accelerate differentiation of
317729. RAW264.7 cells into osteoclast-like cells. J Biol Chem
61. Gil-Henn H, Volohonsky G, Toledano-Katchalski H, 277:4736672.
Gandre S, Elson A (2000) Generation of novel cyto- 78. Hruska KA, Rolnick F, Huskey M, Alvarez U, Cheresh
plasmic forms of protein tyrosine phosphatase epsilon D (1995) Engagement of the osteoclast integrin avb3
by proteolytic processing and translational control. by osteopontin stimulates phosphatidylinositol 3-
Oncogene 19:437584. hydroxyl kinase activity. Ann NY Acad Sci 760:151
62. Green MC, Shultz LD (1975) Motheaten, an immunod- 65.
ecient mutant of the mouse. I. Genetics and pathol- 79. Hsu H, Lacey DL, Dunstan CR, Solovyev I, Colombero
ogy. J Hered 66:2508. A, Timms E, et al. (1999) Tumor necrosis factor recep-
63. Grey A, Chen Y, Paliwal I, Carlberg K, Insogna K (2000) tor family member RANK mediates osteoclast differ-
Evidence for a functional association between phos- entiation and activation induced by osteoprotegerin
phatidylinositol 3-kinase and c-src in the spreading ligand. Proc Natl Acad Sci USA 96:35405.
response of osteoclasts to colony-stimulating factor-1. 80. Hunter T (1995) Protein kinases and phosphatases: the
Endocrinology 141:212938. yin and yang of protein phosphorylation and signal-
64. Grigoriadis AE, Wang Z-Q, Cecchini MG, Hofstetter W, ing. Cell 80:22536.
Felix R, Fleisch HA, et al. (1994) c-Fos: a key regulator 81. Insogna KL, Sahni M, Grey AB, Tanaka S, Horne WC,
of osteoclast-macrophage lineage determination and Neff L, et al. (1997) Colony-stimulating factor-1
bone remodeling. Science 266:4438. induces cytoskeletal reorganization and c-src-depen-
65. Hakak Y, Martin GS (1999) Ubiquitin-dependent dent tyrosine phosphorylation of selected cellular
degradation of active Src. Curr. Biol 9:103942. proteins in rodent osteoclasts. J Clin Invest 100:2476
66. Hakola HP (1972) Neuropsychiatric and genetic 85.
aspects of a new hereditary disease characterized by 82. Joazeiro CAP, Wing SS, Huang H-K, Leverson JD,
progressive dementia and lipomembranous polycystic Hunter T, Liu Y-C (1999) The tyrosine kinase negative
osteodysplasia. Acta Psychiatr Scand Suppl 232:1173. regulator c-Cbl as a RING-type, E2- dependent
67. Hall TJ, Chambers TJ (1989) Optimal bone resorption ubiquitin-protein ligase. Science 286:30912.
by isolated rat osteoclasts requires chloride/bicarbon- 83. Kaifu T, Nakahara J, Inui M, Mishima K, Momiyama
ate exchange. Calcif Tissue Int 45:37880. T, Kaji M, et al. (2003) Osteopetrosis and thalamic
68. Hamilton JA (1997) CSF-1 signal transduction. J hypomyelinosis with synaptic degeneration in DAP12-
Leukoc Biol 62:14555. decient mice. J Clin Invest 111:32332.
69. Haque SJ, Harbor P, Tabrizi M, Yi T, Williams BRG 84. Kallio DM, Garant PR, Minkin C (1972) Ultrastructural
(1998) Protein-tyrosine phosphatase Shp-1 is a nega- effects of calcitonin on osteoclasts in tissue culture. J
tive regulator of IL-4- and IL-13-dependent signal Ultrastruct Res 39:20516.
transduction. J Biol Chem 273:338936. 85. Karin M, Cao Y, Greten FR, Li ZW (2002) NF-kB in
70. Hattersley G, Dorey E, Horton MA, Chambers TJ (1988) cancer: from innocent bystander to major culprit. Nat
Human macrophage colony-stimulating factor inhibits Rev Cancer 2:30110.
bone resorption by osteoclasts disaggregated from rat 86. Khaled AR, Butloski EJ, Sobel ES, Schiffenbauer J
bone. J Cell Physiol 137:199203. (1998) Functional consequences of the SHP-1 defect in
71. Hayashi T, Kaneda T, Toyama Y, Kumegawa M, Hakeda motheaten viable mice: role of NF-kB. Cell Immunol
Y (2002) Regulation of receptor activator of NF-kB 185:4958.
ligand-induced osteoclastogenesis by endogenous 87. King KL, DAnza JJ, Bodary S, Pitti R, Siegel M, Lazarus
interferon-b (INF-b) and suppressors of cytokine sig- RA, et al. (1994) Effects of kistrin on bone resorption
54 Bone Resorption

in vitro and serum calcium in vivo. J Bone Miner Res 103. Li L, Dixon JE (2000) Form, function, and regulation of
9:3817. protein tyrosine phosphatases and their involvement
88. Klemke RL, Leng J, Molander R, Brooks PC, Vuori K, in human diseases. Semin Immunol 12:7584.
Cheresh DA (1998) CAS/Crk coupling serves as a mol- 104. Li X, Udagawa N, Itoh K, Suda K, Murase Y, Nishihara
ecular switch for induction of cell migration. J Cell T, et al. (2002) p38 MAPK-mediated signals are
Biol 140:96172. required for inducing osteoclast differentiation but not
89. Klingmuller U, Lorenz U, Cantley LC, Neel BG, Lodish for osteoclast function. Endocrinology 143:310513.
HF (1995) Specic recruitment of SH-PTP1 to the ery- 105. Li Y-P, Chen W, Liang Y, Li E, Stashenko P (1999) Atp6i-
thropoietin receptor causes inactivation of JAK2 and decient mice exhibit severe osteopetrosis due to loss
termination of proliferative signals. Cell 80:72938. of osteoclast-mediated extracellular acidication. Nat
90. Kobayashi N, Kadono Y, Naito A, Matumoto K, Genet 23:44751.
Yamamoto T, Tanaka S, et al. (2001) Segregation of 106. Lomaga MA, Yeh W-C, Sarosi I, Duncan GS, Furlonger
TRAF6-mediated signaling pathways claries its role C, Ho A, et al. (1999) TRAF6 deciency results in
in osteoclastogenesis. EMBO J 20:127180. osteopetrosis and defective interleukin-1, CD40, and
91. Kornak U, Kasper D, Bosl MR, Kaiser E, Schweizer M, LPS signaling. Genes Dev 13:101524.
Schulz A, et al. (2001) Loss of the ClC-7 chloride 107. Lorenz U, Ravichandran KS, Burakoff SJ, Neel BG
channel leads to osteopetrosis in mice and man. Cell (1996) Lack of SHPTP1 results in src-family kinase
104:20515. hyperactivation and thymocyte hyperresponsiveness.
92. Kornak U, Schulz A, Friedrich W, Uhlhaas S, Kremens Proc Natl Acad Sci USA 93:96249.
B,Voit T, et al. (2000) Mutations in the a3 subunit of the 108. Lowell CA, Niwa M, Soriano P, Varmus HE (1996) De-
vacuolar H(+)-ATPase cause infantile malignant ciency of the Hck and Src tyrosine kinases results in
osteopetrosis. Hum Mol Genet 9:205963. extreme levels of extramedullary hematopoiesis. Blood
93. Krueger NX, Streuli M, Saito H (1990) Structural diver- 87:178092.
sity and evolution of human receptor-like protein tyro- 109. Ma YL, Cain RL, Halladay DL, Yang X, Zeng Q, Miles
sine phosphatases. EMBO J 9:324152. RR, et al. (2001) Catabolic effects of continuous human
94. Lacey DL, Timms E, Tan H-L, Kelley MJ, Dunstan CR, PTH (138) in vivo is associated with sustained stim-
Burgess T, et al. (1998) Osteoprotegerin ligand is a ulation of RANKL and inhibition of osteoprotegerin
cytokine that regulates osteoclast differentiation and and gene-associated bone formation. Endocrinology
activation. Cell 93:16576. 142:404754.
95. Lakkakorpi PT, Nakamura I, Nagy RM, Parsons JT, 110. Mansky KC, Sankar U, Han J, Ostrowski MC (2002)
Rodan GA, Duong LT (1999) Stable association of Microphthalmia transcription factor is a target of the
PYK2 and p130Cas in osteoclasts and their co- p38 MAPK pathway in response to receptor activator
localization in the sealing zone. J Biol Chem 274:4900 of NF-kB ligand signaling. J Biol Chem 277:1107783.
7. 111. Masarachia P, Yamamoto M, Leu CT, Rodan G, Duong
96. Lakkakorpi PT, Nakamura I, Young M, Lipfert L, Rodan L (1998) Histomorphometric evidence for echistatin
GA, Duong LT (2001) Abnormal localisation and inhibition of bone resorption in mice with secondary
hyperclustering of aVb3 integrins and associated pro- hyperparathyroidism. Endocrinology 139:140110.
teins in Src-decient or tyrphostin A9-treated osteo- 112. Matsumoto M, Sudo T, Saito T, Osada H, Tsujimoto M
clasts. J Cell Sci 114:14960. (2000) Involvement of p38 mitogen-activated protein
97. Lakkakorpi PT, Vaananen HK (1991) Kinetics of the kinase signaling pathway in osteoclastogenesis medi-
osteoclast cytoskeleton during the resorption cycle in ated by receptor activator of NF-kB ligand (RANKL). J
vitro. J Bone Miner Res 6:81726. Biol Chem 275:3115561.
98. Lakkakorpi PT, Wesolowski G, Zimolo Z, Rodan GA, 113. Matthews RJ, Bowne DB, Flores E, Thomas ML (1992)
Rodan SB (1997) Phosphatidylinositol 3-kinase associ- Characterization of hematopoietic intracellular
ation with the osteoclast cytoskeleton, and its involve- protein tyrosine phosphatases: description of a phos-
ment in osteoclast attachment and spreading. Exp Cell phatase containing an SH2 domain and another
Res 237:296306. enriched in proline-, glutamic acid-, serine-, and
99. Lee PSW, Wang Y, Dominguez MG, Yeung Y-G, Murphy threonine-rich sequences. Mol Cell Biol 12:2396
MA, Bowtell DDL, et al. (1999) The Cbl protooncopro- 405.
tein stimulates CSF-1 receptor multiubiquitination and 114. Mauro LJ, Olmsted EA, Davis AR, Dixon JE (1996)
endocytosis, and attenuates macrophage proliferation. Parathyroid hormone regulates the expression of the
EMBO J 18:361628. receptor protein tyrosine phosphatase, OST-PTP, in rat
100. Lee SW, Han SI, Kim HH, Lee ZH (2002) TAK1- osteoblast-like cells. Endocrinology 137:92533.
dependent activation of AP-1 and c-Jun N-terminal 115. Mauro LJ, Olmsted EA, Skrobacz BM, Mourey RJ, Davis
kinase by receptor activator of NF-kB. J Biochem Mol AR, Dixon JE (1994) Identication of a hormonally
Biol 35:3716. regulated protein tyrosine phosphatase associated
101. Levkowitz G, Waterman H, Ettenberg SA, Katz M, with bone and testicular differentiation. J Biol Chem
Tsygankov AY, Alroy I, et al. (1999) Ubiquitin ligase 269:3065967.
activity and tyrosine phosphorylation underlie sup- 116. McHugh KP, Hodivala-Dilke K, Zheng M-H, Namba N,
pression of growth factor signaling by c-Cbl/Sli-1. Mol Lam J, Novack D, et al. (2000) Mice lacking b3 integrins
Cell 4:102940. are osteosclerotic because of dysfunctional osteoclasts.
102. Li J, Sarosi I, Yan X-Q, Morony S, Capparelli C, Tan J Clin Invest 105:43340.
H-L, et al. (2000) RANK is the intrinsic hematopoietic 117. Miyazaki T, Katagiri H, Kanegae Y, Takayanagi H,
cell surface receptor that controls osteoclastogenesis Sawada Y, Yamamoto A, et al. (2000) Reciprocal role of
and regulation of bone mass and calcium metabolism. ERK and NF-kB pathways in survival and activation of
Proc Natl Acad Sci USA 97:156671. osteoclasts. J Cell Biol 148:33342.
Regulation of Osteoclast Activity 55

118. Miyazaki T, Neff L, Tanaka S, Horne WC, Baron R 134. Paloneva J, Manninen T, Christman G, Hovanes K,
(2003) Regulation of cytochrome c oxidase activity by Mandelin J, Adolfsson R, et al. (2002) Mutations in two
c-Src in osteoclasts. J Cell Biol 160:70918. genes encoding different subunits of a receptor signal-
119. Mizukami J, Takaesu G, Akatsuka H, Sakurai H, ing complex result in an identical disease phenotype.
Ninomiya-Tsuji J, Matsumoto K, et al. (2002) Receptor Am J Hum Genet 71:65662.
activator of NF-kB ligand (RANKL) activates TAK1 135. Paniccia R, Riccioni T, Zani BM, Zigrino P, Scotlandi K,
mitogen-activated protein kinase kinase kinase Teti A (1995) Calcitonin down-regulates immediate
through a signaling complex containing RANK, TAB2, cell signals induced in human osteoclast-like cells by
and TRAF6. Mol Cell Biol 22:9921000. the bone sialoprotein-IIA fragment through a postin-
120. Moller NPH, Moller KB, Lammers R, Kharitonenkov A, tegrin receptor mechanism. Endocrinology 136:1177
Hoppe E, Wiberg FC, et al. (1995) Selective down- 86.
regulation of the insulin receptor signal by protein- 136. Plutzky J, Neel BG, Rosenberg RD (1992) Isolation of a
tyrosine phosphatases a and e. J Biol Chem 270: src homology 2-containing tyrosine phosphatase. Proc
2312631. Natl Acad Sci USA 89:11237.
121. Nakamura I, Jimi E, Duong LT, Sasaki T, Takahashi N, 137. Plutzky J, Neel BG, Rosenberg RD, Eddy RL, Byers MG,
Rodan GA, et al. (1998) Tyrosine phosphorylation of Jani-Sait S, et al. (1992) Chromosomal localization of
p130Cas is involved in actin organization in osteoclasts. an SH2-containing tyrosine phosphatase (PTPN6).
J Biol Chem 273:111449. Genomics 13:86972.
122. Nakamura I, Lipfert L, Rodan GA, Duong LT (2001) 138. Quinn JMW, Elliott J, Gillespie MT, Martin TJ (1998) A
Convergence of avb3 integrin- and macrophage combination of osteoclast differentiation factor and
colony stimulating factor-mediated signals on phos- macrophage-colony stimulating factor is sufcient for
pholipase Cg in prefusion osteoclasts. J Cell Biol 152: both human and mouse osteoclast formation in vitro.
36173. Endocrinology 139:44247.
123. Nakamura I, Pilkington MF, Lakkakorpi PT, Lipfert L, 139. Rajapurohitam V, Chalhoub N, Benachenhou N, Neff L,
Sims SM, Dixon SJ, et al. (1999) Role of avb3 integrin in Baron R, Vacher J (2001) The mouse osteopetrotic
osteoclast migration and formation of the sealing grey-lethal mutation induces a defect in osteoclast
zone. J Cell Sci 112:398593. maturation/function. Bone 28:51323.
124. Nakamura I, Takahashi N, Sasaki T, Jimi E, Kurokawa 140. Saftig P, Hunziker E, Wehmeyer O, Jones S, Boyde A,
T, Suda T (1996) Chemical and physical properties of Rommerskirch W, et al. (1998) Impaired osteoclastic
the extracellular matrix are required for the actin ring bone resorption leads to osteopetrosis in cathepsin-K-
formation in osteoclasts. J Bone Miner Res 11:18739. decient mice. Proc Natl Acad Sci USA 95:13453
125. Nakamura I, Takahashi N, Sasaki T, Tanaka S, Udagawa 8.
N, Murakami H, et al. (1995) Wortmannin, a specic 141. Salo J, Lehenkari P, Mulari M, Metsikko K, Vaananen
inhibitor of phosphatidylinositol-3 kinase, blocks HK (1997) Removal of osteoclast bone resorption
osteoclastic bone resorption. FEBS Lett 361:7984. products by transcytosis. Science 276:2703.
126. Nakamura I, Tanaka H, Rodan GA, Duong LT (1998) 142. Sanjay A, Horne WC, Baron R (2001) The Cbl family:
Echistatin inhibits the migration of murine prefusion ubiquitin ligases regulating signaling by tyrosine
osteoclasts and the formation of multinucleated osteo- kinases. Sciences STKE. http://stke.sciencemag.org/
clast-like cells. Endocrinology 139:518293. cgi/content/full/OC_sigtrans;2001/110/pe40.
127. Nakamura K, Mizuno Y, Kikuchi K (1996) Molecular 143. Sanjay A, Houghton A, Neff L, Didomenico E, Bardelay
cloning of a novel cytoplasmic protein tyrosine phos- C, Antoine E, et al. (2001) Cbl associates with Pyk2
phatase PTP e. Biochem Biophys Res Commun 218: and Src to regulate Src kinase activity, avb3 integrin-
72632. mediated signaling, cell adhesion, and osteoclast
128. Naro F, Perez M, Migliaccio S, Galson DL, Orcel P, Teti motility. J Cell Biol 152:18195.
A, et al. (1998) Phospholipase D- and protein kinase C 144. Sato M, Sardana MK, Grasser WA, Garsky VM, Murray
isoenzyme-dependent signal transduction pathways JM, Gould RJ (1990) Echistatin is a potent inhibitor
activated by the calcitonin receptor. Endocrinology of bone resorption in culture. J Cell Biol 111:1713
139:32418. 23.
129. Nesbitt S, Nesbit A, Helfrich M, Horton M (1993) Bio- 145. Schlaepfer DD, Broome MA, Hunter T (1997)
chemical characterization of human osteoclast inte- Fibronectin-stimulated signaling from a focal adhe-
grins. Osteoclasts express avb3, a2b1, and avb1 sion kinase-c-Src complex: involvement of the Grb2,
integrins. J Biol Chem 268:1673745. p130cas, and Nck adaptor proteins. Mol Cell Biol
130. Nesbitt SA, Horton MA (1997) Trafcking of matrix 17:170213.
collagens through bone-resorbing osteoclasts. Science 146. Schlaepfer DD, Hauck CR, Sieg DJ (1999) Signaling
276:2669. through focal adhesion kinase. Prog Biophys Mol Biol
131. Ota J, Sato K, Kimura F, Wakimoto N, Nakamura Y, 71:435378.
Nagata N, et al. (2000) Association of Cbl with Fms and 147. Schlessinger J (2000) New roles for Src kinases in
p85 in response to macrophage colony-stimulating control of cell survival and angiogenesis. Cell 100:
factor. FEBS Lett 466:96100. 2936.
132. Palacio S, Felix R (2001) The role of phosphoinositide 148. Schwartzberg PL, Xing L, Hoffmann O, Lowell CA,
3-kinase in spreading osteoclasts induced by colony- Garrett L, Boyce BF, et al. (1997) Rescue of osteoclast
stimulating factor-1. Eur J Endocrinol 144:43140. function by transgenic expression of kinase-decient
133. Paloneva J, Kestila M, Wu J, Salminen A, Bohling T, Src in src-/- mutant mice. Genes Dev 11:283544.
Ruotsalainen V, et al. (2000) Loss-of-function muta- 149. Scimeca JC, Franchi A, Trojani C, Parrinello H,
tions in TYROBP (DAP12) result in a presenile demen- Grosgeorge J, Robert C, et al. (2000) The gene encod-
tia with bone cysts. Nat Genet 25:35761. ing the mouse homologue of the human osteoclast-
56 Bone Resorption

specic 116-kDa V-ATPase subunit bears a deletion in 164. Takayanagi H, Kim S, Matsuo K, Suzuki H, Suzuki T,
osteosclerotic (oc/oc) mutants. Bone 26:20713. Sato K, et al. (2002) RANKL maintains bone home-
150. Shaw LM, Rabinovitz I, Wang HH-F, Toker A, Mercurio ostasis through c-Fos-dependent induction of inter-
AM (1997) Activation of phosphoinositide 3-OH feron-b. Nature 416:7449.
kinase by the a6b4 integrin promotes carcinoma inva- 165. Takayanagi H, Ogasawara K, Hida S, Chiba T, Murata S,
sion. Cell 91:94960. Sato K, et al. (2000) T-cell-mediated regulation of
151. Shen S-H, Bastien L, Posner BI, Chretien P (1991) A osteoclastogenesis by signalling cross- talk between
protein-tyrosine phosphatase with sequence similarity RANKL and IFN-g. Nature 408:6005.
to the SH2 domain of the protein-tyrosine kinases. 166. Takeshita S, Namba N, Zhao JJ, Jiang Y, Genant HK,
Nature 352:7369. Silva MJ, et al. (2002) SHIP-decient mice are severely
152. Shui C, Riggs BL, Khosla S (2002) The Immunosup- osteoporotic due to increased numbers of hyper-
pressant rapamycin, alone or with transforming resorptive osteoclasts. Nat Med 8:9439.
growth factor-b, enhances osteoclast differentiation of 167. Tanaka S, Amling M, Neff L, Peyman A, Uhlmann E,
RAW264.7 monocyte-macrophage cells in the presence Levy JB, et al. (1996) c-Cbl is downstream of c-Src in a
of RANK-ligand. Calcif Tissue Int 71:43746. signalling pathway necessary for bone resorption.
153. Shultz LD, Schweitzer PA, Rajan TV, Yi T, Ihle JN, Nature 383:52831.
Matthews RJ, et al. (1993) Mutations at the murine 168. Tanaka S, Neff L, Baron R, Levy JB (1995) Tyrosine
motheaten locus are within the hematopoietic cell phosphorylation and translocation of the c-Cbl
protein-tyrosine phosphatase (Hcph) gene. Cell 73: protein after activation of tyrosine kinase signaling
144554. pathways. J Biol Chem 270:1434751.
154. Shultz LD, Sidman CL (1987) Genetically determined 169. Tanuma N, Nakamura K, Kikuchi K (1999) Distinct
murine models of immunodeciency. Annu Rev promoters control transmembrane and cytosolic
Immunol 5:367403. protein tyrosine phosphatase e expression during
155. Shyu J-F, Inoue D, Baron R, Horne WC (1996) The dele- macrophage differentiation. Eur J Biochem 259:4654.
tion of 14 amino acids in the seventh transmembrane 170. Tanuma N, Nakamura K, Shima H, Kikuchi K (2000)
domain of a naturally occurring calcitonin receptor Protein-tyrosine phosphatase PTPeC inhibits Jak-
isoform alters ligand binding and selectively abolishes STAT signaling and differentiation induced by inter-
coupling to phospholipase C. J Biol Chem 271:31127 leukin-6 and leukemia inhibitory factor in M1
34. leukemia cells. J Biol Chem 275:2821621.
156. Shyu J-F, Zhang Z, Hernandez-Lagunas L, Camerino C, 171. Tanuma N, Shima H, Nakamura K, Kikuchi K (2001)
Chen Y, Inoue D, et al. (1999) Protein kinase C antago- Protein tyrosine phosphatase eC selectively inhibits
nizes pertussis-toxin-sensitive coupling of the calci- interleukin-6- and interleukin- 10-induced JAK-STAT
tonin receptor to adenylyl cyclase. Eur J Biochem signaling. Blood 98:30304.
262:95101. 172. Tanuma N, Shima H, Shimada S, Kikuchi K (2003)
157. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang Reduced tumorigenicity of murine leukemia cells
M-S, Luthy R, et al. (1997) Osteoprotegerin: a novel expressing protein-tyrosine phosphatase, PTPeC.
secreted protein involved in the regulation of bone Oncogene 22:175862.
density. Cell 89:30919. 173. Teitelbaum SL (2000) Bone resorption by osteoclasts.
158. Sims NA, Aoki K, Bogdanovich Z, Maragh M, Okigaki Science 289:15048.
M, Logan S, et al. (1999) Impaired osteoclast function 174. Thien CBF, Langdon WY (2001) Cbl: many adaptations
in Pyk2 knockout mice and cumulative effects in to regulate protein tyrosine kinases. Nat Rev Mol Cell
Pyk2/Src double knockout. J Bone Miner Res 14 (Suppl. Biol 2:294305.
1):S183. 175. Thomas SM, Brugge JS (1997) Cellular functions regu-
159. Sly WS, Hewett-Emmett D, Whyte MP, Yu Y-S, Tashian lated by Src family kinases. Annu. Rev Cell Dev Biol
RE (1983) Carbonic anhydrase II deciency identied 13:513609.
as the primary defect in the autosomal recessive syn- 176. Toledano-Katchalski H, Kraut J, Sines T, Granot-Attas
drome of osteopetrosis with renal tubular acidosis and S, Shohat G, Gil-Henn H, et al. (2003) Protein tyrosine
cerebral calcication. Proc Natl Acad Sci USA 80: phosphatase e inhibits signaling by mitogen-activated
27526. protein kinases. Mol Cancer Res 1:54150.
160. Soriano P, Montgomery C, Geske R, Bradley A (1991) 177. Tomic S, Greiser U, Lammers R, Kharitonenkov A,
Targeted disruption of the c-src proto-oncogene leads Imyanitov E, Ullrich A, et al. (1995) Association of
to osteopetrosis in mice. Cell 64:693702. SH2 domain protein tyrosine phosphatases with the
161. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, epidermal growth factor receptor in human tumor
Martin TJ (1999) Modulation of osteoclast differentia- cells. Phosphatidic acid activates receptor dephos-
tion and function by the new members of the tumor phorylation by PTP1C. J Biol Chem 270:2127784.
necrosis factor receptor and ligand families. Endocr 178. Tonks NK, Neel BG (2001) Combinatorial control of the
Rev 20:34557. specicity of protein tyrosine phosphatases. Curr Opin
162. Suhr SM, Pamula S, Baylink DJ, Lau K-HW (2001) Anti- Cell Biol 13:18295.
sense oligodeoxynucleotide evidence that a unique 179. Umeda S, Beamer WG, Takagi K, Naito M, Hayashi S-I,
osteoclastic protein-tyrosine phosphatase is essential Yonemitsu H, et al. (1999) Deciency of SHP-1 protein-
for osteoclastic resorption. J Bone Miner Res 16: tyrosine phosphatase activity results in heightened
17951803. osteoclast function and decreased bone density. Am J
163. Sully V, Pownall S, Vincan E, Bassal S, Borowski AH, Pathol 155:22333.
Hart PH, et al. (2001) Functional abnormalities in 180. Vaananen HK, Karhukorpi EK, Sundquist K, Roininen
protein tyrosine phosphatase e-decient macrophages. I, Hentunen T, Tuukkanen J, et al. (1990) Evidence for
Biochem Biophys Res Commun 286:1848. the presence of a proton pump of the vacuolar H+-
Regulation of Osteoclast Activity 57

ATPase type in the rufed border of osteoclasts. J Cell induced by receptor activator of nuclear factor kB
Biol 111:130511. ligand. J Bone Miner Res 17:61221.
181. Vaananen HK, Zhao H, Mulari M, Halleen JM (2000) 194. Yamamoto M, Fisher JE, Gentile M, Seedor JG, Leu
The cell biology of osteoclast function. J Cell Sci 113: C-T, Rodan SB, et al. (1998) The integrin ligand echis-
37781. tatin prevents bone loss in ovariectomized mice and
182. Vuori K, Hirai H, Aizawa S, Ruoslahti E (1996) Intro- rats. Endocrinology 139:141119.
duction of p130cas signaling complex formation upon 195. Yan T, Riggs BL, Boyle WJ, Khosla S (2001) Regulation
integrin-mediated cell adhesion: a role for Src family of osteoclastogenesis and RANK expression by TGF-
kinases. Mol Cell Biol 16:260613. b1. J Cell Biochem 83:3205.
183. Wabakken T, Hauge H, Finne EF,Wiedlocha A,Aasheim 196. Yi T, Ihle JN (1993) Association of hematopoietic cell
H (2002) Expression of human protein tyrosine phosphatase with c-Kit after stimulation with c-Kit
phosphatase epsilon in leucocytes: a potential ERK ligand. Mol Cell Biol 13:33508.
pathway-regulating phosphatase. Scand J Immunol 56: 197. Yi T, Mui AL, Krystal G, Ihle JN (1993) Hematopoietic
195203. cell phosphatase associates with the interleukin-3
184. Wei S, Wang MW-H, Teitelbaum SL, Ross FP (2002) (IL-3) receptor beta chain and down-regulates IL-3-
Interleukin-4 reversibly inhibits osteoclastogenesis via induced tyrosine phosphorylation and mitogenesis.
inhibition of NF-kB and mitogen-activated protein Mol Cell Biol 13:757786.
kinase signaling. J Biol Chem 277:662230. 198. Yi TL, Cleveland JL, Ihle JN (1992) Protein tyrosine
185. Weinreb M, Halperin D (1998) Rat osteoclast precur- phosphatase containing SH2 domains: characteriza-
sors in vivo express a vitronectin receptor and a tion, preferential expression in hematopoietic cells,
chloride-bicarbonate exchanger. Connect Tissue Res and localization to human chromosome 12p12-p13.
37:17782. Mol Cell Biol 12:83646.
186. Wennerberg K, Lohikangas L, Gullberg D, Pfaff M, 199. Yokouchi M, Kondo T, Houghton A, Bartkiewicz M,
Johansson S, Fassler R (1996) b1 integrin-dependent Horne WC, Zhang H, et al. (1999) Ligand-induced
and -independent polymerization of bronectin. J Cell ubiquitination of the epidermal growth factor recep-
Biol 132:22738. tor involves the interaction of the c-Cbl RING nger
187. Witke W, Sharpe AH, Hartwig JH, Azuma T, Stossel TP, and UbcH7. J Biol. Chem 274:3170712.
Kwiatkowski DJ (1995) Hemostatic, inammatory, and 200. Yu Z, Su L, Hoglinger O, Jaramillo ML, Banville D, Shen
broblast responses are blunted in mice lacking gel- SH (1998) SHP-1 associates with both platelet-derived
solin. Cell 81:4151. growth factor receptor and the p85 subunit of
188. Wong BR, Besser D, Kim N, Arron JR, Vologodskaia M, phosphatidylinositol 3-kinase. J Biol Chem 273:3687
Hanafusa H, et al. (1999) TRANCE, a TNF family mem- 9364.
ber, activates Akt/PKB through a signaling complex 201. Zaidi M, Datta HK, Moonga BS, MacIntyre I (1990) Evi-
involving TRAF6 and c-Src. Mol Cell 4:10419. dence that the action of calcitonin on rat osteoclasts is
189. Wu C, Hughes PE, Ginsberg MH, McDonald JA (1996) mediated by two G proteins acting via separate post-
Identication of a new biological function for the inte- receptor pathways. J Endocrinol 126:47381.
grin avb3: initiation of bronectin matrix assembly. 202. Zhang X, Chattopadhyay A, Ji QS, Owen JD, Ruest PJ,
Cell Adhes Commun 4:14958. Carpenter G, et al. (1999) Focal adhesion kinase pro-
190. Wu H, Byrne MH, Stacey A, Goldring MB, Birkhead JR, motes phospholipase C-g1 activity. Proc Natl Acad Sci
Jaenisch R, et al. (1990) Generation of collagenase- USA 96:90216.
resistant collagen by site-directed mutagenesis of 203. Zhang Z, Baron R, Horne WC (2000) Integrin engage-
murine proa1(I) collagen gene. Proc Natl Acad Sci USA ment, the actin cytoskeleton, and c-Src are required for
87:588892. the calcitonin-induced tyrosine phosphorylation of
191. Wu L-W, Baylink DJ, Lau K-H (1996) Molecular cloning paxillin and HEF1, but not for calcitonin-induced
and expression of a unique rabbit osteoclastic phos- Erk1/2 phosphorylation. J Biol Chem 275:3721923.
photyrosyl phosphatase. Biochem J 316:51523. 204. Zhang Z, Hernandez-Lagunas L, Horne WC, Baron R
192. Xing L, Bushnell TP, Carlson L, Tai Z, Tondravi M, (1999) Cytoskeleton-dependent tyrosine phosphoryla-
Siebenlist U, et al. (2002) NF-kB p50 and p52 expres- tion of the maily member HEF1 downstream of the G
sion is not required for RANK-expressing osteoclast protein-couopled calcitonin receptor. Calcitonin
progenitor formation but is essential for RANK- and induces the association of HEF1, paxillin, and focal
cytokine-mediated osteoclastogenesis. J Bone Miner adhesion of kinase. J Biol Chem 274:250938.
Res 17:120010. 205. Zimolo Z, Weolowski G, Tanaka H, Hyman JL, Hoyer
193. Yamamoto A, Miyazaki T, Kadono Y, Takayanagi H, JR, Rodan GA (1994) Soluble avb3-integrin ligands
Miura T, Nishina H, et al. (2002) Possible involvement raise [Ca2+]I in rat osteoclasts and mouse-derived
of IkB kinase 2 and MKK7 in osteoclastogenesis osteoclast-like cells. Am J Physiol 266:C37681.
4.
Structural Aspects of Bone Resorption
Steven D. Bain and Ted S. Gross

focus on the osteoclastic behaviors that modu-


Introduction late bone mass and architecture.

The skeleton is a metabolically active organ Resorption of Cortical Bone


system, comprised of tissue-specic cells and an
extracellular matrix that undergoes continuous Bone remodeling in the adult skeleton is the
removal and replacement throughout life. At a process whereby bone matrix is rst removed by
fundamental level, the removal and replacement osteoclasts and then replaced by osteoblasts.
of the extracellular matrix is necessary to estab- The cellular basis of the remodeling cycle was
lish and maintain bone mass and architecture. rst described by Frost, who named this phy-
However, the removal and replacement of the siologic relationship the basic multicellular
matrix is regulated within a physiologic frame- unit or the BMU [26, 28]. Known also as a bone
work that requires the bone tissue to function remodeling unit or BRU [70], the BMU
simultaneously in locomotion, as support and describes the sequential activities of osteoclasts
site of muscle attachment, as a protector of vital and osteoblasts that are spatially and temporally
internal organs, and as a storehouse of calcium coupled to ensure that the removal of mineral-
and phosphorus ions. Interestingly, these ized matrix is replaced by an equivalent quan-
diverse structural and metabolic functions are tity of new bone [27, 48, 69]. Thus, over
principally driven by the interplay between just innumerable remodeling cycles the collective
two cell types; the bone-resorbing osteoclast activities of individual BMUs ensure that an
and the bone-forming osteoblast. Moreover, it organisms bone mass remains in constant
is recognized that the interplay between osteo- balance and its skeletal architecture relatively
clast and osteoblast must be nely balanced as unchanged [29, 43, 71].
even minor disturbances in the resorbing and It is now evident from investigations of the
forming activities of these cell types can lead to bone remodeling cycle that the skeletons mass
both structural and metabolic pathologies. and its corresponding architecture are depen-
From a structural perspective, it is the osteo- dent on the coordinated activity of the BMU,
clast that is responsible for initiating the bone which is commonly separated into four distinct
renewal process, which in the adult skeleton is phases: activation, resorption, reversal, and
termed bone remodeling. Thus, it is only by rst formation [71, 85, 93, 94]. In both cortical and
removing preexisting bone matrix that the cancellous bone, the BMU life cycle begins with
tissue is able to alter its shape and morphology the activation of a quiescent (i.e., resting) bone
in response to the demands engendered by loco- surface in response to an enabling stimulus
motor and/or metabolic stimuli. The objective that triggers differentiation and recruitment
of this chapter is to provide an overview of the of osteoclast precursors, which fuse to form
structural aspects of this process with a specic mature, multinucleated, bone-resorbing osteo-
58
Structural Aspects of Bone Resorption 59

clasts (for review see [91]). The multinucleated lamellar structure that is termed a bone struc-
osteoclast then attaches to and transits over the tural unit or BSU [36]. This structure is also
bone surface, dissolving and resorbing the min- known as Haversian canal or a secondary osteon.
eralized matrix. In this fashion, groups of osteo- Studies of the temporal nature of osteoclastic
clasts are recruited to the resorption site until activity have shown that the time to complete
a discrete packet or quantum of bone matrix resorption at one BMU level (i.e., the resorption
is removed [28, 30, 36, 40, 69, 71]. As the osteo- sigma), is 915 days, depending on the species
clastic activity subsides, a resorption cavity, [70]. This time period is roughly equivalent to
termed a Howships lacunae, is formed [35]. the 1012-day life span of individual osteoclasts
Within this anatomic framework the struc- [38], which at a longitudinal resorption rate of
tural outcomes that result from bone resorption 35 mm per day would burrow several hundred
are more difcult to assess than those arising mm through the cortex. However, as the com-
from bone formation, simply because the osteo- pleted BSU is from 26 mm in length, it is
clast behaviors must be inferred from the evident that the intracortical progression of
anatomic changes that result from osteoclast bone remodeling requires the continual recruit-
resorption, whereas osteoblast activity creates ment and mobilization of osteoclasts. This
a mineralized record that can be measured recruitment has been substantiated by autora-
directly [16, 20, 70, 75]. This limitation not with- diographic studies conducted in dogs which
standing, reconstruction of the BMU in cortical have shown that the entry of new nuclei into the
bone (Figure 4.1) has provided the basis for osteoclast population is approximately 8% per
quantifying the spatial and temporal behavior day [37]. Thus, when the life span and resorp-
of osteoclasts in bone resorption [48, 76, 77]. tion rates of individual osteoclasts are superim-
Spatially, the activation of the osteoclast posed over the spatial construct of a cortical
machinery in cortical bone populates a resorp- bone BMU it becomes possible to dene a struc-
tive front that tunnels through the cortex. The ture that travels roughly 4,000 mm through the
apex of this structure is termed the cutting cone cortex at about 20 mm per day, with a life span
[44]. Data from studies in non-human primates of 200 days [79].
[70, 88] and canines [39] indicate that osteo- When considering the impact that normal
clasts are able to tunnel longitudinally at rates Haversian remodeling has on cortical bone
of 3540 mm per day. Furthermore, as the structure it can be assumed that the process of
resorption front proceeds through the cortex it bone resorption is ultimately governed by the
expands radially at a rate of about 67 mm per physiologic purposes of bone remodeling [76].
day, which reects the matrix resorbing capac- Bone remodeling replaces old bone with new, a
ity of the individual osteoclast [20, 70]. These stochastic process that prevents mean bone age
data are similar to those reported in human from exceeding an acceptable level [5, 56, 74,
subjects [70]. The resorptive tunnel excavated 79, 84], and is a directed process that removes
by the osteoclasts is eventually lled in by osteo- bone matrix that is micro- or macroscopically
blastic bone formation, producing a mature damaged [4, 7, 25, 53, 62], or has been function-

Figure 4.1 Diagram of a longitudinal


section of a cortical BMU or Haversian
system. The cortical BMU moves toward
the left behind the resorptive front which
is termed the cutting cone. By covering
the upper half of the diagram it is possi-
ble to visualize the structure of the BMU
in cancellous bone. (Reproduced from
Baron R 2003 General Principles of Bone
Biology. In Favus M (ed.) Primer on the
Metabolic Bone Diseases and Disorders
of Mineral Metabolism, 5th ed. American
Society for Bone and Mineral Research,
Washington DC, USA, pp. 18 with per-
mission of the Amercian Society for Bone
and Mineral Research)
60 Bone Resorption

ally suboptimal [14, 50, 51, 66, 86, 95]. Regard-


less of the remodeling stimuli, the amount of
normal adult skeleton undergoing intracortical
remodeling at any point in time is about 5% 1
[70]. Therefore, given the evolutionary success
of terrestrial vertebrates, it is likely that this 2
underlying level of remodeling (and by infer-
ence, resorptive activity) has been genetically
programmed to maintain the skeletons struc-
tural competence [77].
3
Within this evolutionary context, Martin has
proposed that stochastic osteoclast tunneling
would have the effect of frequently replacing
osteocytes so that the homeostatic function of
the osteocyte network remains intact [58].
Furthermore, targeted osteoclastic resorption
would not only serve to remove microdamage 4
[6, 79], but also the new ber-matrix structure
created by the BMU. It is clear that multiple
factors are involved in selecting for osteoclast Figure 4.2 Two-dimensional representation of the life cycle
behaviors that are structurally benecial [79] stages of the BMU on a cancellous bone surface. Activation of a
and that these behaviors need not be 100% ef- resting surface (1) results in the recruitment and activation of
cient to repair tissue. Indeed, Partt argues con- bone resorbing osteoclasts, which work in teams to resorb bone
vincingly that most remodeling is redundant, matrix (2). Osteoclastic resorption subsides during the reversal
phase (3) at which time osteoblasts are recruited to refill the
surplus, spare or stochastic [78], as evidenced resorption cavity with organic matrix (4) that subsequently
by the lack of harm caused by low bone turnover mineralizes, returning the remodeling site to the resting or
in hypothyroid [22] and hypoparathyroid states quiescent phase. (Reproduced from Baron R 2003 General
[49]. Conversely, it is important to note that Principles of Bone Biology. In Favus M (ed.) Primer on the Meta-
recent experimental evidence indicates that bis- bolic Bone Diseases and Disorders of Mineral Metabolism, 5th
phosphonates can suppress both targeted and ed. American Society for Bone and Mineral Research, Washing-
non-targeted remodeling, which may lead to the ton DC, USA, pp. 18 with permission of the Amercian Society
accumulation of microdamage [54]. However, for Bone and Mineral Research.)
the extent to which the suppression of re-
modeling by therapeutic agents actually clini-
cally compromises the skeletons functional role However, such representations ignore the
remains to be elucidated. two-dimensional organization of cancellous
BMUs and promote the mistaken concept that
osteoclasts and osteoblasts are never present at
Resorption of Cancellous Bone the same BMU simultaneously [82]. In actuality,
the cancellous BMU is hemi-osteonal in nature
Because of its solid nature, the replacement of [31, 76], with the principal difference that rather
cortical bone occurs only at sites where bone than boring a cylindrical tunnel through the
has been resorbed by osteoclasts recruited for cortex (as depicted in Figure 4.1) the osteoclasts
that purpose. This physical constraint does not resorb a trench-like structure across the bones
apply to cancellous or cortical endosteal sur- surface (Figure 4.3) [15]. The hemi-osteonal
faces which are surrounded by open spaces model of the cancellous BMU has the added
containing both osteoclast and osteoblast advantage of depicting the cellular relationships
progenitors, but there is little doubt that resorp- that exist within the remodeling space, which
tion and formation in cancellous bone are spa- allows for a more accurate visualization of how
tially and temporally coupled as well [2729, 68, these cells might communicate in vivo [76].
69]. This relationship can be depicted in the Reconstruction of the resorptive phase in
classic up-down or life-cycle diagram repre- cancellous bone has revealed at least four dis-
sented in Figure 4.2, which faithfully reects the tinct variables associated with bone resorption:
sequential nature of BMU activities at discrete erosion depth, active erosion period, erosion
two-dimensional locations. rate, and bone resorption rate [20]. Of these
Structural Aspects of Bone Resorption 61

Figure 4.3 A diagrammatic represen-


tation of the cancellous BMU based on a
histomorphometric reconstruction that
shows the duration and depth of the
phases of the bone remodeling sequence.
(Reproduced with permission form
Eriksen EF, Axelrod DW, Melsen F. Bone
Histomorphometry. Lippincott Williams
& Wilkins. 1994, 7.)

Table 4.1. Methodology for measuring cancellous bone resorption depths (i.e., erosion depth) that have been reported in
the literature (modified from Compston and Croucher [9]).
Reference Methodology Resorption
Depth (mm)
Eriksen et al. [17] Erosion depths are measured by counting the number of eroded lamellae that are a) 19.0
microscopically identified below a) osteoclasts, b) mononuclear cells, and b) 49.1
c) preosteoblast-like cells. The number of lamellae is then multiplied by the c) 62.6
lamellar thickness.
Eriksen et al. [21] Age-related differences in resorption depth of lacunae underlying preosteoblastic a) 57.2
cells measured by counting lamellae in a) females and b) males b) 55.0
Palle et al. [67] Erosion depths determined by counting lamellae but without cell characterization a) 33.0
in a) middle-aged males (33 yr), b) elderly males (67 yr), and c) elderly females b) 28.9
(64 yr). c) 27.2
Eriksen et al. [18] Maximum erosion depth was determined as in [17] and [21] for a) osteoporotics a) 55.0
and b) age-matched controls b) 49.1a
Croucher et al. [10] Eroded bone surface is reconstructed by fitting a smooth line connecting the a) 15.7b
bone surface on each side of a resorption cavity. The line is used as baseline to b) 17.0b
determine mean and maximum erosion depths. Values were determined for
a) females and b) males.
Cohen-Solal et al. [8] Erosion depth determined at sites where resorption had been completed. The 40.8
bone surface was reconstructed by drawing a line joining the adjacent quiescent
surfaces, creating a smooth contour over resorption site. Orthogonal intercept
lines were then drawn between this contour and closest cement line.

Values for resorption depth represent mean values, amedian, or bgeometric mean.

variables, erosion depth has the greatest impact resorption cavity. While a detailed explanation
on bone structure as it relates directly to the of each method and the morphometric assump-
quantum (i.e., packet) of bone that must be tions associated with their application is beyond
replaced by osteoblastic bone formation [8, 9]. the scope of this chapter, Table 4.1 summarizes
Consequently, if the bone formed in the resorp- the basic methodologies that have been used
tion cavity is insufcient to replace the amount to assess bone resorption and the results from
resorbed, due either to an increase in the several prominent investigations. Perhaps not
amount resorbed, a decrease in the amount surprisingly, the values for erosion depth vary,
formed, or some combination of the two, a neg- depending on the methodology employed.
ative balance and a potentially irreversible bone However, it is instructive to note that when
loss will occur at the remodeling site. resorption cavities are lled with new bone, the
As in cortical bone, osteoclast behaviors and resulting values for wall width are less variable.
their outcomes, including erosion depths, must Indeed, the values for wall width in normal
be inferred from what has been removed. subjects are generally on the order of 4050 mm
Several methods exist for measuring erosion [11, 16, 55, 61, 80, 89, 90], which indicates that
depth and each involves reconstruction of the erosion depth values less than 40 mm are most
62 Bone Resorption

likely underestimating the structural impact of cally, longitudinal tunneling of trabeculae that
osteoclast resorption. have been thickened by parathyroid hormone
treatment has been shown to convert them into
Structural Implications of multiple trabeculae (Figure 4.5). These trabecu-
lae have a normal thickness, but the resulting
Bone Resorption increase in trabecular number has a positive
Regardless of the methodological debate sur- effect on cancellous bone volumes [42].
rounding measurement of erosion depth, there The ability of the osteoclast to modify bone
is no argument about the importance of osteo- structure can also be substantiated by studies
clastic resorption in the maintenance of bone that have classied resorption lacunae accord-
mass and tissue architecture. As discussed ing to their microstructural proles. In that
previously, intracortical and cancellous bone regard, Gentzsch et al. [33] have used light and
resorption are clearly initiating events in the scanning electron microscopy to show that
removal of aged [5, 56, 74, 79, 84] and/or osteoclasts not only resorb cancellous bone in
damaged bone tissue [4, 7, 25, 53, 62], which is the familiar hemi-osteonal fashion (i.e., longitu-
subsequently replaced by osteoblastic bone for- dinally extended resorption lacunae), but they
mation. This activity serves continually to reju- also sculpt bone surfaces in reticulate patches.
venate the bone mineral and optimizes bone The reticulate patch morphology is particularly
architecture in response to functional loading reminiscent of the resorption pits that have
[14, 50, 64, 66, 95]. Recent studies have also pro- been observed when osteoclasts resorb miner-
vided insight into how osteoclast resorption alized matrices in vitro [45, 65].
participates in the modication of cancellous In addition to the osteoclasts role in main-
bone structures to increase bone mass. Speci- taining and/or modifying normal bone
structure, osteoclastic activities are major con-
tributors to cancellous bone pathologies. For
example, in osteoporosis, the early, rapid bone
A. loss experienced at the menopause is the result
of osteoclast hyperactivity and increased
resorption depth [2, 18, 19, 81], which leads to
focal trabecular perforations and subsequent
loss of complete structural elements [3, 9, 73,
81]. These perforations occur either via direct
tunneling through individual trabeculae [33] or
by excessive lacunar resorption on trabecular
surfaces [33, 63]. The net result of such activity
is the conversion of the plate-like cancellous

B.

Figure 4.4 Three-dimensional micro-CT images of trabeculae Figure 4.5 Photomicrograph of osteoclast cutting cone in
from the distal femur of C57/B6 mice. The normal highly con- cancellous bone of non-human primate treated with parathy-
nected trabecular structure (A) is drastically eroded by just 3 roid hormone. The cutting cone is dividing a single, thickened
weeks of limb disuse (B). trabeculae into two trabeculae of normal thickness.
Structural Aspects of Bone Resorption 63

structure to rods [81, 83], producing a dis- tion of osteoclasts from marrow precursors
proportionate loss of strength due to loss of [87]. Alternately, disuse rapidly induces osteo-
connectivity [72, 73]. This weakness in the cyte hypoxia [13], and it has been observed that
trabecular structure is only partially compen- direct oxygen deprivation of marrow culture
sated for by the increased thickness of the stimulates signicant osteoclastogenesis [1].
remaining trabeculae. In the context of structural adaptation in
The structural pathologies documented by adult mammalian cortical bone, disuse rapidly
bone histomorphometry have been substanti- precipitates substantial loss of bone mass. This
ated by Thomsen et al. [92], who have used a bone loss is the net result of rapid and profound
computer simulation model to demonstrate that osteoclastic resorption on endocortical and
the number of cancellous perforations was intracortical surfaces [52]. Periosteal resorption
correlated with the nal resorption depth. In is not evident in adult mammalian and avian
addition, analysis of digital microimages of models of disuse-induced bone loss [34, 41].
trabecular bone obtained by in vivo magnetic Endocortical resorption resulting in reduced
resonance imaging attributed the perforation cortical thickness is the primary contributor to
of trabecular plates and rods to osteoclastic disuse-induced bone loss [32], as intracortical
resorption [96]. Furthermore, microCT analyses resorption precipitated by disuse is eventually
of tibial cancellous bone have yielded three- coupled with subsequent osteoblastic recruit-
dimensional conrmation of the age-related ment that aspires to ll in each excavated cone
conversion of the cancellous bone structure of bone. In young adults, nearly all of bone
from plates to rods [12]. However, it should be resorbed intracortically is replaced by
emphasized that even though osteoclast hy- osteoblasts, while in older adults intracortical
peractivity and increased erosion depth may resorption results in a net loss of intracortical
serve as the catalyst for trabecular perforations, bone [32].
the incomplete lling of resorption cavities by Following an acute period of disuse, such as
osteoblasts still plays a major role in the struc- 4 weeks, substantial endocortical and intracor-
tural pathologies associated with osteoporotic tical erosion has ensued, while intracortical
bone loss [18, 71, 92]. remodeling is incomplete, even in the young
While there is an extensive literature detail- adult. Degradation in bone strength in response
ing how bone structure is altered by post- to acute disuse therefore arises from both endo-
menopausal estrogen depletion and aging, both cortical expansion and heightened intracortical
pathologies result from changes in osteoclast porosity. Although the loss of bone strength in
and osteoblast function that occur over a time humans due to acute disuse is obviously not
course of years. In contrast, changes in bone directly assessable, it is possible to generate a
structure induced by disuse occur much more bracketing estimate from the literature. An
rapidly, providing a means to explore the expanded endocortical surface diminishes
question of how bone resorption affects bone bones moment of inertia and thereby limits
structure without the confounding inuence resistance to bending or torsion. The endocor-
of systemic abnormalities and metabolic tical surface can be expected to expand 515%
insufciencies. within 4 weeks following the onset of disuse,
Disuse-engendered bone resorption is pre- depending on species and model [34, 46, 47].
cipitated when the bones normal loading Assuming uniform endocortical erosion, a
environment is withdrawn or substantially 110% decrease in the bones moment of inertia
diminished. Thus, disuse-induced bone loss is about the primary axis of bending would be
provoked by a variety of conditions, including anticipated, depending upon the initial propor-
extended bedrest or motor-neuron-deciency tions of the cortex (e.g., a small diameter, thick
induced paralysis [23, 47]. Distinct from bone cortex bone would be less affected than a large
loss induced by perturbations of systemic hor- diameter, thin cortex bone). As a result, ultimate
mones or cytokines, disuse-induced bone loss is bone strength (if loaded about this axis) would
conned to the bones deprived of loading. The be expected to decline proportionately. Elevated
specic stimulus for disuse induced osteoclas- intracortical porosity serves to elevate local
togenesis is relatively unexplored, but it has tissue strains for a given load and substantially
been speculated that the loss of physical stimuli degrades fracture toughness [97]. Intracortical
directly removes some factor inhibiting forma- porosity can be estimated to double from 510%
64 Bone Resorption

under normal conditions and 1020% following 7. Burr DB, Martin RB, Schafer MB, Radin EL (1985)
acute disuse [24, 34, 59]. Mechanical testing data Bone remodeling in response to in vivo fatigue micro-
damage. J Biomech 18:189200.
suggest that such an alteration would degrade 8. Cohen-Solal ME, Shih MS, Lundy MW, Partt AM (1991)
bone strength 1025% [60]. As a comparison, it A new method for measuring cancellous bone erosion
has been estimated that cortical bone strength depth: application to the cellular mechanisms of bone
degrades 1520% between the ages of 35 and 70 loss in postmenopausal osteoporosis. J Bone Miner Res
6:13318.
[57]. 9. Compston JE, Croucher PI (1991) Histomorphometric
assessment of trabecular bone remodelling in osteo-
porosis. Bone Miner 14:91102.
10. Croucher PI, Garrahan NJ, Mellish RW, Compston JE
Conclusion (1991) Age-related changes in resorption cavity char-
acteristics in human trabecular bone. Osteoporos Int
1:25761.
Osteoclastic resorption plays a central role in 11. Croucher PI, Mellish RW, Vedi S, Garrahan NJ,
the regulation of bone mass and architecture. Compston JE (1989) The relationship between resorp-
tion depth and mean interstitial bone thickness: age-
Moreover, osteoclast resorption is a prerequisite related changes in man. Calcif Tissue Int 45:1519.
to maintaining the structural health of cortical 12. Ding M, Hvid I (2000) Quantication of age-related
and cancellous bone by removing aged tissue changes in the structure model type and trabecular
and by initiating the repair of bone micro- thickness of human tibial cancellous bone. Bone 26:
2915.
damage. Conversely, hyperactive osteoclasts and 13. Dodd JS, Raleigh JA, Gross TS (1999) Osteocyte hypoxia:
excessive bone resorption can lead to irre- a novel mechanotransduction pathway. Am J Phys Cell
versible bone loss and structurally weakened Phys 277:C589602.
bone. From these observations we have arrived 14. Epker BN, Frost HM (1965) Correlation of Bone Resorp-
at an understanding of how osteoclast behaviors tion and Formation with the Physical Behavior of
Loaded Bone. J Dent Res 44:3341.
participate in both normal and pathological 15. Eriksen EF (1986) Normal and pathological remodeling
changes to bone structure but investigations of human trabecular bone: three dimensional recon-
into the regulatory cues that target osteoclasts struction of the remodeling sequence in normals and in
to bone remodeling sites are only now begin- metabolic bone disease. Endocr Rev 7:379408.
16. Eriksen EF (1993) Assessment of erosion depth by
ning. Future work aimed at understanding these lamellar counting. Bone 14:4437.
pathways should therefore provide a scientic 17. Eriksen EF, Gundersen HJ, Melsen F, Mosekilde L (1984)
basis for the design and development of Reconstruction of the formative site in iliac trabecular
therapeutic modalities that will optimize bone bone in 20 normal individuals employing a kinetic
structure. model for matrix and mineral apposition. Metab Bone
Dis Relat Res 5:24352.
18. Eriksen EF, Hodgson SF, Eastell R, Cedel SL, OFallon
WM, Riggs BL (1990) Cancellous bone remodeling in
type I (postmenopausal) osteoporosis: quantitative
References assessment of rates of formation, resorption, and bone
loss at tissue and cellular levels. J Bone Miner Res 5:
31119.
1. Arnett TR, Gibbons DC, Utting JC, Orriss IR, Hoebertz 19. Eriksen EF, Langdahl B,Vesterby A, Rungby J, Kassem M
A, Rosendaal M, et al. (2003) Hypoxia is a major stimu- (1999) Hormone replacement therapy prevents osteo-
lator of osteoclast formation and bone resorption. J Cell clastic hyperactivity: A histomorphometric study in
Physiol 196:28. early postmenopausal women. J Bone Miner Res 14:
2. Arnold JS (1970) Focal excessive endosteal resorption 121721.
in aging and senile osteoporosis, edn. In: Barzel US, 20. Eriksen EF, Melsen F, Mosekilde L (1984) Reconstruc-
editor. Osteoporosis. Grune and Stratton, Inc., New York, tion of the resorptive site in iliac trabecular bone: a
80100. kinetic model for bone resorption in 20 normal indi-
3. Arnold JS (1981) Trabecular patterns and shapes in viduals. Metab Bone Dis Relat Res 5:23542.
aging and osteoporosis, edn. In: Jee WSS, Partt AM, 21. Eriksen EF, Mosekilde L, Melsen F (1985) Trabecular
editors(eds). Bone Histomorphometry: Third Interna- bone resorption depth decreases with age: differences
tional Workshop. Armour Montagu, Paris, 297308. between normal males and females. Bone 6:1416.
4. Bentolila V, Boyce TM, Fyhrie DP, Drumb R, Skerry TM, 22. Eriksen EF, Steiniche T, Mosekilde L, Melsen F (1989)
Schafer MB (1998) Intracortical remodeling in adult Histomorphometric analysis of bone in metabolic
rat long bones after fatigue loading. Bone 23:27581. bone disease. Endocrinol Metab Clin North Am 18:
5. Boivin G, Meunier PJ (2002) Changes in bone remodel- 91954.
ing rate inuence the degree of mineralization of bone. 23. Eser P, Frotzler A, Zehnder Y, Wick L, Knecht H, Denoth
Connect Tissue Res 43:5357. J, et al. (2004) Relationship between the duration of
6. Burr DB (2002) Targeted and nontargeted remodeling. paralysis and bone structure: a pQCT study of spinal
Bone 30:24. cord injured individuals. Bone 34:86980.
Structural Aspects of Bone Resorption 65

24. Feik SA, Thomas CD, Clement JG (1997) Age-related 45. Jones SJ, Boyde A, Ali NN, Maconnachie E (1986) Varia-
changes in cortical porosity of the midshaft of the tion in the sizes of resorption lacunae made in vitro.
human femur. J Anat 191(Pt 3):40716. Scan Electron Microsc 15:7180.
25. Frost HM (1960) Presence of microcracks in vivo in 46. Judex S, Garman R, Squire M, Busa B, Donahue LR,
bone. Henry Ford Hosp Med J 8:2535. Rubin C (2004) Genetically linked site-specicity of
26. Frost HM (1964) Dynamics of bone remodeling, edn. In: disuse osteoporosis. J Bone Miner Res 19:60713.
Frost HM, editor. Bone Biodynamics. Little, Brown and 47. Kiratli BJ, Smith AE, Nauenberg T, Kallfelz CF, Perkash I
Co, Boston, 31533. (2000) Bone mineral and geometric changes through
27. Frost HM (1966) Bone dynamics in osteoporosis and the femur with immobilization due to spinal cord
osteomalacia, edn. In: (ed) Series Titl. Charles C. injury. J Rehabil Res Dev 37:22533.
Thomas, Springeld, IL, pp . 48. Landeros O, Frost HM (1964) A Cell System in Which
28. Frost HM (1969) Tetracycline-based histological analy- Rate and Amount of Protein Synthesis Are Separately
sis of bone remodeling. Calcif Tissue Int 3:21137. Controlled. Science 145:13234.
29. Frost HM (1973) Bone Remodeling and its Relationship 49. Langdahl BL, Mortensen L, Vesterby A, Eriksen EF,
to Metabolic Bone Disease, edn. In: (ed) Series Titl. Charles P (1996) Bone histomorphometry in hypo-
Charles C. Thomas, Springeld, IL, pp . parathyroid patients treated with vitamin D. Bone 18:
30. Frost HM (1983) The skeletal intermediary organiza- 1038.
tion. Metab Bone Dis Relat Res 4:28190. 50. Lanyon LE (1984) Functional strain as a determinant for
31. Frost HM (1986) Intermediary Organization of the bone remodeling. Calcif Tissue Int 36:S5661.
Skeleton, edn. In: (ed) Series Title. CRC Press, Inc., Boca 51. Lanyon LE (1992) The success and failure of the adap-
Raton, FL, pp . tive response to functional load- bearing in averting
32. Frost HM (1992) Perspectives: bones mechanical usage bone fracture. Bone 13:S1721.
windows. Bone Miner 19:25771. 52. Leblanc AD, Schneider VS, Evans HJ, Engelbretson DA,
33. Gentzsch C, Delling G, Kaiser E (2003) Microstructural Krebs JM (1990) Bone mineral loss and recovery after
classication of resorption lacunae and perforations 17 weeks of bed rest. J Bone Miner Res 5:84350.
in human proximal femora. Calcif Tissue Int 72:698 53. Lee TC, Staines A, Taylor D (2002) Bone adaptation to
709. load: microdamage as a stimulus for bone remodelling.
34. Gross TS, Rubin CT (1995) Uniformity of resorptive J Anat 201:43746.
bone loss induced by disuse. J Orthop Res 13:70814. 54. Li J, Mashiba T, Burr DB (2001) Bisphosphonate treat-
35. Howship J (1817) Experiments and observations in ment suppresses not only stochastic remodeling but
order to determine the means employed by the animal also the targeted repair of microdamage. Calcif Tissue
economy in the formation of bone. Transactions of the Int 69:2816.
Medico-Chirurgical Society (London). 6:263301. 55. Lips P, Courpron P, Meunier PJ (1978) Mean wall thick-
36. Jaworski ZF (1984) Lamellar bone turnover system and ness of trabecular bone packets in the human iliac crest:
its effector organ. Calcif Tissue Int 36(Suppl 1):S4655. changes with age. Calcif Tissue Res 26:1317.
37. Jaworski ZF, Duck B, Sekaly G (1981) Kinetics of osteo- 56. Martin B (1993) Aging and strength of bone as a struc-
clasts and their nuclei in evolving secondary Haversian tural material. Calcif Tissue Int 53:S349; discussion
systems. J Anat 133:397405. S3940.
38. Jaworski ZF, Hooper C (1980) Study of cell kinetics 57. Martin B (1993) Aging and strength of bone as a struc-
within evolving secondary Haversian systems. J Anat tural material. Calcif Tissue Int 53(Suppl 1):S349; dis-
131:91102. cussion S3940.
39. Jaworski ZF, Lok E (1972) The rate of osteoclastic bone 58. Martin RB (2002) Is all cortical bone remodeling initi-
erosion in Haversian remodeling sites of adult dogs rib. ated by microdamage? Bone 30:813.
Calcif Tissue Res 10:10312. 59. Martin RB, Pickett JC, Zinaich S (1980) Studies of
40. Jaworski ZFG (1981) The quantum concept of bone skeletal remodeling in aging men. Clin Orthop 268
remodeling in adults, edn. In: Norman AM, Schaefer K, 82.
Coburn JW, DeLuca HF, Fraser D, Grigoleit HG, et al., 60. McCalden RW, McGeough JA, Barker MB, Court-Brown
editors(eds). Vitamin D: Biochemical, Chemical, and CM (1993) Age-related changes in the tensile properties
Clinical Aspects Related to Calcium Metabolism. of cortical bone. The relative importance of changes in
Degruyter, Berlin. porosity, mineralization, and microstructure. J Bone
41. Jaworski ZFG, Uthoff HK (1986) Reversibility of non- Joint Surg Am 75:1193205.
traumatic disuse osteoporosis during its active phase. 61. Meunier PJ (1983) Histomorphometry of the skeleton,
Bone 7:4319. edn. In: Peck WA, editor. Bone and Mineral Research.
42. Jerome CP, Burr DB, Van Bibber T, Hock JM, Brommage Annual 1. Excerpta Medica, Amsterdam, 191222.
R (2001) Treatment with human parathyroid hormone 62. Mori S, Burr DB (1993) Increased intracortical remod-
(134) for 18 months increases cancellous bone volume eling following fatigue damage. Bone 14:1039.
and improves trabecular architecture in ovariectomized 63. Mosekilde L (1990) Consequences of the remodelling
cynomolgus monkeys (Macaca fascicularis). Bone 28: process for vertebral trabecular bone structure: a
1509. scanning electron microscopy study (uncoupling of
43. Jilka RL (2003) Biology of the basic multicellular unit unloaded structures). Bone Miner 10:1335.
and the pathophysiology of osteoporosis. Med Pediatr 64. Mosekilde L (2000) Age-related changes in bone mass,
Oncol 41:1825. structure, and strength effects of loading. Z Rheuma-
44. Johnson LC (1964) Morphological analysis in pathol- tol 59(Suppl 1):19.
ogy: the kinetics of disease and general biology of bone, 65. Murrills RJ, Shane E, Lindsay R, Dempster DW (1989)
edn. In: Frost HM, editor. Bone Biodynamics. Little Bone resorption by isolated human osteoclasts in vitro:
Brown, Boston, 543654. effects of calcitonin. J Bone Miner Res. 4:25968.
66 Bone Resorption

66. Noble BS, Peet N, Stevens HY, Brabbs A, Mosley JR, Reilly and cellular mechanisms of bone loss. J Clin Invest
GC, et al. (2003) Mechanical loading: biphasic osteocyte 72:1396409.
survival and targeting of osteoclasts for bone destruc- 82. Partt AM, Mundy GR, Roodman GD, Hughes DE, Boyce
tion in rat cortical bone. Am J Physiol Cell Physiol BF (1996) A new model for the regulation of bone
284:C93443. resorption, with particular reference to the effects of
67. Palle S, Chappard D, Vico L, Riffat G, Alexandre C (1989) bisphosphonates. J Bone Miner Res 11:1509.
Evaluation of the osteoclastic population in iliac crest 83. Parisien MV, McMahon D, Pushparaj N, Dempster DW
biopsies from 36 normal subjects: a histoenzymologic (1988) Trabecular architecture in iliac crest bone biop-
and histomorphometric study. J Bone Miner Res 4: sies: infra-individual variability in structural parame-
5016. ters and changes with age. Bone 9:28995.
68. Partt AM (1979) Quantum concept of bone remodel- 84. Polig E, Jee WS (1987) Bone age and remodeling: a math-
ing and turnover: implications for the pathogenesis of ematical treatise. Calcif Tissue Int 41:1306.
osteoporosis. Calcif Tissue Int 28:15. 85. Raisz LG (1999) Physiology and pathophysiology of
69. Partt AM (1982) The coupling of bone formation to bone remodeling. Clin Chem 45:13538.
bone resorption: a critical analysis of the concept and 86. Rubin CT (1984) Skeletal strain and the functional
of its relevance to the pathogenesis of osteoporosis. signicance of bone architecture. Calcif Tissue Int
Metab Bone Dis Relat Res 4:16. 36:S1118.
70. Partt AM (1983) The physiologic and clinical signi- 87. Rubin J, Fan X, Biskobing DM, Taylor WR, Rubin CT
cance of bone histomorphometric data, edn. In: Recker (1999) Osteoclastogenesis is repressed by mechanical
RR, editor. Bone Histomorphometry: Techniques and strain in an in vitro model. J Orthop Res 17:63945.
Interpretation. CRC Press, Boca Raton, FL, 143224. 88. Schock CC, Noyes FR, Villanueva AR (1972) Measure-
71. Partt AM (1984) The cellular basis of bone remodel- ment of haversian bone remodeling by means of tetra-
ing: the quantum concept reexamined in light of recent cycline labeling in rib of Rhesus monkeys. Henry Ford
advances in the cell biology of bone. Calcif Tissue Int Hosp Med J 20:13144.
36(Suppl 1):S3745. 89. Steiniche T, Christiansen P, Vesterby A, Hasling C,
72. Partt AM (1987) Trabecular bone architecture in the Ullerup R, Mosekilde L, et al. (1994) Marked changes in
pathogenesis and prevention of fracture. Am J Med iliac crest bone structure in postmenopausal osteo-
82:6872. porotic patients without any signs of disturbed bone
73. Partt AM (1992) Implications of architecture for the remodeling or balance. Bone 15:739.
pathogenesis and prevention of vertebral fracture. Bone 90. Steiniche T, Mosekilde L, Christensen MS, Melsen F
13(Suppl 2):S417. (1989) Histomorphometric analysis of bone in idio-
74. Partt AM (1993) Bone age, mineral density, and fatigue pathic hypercalciuria before and after treatment with
damage. Calcif Tissue Int 53(Suppl 1):S825; discussion thiazide. Apmis 97:3028.
S856. 91. Teitelbaum SL (2000) Bone resorption by osteoclasts.
75. Partt AM (1993) Morphometry of bone resorption: Science 289:15048.
introduction and overview. Bone 14:43541. 92. Thomsen JS, Mosekilde L, Mosekilde E (1996) Quanti-
76. Partt AM (1994) Osteonal and hemi-osteonal remod- cation of remodeling parameter sensitivity assessed
eling: the spatial and temporal framework for signal by a computer simulation model. Bone 19:50511.
trafc in adult human bone. J Cell Biochem 55:27386. 93. Tran Van P, Vignery A, Baron R (1982) An electron-
77. Partt AM (1996) Skeletal heterogeneity and the pur- microscopic study of the bone-remodeling sequence in
poses of bone remodeling, edn. In: Marcus R, Feldman the rat. Cell Tissue Res 225:28392.
B, Kelsey J, editors(eds). Osteoporosis. Academic Press, 94. Tran Van PT, Vignery A, Baron R (1982) Cellular
San Diego. kinetics of the bone remodeling sequence in the rat.
78. Partt AM (2000) BMU origination and progression: Anat Rec 202:44551.
relationship to targeted and nontargeted remodeling. 95. Turner CH, Pavalko FM (1998) Mechanotransduction
J Bone Mine Res 15:823. and functional response of the skeleton to physical
79. Partt AM (2002) Targeted and nontargeted bone stress: the mechanisms and mechanics of bone adapta-
remodeling: relationship to basic multicellular unit tion. J Orthop Sci 3:34655.
origination and progression. Bone 30:57. 96. Wehrli FW, Gomberg BR, Saha PK, Song HK, Hwang SN,
80. Partt AM, Foldes J (1991) The ambiguity of interstitial Snyder PJ (2001) Digital topological analysis of in vivo
bone thickness: a new approach to the mechanism of magnetic resonance microimages of trabecular bone
trabecular thinning. Bone 12:11922. reveals structural implications of osteoporosis. J Bone
81. Partt AM, Mathews CH,Villanueva AR, Kleerekoper M, Miner Res 16:152031.
Frame B, Rao DS (1983) Relationships between surface, 97. Yeni YN, Brown CU, Wang Z, Norman TL (1997) The
volume, and thickness of iliac trabecular bone in aging inuence of bone morphology on fracture toughness of
and in osteoporosis. Implications for the microanatomic the human femur and tibia. Bone 21:4539.
5.
Inflammatory Cytokines
Mark S. Nanes and Roberto Pacici

clastogenesis. NfkB suppresses the expression of


Introduction osteoblast matrix protein genes, stimulates the
production of matrix-degrading metallopro-
Inammatory cytokines have a major role in teinases, causes resistance to 1,25-dihydroxyvit-
bone disease. The estrogen deciency of amin D3 (1,25-(OH)2D3) and inhibits the
menopause results in an indolent inammatory differentiation of osteoblast progenitors. Simi-
state with elevated production of cytokines, larly, activation of MAPK by TNF and IL-1 leads
most notably tumor necrosis factor-a (TNF) to the phosphorylation of other transcription
and interleukin-1 (IL-1). A local effect of factors of the activator protein (AP-1) class that
cytokines in inammatory arthritis also con- contribute to the catabolic direction of gene
tributes to periarticular bone destruction. expression. This chapter will focus on the source
Cytokines act as skeletal catabolic agents that of cytokines in bone during estrogen deciency,
stimulate osteoclastogenesis while simultane- cytokine intracellular signal pathways, and
ously inhibiting osteoblast differentiation and the molecular mechanism through which
function. The postmenopausal state is associ- cytokines regulate the genes that result in bone
ated with inltration of marrow by TNF- loss. A large number of inammatory cytokines
producing T cells, which percolate through bone contribute to skeletal pathology, but attention
and stimulate the production of an expanded will be focused primarily on TNF and IL-1 as
local cytokine cascade. The skeletal catabolic central players. Inammatory cytokines and
state is achieved by cytokine regulation of genes their signal pathways are important targets
in hematopoietic cells, marrow stromal cells, in the development of new therapies for
osteoblasts, and osteoclasts that tilt the balance osteoporosis.
of formation and resorption toward net bone
loss. TNF and IL-1 engage initially distinct sig-
naling pathways that converge with activation
of the transcription factor, nuclear factor kappa
Sources of Cytokines in Bone
B (NfkB), and stimulation of the mitogen-
activated protein kinase (MAPK) system. The Early Data that Estrogen Deficiency
combined effect of these two cytokines provides Stimulates Cytokine Production
a potent signal that stimulates differentiation
and prolongs lifespan of osteoclasts, while A major focus of osteoporosis research has
inhibiting differentiation of osteoblasts and been to understand the reasons for accelerated
promoting their apoptosis. NfkB regulates an bone resorption following menopause; thus, an
array of genes that include the receptor activa- obvious question has been how estrogen de-
tor of NfkB ligand (RANKL) and its receptor, ciency leads to increased osteoclast activity
RANK, both necessary and sufcient for osteo- and ultimately osteoporosis. Early work by
67
68 Bone Resorption

McSheehy et al. and Thompson et al. showed TNF and RANKL. T-cell-decient nude mice do
that conditioned media derived from osteo- not respond to IL-7 with bone loss, though they
blasts that were stimulated by a variety of do so after adoptive T-cell transfer [211].
factors increased osteoclastogenesis [142144, Whether the in vivo action of IL-7 requires pro-
207, 208]. These results suggested that sub- duction of TNF is, as yet, unknown. Surpris-
stances were produced by osteoblasts that ingly, IL-7 inhibits osteoclastogenesis in an ex
mediated differentiation of osteoclast precur- vivo system of cultured marrow cells [125].
sors. After several inammatory cytokines were Marrow cells from IL-7-decient mice exhibit
found capable of producing this response, an increased osteoclastogenic response to a
research focused on discerning their relative variety of stimuli, including 1,25-dihydroxyvit-
contributions in estrogen deciency bone loss. amin D, PTH, and mCSF/RANKL. To make
A lively debate ensued with proponents for IL- matters more confusing, marrow cells derived
1, TNF, or IL-6 as the major players. The osteo- from IL-7-receptor-decient mice had the oppo-
clastogenic response to all three cytokines in site outcome [125]. Thus, the exact role of IL-7
vitro was based on sound data [138, 166]. in bone is yet to be established. Understanding
However, the use of different experimental IL-7 may require knowing both its systemic
models may have yielded different results. action on T-cell expansion and direct local
Several reports suggested increased produc- actions on skeletal cells.
tion of TNF by cultures of mononuclear cells In premenopausal women the production of
derived from postmenopausal women, an effect TNF, interleukin-1, -4, -6, and IFNg by blood
reversed by estrogen replacement [167, 177, cells varies inversely with the estrogen level.
180]. In this model, secretion of IL-1, but not Interestingly, the plasma levels of these
always of IL-6, mirrored that of TNF [67, 180]. cytokines also correlate inversely with bone
Experimental models in which estrogen did not density after menopause; this strengthens the
seem to regulate TNF expression included cul- hypothesis that cytokines play an important
tured stromal cells, some osteoblastic cell lines, role in menopausal bone loss [186, 220, 242].
and bone biopsies [1, 37, 176, 178]. Although
small amounts of TNF are produced by many of
these cell types, the lack of regulation by estro- Central Role for TNF in Estrogen
gen in all but monocytes hinted at an uncer- Deficiency Bone Loss
tainty as to the genuine source. To add to the
confusion, phytohemaglutinin stimulation was Substantial in vitro and in vivo evidence sug-
required to demonstrate increased TNF pro- gests that TNF is a central player in the patho-
duction from monocytes. Another level of com- physiology of skeletal loss following menopause
plexity was evident in the study by Atkins et al., [82, 166]. As noted above, TNF and IL-1 are
in which peripheral blood mononuclear cells potent stimuli for the formation of osteoclasts
were shown to secrete TNF, IL-1, and IL-6 after in vitro. This is also true in animal models with
co-culture with the stromal-like cell line ST-2 reference to ectopic bone formation, estrogen-
[6]. Thus, cytokine production is also modu- deciency-induced bone loss, and to periarticu-
lated by cell-cell contact and/or paracrine events lar bone destruction in the inamed joint. In
in the bone microenvironment. More recent these animal models, blockade of TNF or IL-1
data revealed the presence of membrane action alleviates bone loss, thus conrming the
anchored RANKL, part of a potent osteoclasto- role played by TNF and IL-1 in stimulating
genic system that is, itself, dependent on TNF osteoclastogenesis [33, 35, 106108, 110, 167].
(discussed in detail below). The most denitive ndings in support of the
Weitzman et al. have suggested that IL-7 also central role of TNF in mediating osteoporosis
contributes to bone loss following estrogen de- came from experiments that showed that TNF-
ciency [226, 227]. Ovariectomy increases the decient and TNF receptor knockout mice are
level of IL-7 in mice and neutralization of IL-7 resistant to ovariectomy-induced bone loss
in vivo blocks ovariectomy-induced bone loss. [106, 107, 183]. Comparable ndings from IL-6
Exactly where IL-7 is synthesized and where it or IL-1 knockout mice were less denitive. In
ts into the scheme of skeletal regulation is the wild-type mouse, the rise in TNF follow-
unclear. At the physiologic level, IL-7 stimulates ing ovariectomy is immediately followed by
T-cell production of other cytokines, including increases in IL-6 and IL-1, which, in turn,
Inflammatory Cytokines 69

magnify the stimulus to osteoclastogenesis by repressing the compensatory increase in


[17, 37, 77, 178, 182]. bone formation.
Evidence is accumulating to suggest that T
T-Cells as a TNF Source in Estrogen cells also regulate bone resorption in humans.
For example, RANKL-expression by T cells is
Deficiency Bone Loss up-regulated by estrogen deciency and corre-
Substantial evidence is accumulating to suggest lates with increases in bone resorption markers
that T cells play a pivotal role in bone loss and inversely with serum 17b-estradiol [47].
caused by estrogen deciency [33]. T cells are a
major source of TNF, and ovariectomy enhances
the production of T-cell-derived TNF. The Estrogen Effects on TNF
concept that the T cell is the source of TNF is
consistent with other models of T-cell- Production and Action
dependent immunity [187]. TNF acts through
the TNF receptor, TNFR1 (p55), to augment Estrogen Regulation of T-Cell
RANKL-induced osteoclastogenesis and osteo-
clast activity [58]. Athymic, T-cell-decient Clonal Expansion
nude mice are completely protected against the
As discussed above, estrogen regulates the
bone loss and the increase in bone turnover
increase in the mass of TNF-producing cells [33,
induced by ovariectomy [33, 183] (Figure 5.1).
183]. Estrogen also causes the lymphocyte
Reconstitution studies in nude mice have shown
populations to shift toward clones that produce
that the relevant T-cell subpopulation is made
anti-inammatory TH2 cytokines (IL-4, IL-10),
up of CD4+ cells, but not of CD+8 cells, because
thereby causing fewer clones that produce
only CD4+ cells, when implanted in nude mice,
inammatory TH1 cytokines (IL-2, IFNg)). In
restored ovariectomy-induced bone loss trans-
postmenopausal women, production of
fer of CD4+. It is thus apparent that CD4+ T cells
cytokines representative of TH1 lymphocytes is
are an essential mediator of the bone-wasting
increased, an effect reversed by supplemental
effect that is the result of estrogen deciency in
estrogen [97].
vivo. The ovariectomy induced increase in T-cell
The mechanism by which estrogen regulates
production raises the TNF level to a point where
T-cell clonal expansion is shown in Figure 5.2.
it can augment RANKL-induced osteoclastoge-
nesis [33]. TNF may also be pivotal in disrupt-
ing the coupling between bone resorption and Ovx
bone formation, not only by synergizing with
RANKL to stimulate bone resorption, but also IFNg

CIIT A
Sham Ovx Ovx E2
250
MHCII
200
MF Ag Presentation
BMD 150 *
mg/cm2 100 T Cell
Activation
T Cell T Cell
50
Proliferation Lifespan
0 T Cell Number
WT Mouse Nude Mouse
(No T cells) T Cell TNF Production

Figure 5.1 T-cell-deficient mice are protected against RANKL-induced osteoclast ()


ovariectomy-induced bone loss. Reproduced with permission formation and bone loss
from Cenci S, Weitzmann MN, Roggia O, et al. Estrogen defi-
ciency induces bone loss by enhancing T-cell production of TNF- Figure 5.2 Schematic representation of the mechanism by
alpha. J Clin Invest 2000 Nov;106(10):12034. which ovariectomy causes bone loss.
70 Bone Resorption

Ovariectomy increases T-cell activation. This in tion of antigen presentation in macrophages.


turn causes an expansion of the T-cell pool in CIITA expression is regulated by four distinct
bone marrow, spleen and in lymph nodes. T-cell promoters that direct the transcription of four
activation occurs by enhancing antigen presen- separate rst exons spliced to a common second
tation by bone marrow macrophages. This in exon [149, 150]. Initially it was thought that IFNg
turn is due to the up-regulation of the expres- expression in murine macrophages is regulated
sion of MHCII that occurs in estrogen de- exclusively by promoter IV [149, 150, 165].
ciency. The mechanism of T-cell activation However, it is now recognized that promoters I
elicited by estrogen deciency is similar to that and IV together account for IFNg induced CIITA
triggered by infections, but the intensity of in vitro and in vivo [168, 225].
events that follow estrogen withdrawal is signi- CIITA is constitutively expressed in B and
cantly less. With estrogen withdrawal, CD4+ dendritic cells, but not in bone marrow macro-
clones of T cells double. Modulation of anti- phages. IFNg induces CIITA in macrophages.
gen presentation by estrogen is specic for Increased CIITA expression in bone marrow
macrophages, since no change occurs in B cells macrophages from ovariectomized mice is
and dendritic cells, the other two antigen pre- the result of ovariectomy leading to an increase
senting-cell populations (APC) [32]. in both T-cell production of IFNg and the
The relevance of this mechanism in vivo has responsiveness of the CIITA gene to IFNg
been established by utilizing DO11.10 mice, a [32].
strain in which all T cells recognize a single That ovariectomy increases T-cell production
peptide epitope of chicken albumin (ovalbu- of IFNg was demonstrated by measuring the
min), which is not expressed in mice. In the cytokine concentration in the media of puried
absence of ovalbumin, the APC of DO11.10 mice T cells and by uorescent-activated cell sorting
cannot induce T-cell activation. Therefore, if (FACS) analysis of unfractionated bone marrow.
APC are targeted by estrogen, the DOLL.10 mice IFNg production by T cells is induced either by
should be protected from increased T-cell pro- a cyclosporin-A sensitive T-cell receptor (TCR)-
liferation and the bone loss that follows ovariec- dependent mechanism, mediated by T-cell acti-
tomy. Injection of ovalbumin leads to the vation, or by the cytokines IL-12 and IL-18.
generation of the appropriate MHC-peptide The expression of the IL-12 and IL-18 genes
antigen for the DOLL.10 mouse T cells. This in BMM is induced by Nf-kB and AP-1,
restores the series of events in which ovariec- nuclear proteins whose transcriptional activity
tomy causes the T-cell pool to expand and to is directly repressed by estrogen [4, 60, 194].
induce bone loss. It is therefore apparent that Unstimulated bone marrow macrophages, such
the generation of appropriate peptide-MHC as those from estrogen-replete mice, express low
complexes is critical to the process by which or undetectable levels of NFkB and AP-1 [148].
ovariectomy increases T-cell proliferation and Ovariectomy potently increases secretion of IL-
lifespan and leads to bone loss. Furthermore, the 12 and IL-18, whereas treatment with 17b estra-
nding that T cells from ovariectomized mice diol represses it. Thus, estrogen represses CIITA
exhibit an increased response to ovalbumin by decreasing IFNg production via an inhibitory
demonstrates that ovariectomy increases the effect on the production of IL-12 and IL-18.
reactivity of APC to endogenous antigens rather Also, because CIITA expression in T cells
than stimulating the production of a new increases IFNg production, and because IFNg
antigen or modulating antigen levels. stimulates its own inducers, IL-18 and IL-12 and
The effects of ovariectomy on antigen pre- IL-12 receptor expression, ovariectomy triggers
sentation and the resulting changes in T-cell an amplication loop that leads to a further
activation, proliferation, and lifespan come increase in the level of IFNg and to the induc-
about because, as a result of ovariectomy, there tion of CIITA [68, 105, 151].
is an increased expression of the gene that
encodes Class II Transactivator (CIITA). The
product of the CIITA gene is a non-DNA Regulation of IFNg By Estrogen and
binding factor that functions as a transcrip- Skeletal Effects of IFNg
tional coactivator when recruited to the MHC II
promoter by interaction with promoter-bound IFNg is a key factor in a complex pathway by
factors [18, 152, 153]. CIITA expression is, which ovariectomy leads to increased T-cell
indeed, required and sufcient for the stimula- proliferation and T-cell TNF production.
Inflammatory Cytokines 71

Ovariectomy up-regulates IFNg production in inhibits TNF-a-stimulated JNK activity in


vivo, even though estrogen stimulates IFNg gene nonskeletal cells [26]. Thus, estrogen decreases
expression in vitro [55]. The discordance the pool of TNF-producing T-cells, suppresses
between in vitro and in vivo data is further TNF transcription in monocytes, and decreases
exemplied by the nding that IFNg represses sensitivity to the TNF /RANKL stimulus.
osteoclast formation in vitro [56]. In vivo, IFNg
stimulates osteoclast formation by increasing
the T-cell production of TNF. IFNg receptor Cytokine Signal Pathways in
knock-out mice are entirely protected against
ovariectomy-induced bone loss. Furthermore,
Skeletal Cells
ovariectomy fails to increase CIITA expression
in IFNg receptor knock out mice [32]. Thus, the Complex intracellular pathways transmit
data strongly suggest that in estrogen deciency cytokine signals from cell surface receptors to
the indirect pro-resorptive effects of IFNg gene activation. Here we will restrict the discus-
prevail over the direct repression of osteoclas- sion to the signal pathways for TNF and IL-1 as
togenesis. This is consistent with earlier reports they directly affect osteoblast and osteoclast
that indicated bone resorption and/or bone loss function. Although these cytokines initiate their
increased in models of IFNg overexpression [9, signals via unique membrane receptor families,
136]. Other studies that have shown IFNg the different signals converge in similar intra-
decreases bone resorption in vivo have been cellular pathways. These include the activation
carried out in T-cell decient mice [188, 209]. of the transcription factor Nf-kB, activation of
This again demonstrates that the T-cell- mitogen-activated protein kinase (MAPK), and
mediated indirect effects are more potent than the regulation of the apoptotic caspase cascade.
direct repression by IFNg. IFNg has been shown Once activated, NFkB and MAPK elicit cell-
in one study to repress bone resorption in a specic responses in osteoblasts and osteoclasts
T-cell replete model, but the only in vivo data that include a divergent response with regard to
presented were obtained on the calvaria of cell survival.
newborn mice [203]. Further work will be
needed to sort out possible maturation stage TNF Signaling
dependent effects of IFNg.
A trimeric form of TNF binds one of two cell
surface receptors, TNFR1 (TNFRSF1A, p55)
Estrogen Repression Of TNF Gene and TNFR2 (TNFRSF1B, p75), to initiate a
Expression and Action signal cascade that causes inammatory gene
activation, selective gene repression, and for the
Estrogen not only suppresses T-cell prolifera- TNFR1, an apoptotic response (Figure 5.3). Both
tion, but also inhibits monocyte TNF produc- of these receptors are expressed on osteoblasts
tion and TNF action [197]. In a study using and osteoclasts. From receptor to gene, numer-
RAW 264.7 cells (a monocytic precursor that ous signal pathways provide an opportunity
can be stimulated to acquire the osteoclast phe- for divergence of the skeletal response. As will
notype), estrogen decreased transcriptional be discussed below, the TNF signal pathways
activity of the TNF promoter. Mutation of an include features linked specically to stimula-
AP-1 binding site in the promoter abolished the tion of bone resorption, as well as inhibition of
inhibitory effect of estrogen. Furthermore, the bone formation. Stimulation of bone resportion
expression of the AP-1 family proteins cJUN, is due to protease production by osteoblasts
JUND, and also the binding of AP-1 proteins to and the escalation of osteoclastogenesis. Inhi-
the promoter were selectively decreased after bition of bone formation is due to the inhibition
estrogen stimulation, as was the activity of JUN of osteoblast differentiation, suppression of
kinase (JNK), a kinase that would normally mature osteoblast function, and the induction
enhance the AP-1 transcriptional signal. Shevde of osteoblast resistance to 1,25-(OH)2D3. TNFR1
et al. conrmed this in cultured monocytes is the functional form of the receptor in both
and RAW 264.7 pre-osteoclastic cells, where osteoblasts and osteoclasts. Both Abu-Amer
the osteoclastogenic stimulus was RANKL, the et al. [2] and Roggia et al. [183] have reported
membrane anchored protein that binds the that TNF fails to stimulate osteoclastogenesis
TNF-family receptor RANK [194]. Estrogen also in TNFR1 receptor knockout mice (R1-/-).
72 Bone Resorption

TNF-a

TNFR1

cytoplasm SODD

FADD Caspases
TRAD
RIP D
clAP
TRAF2 Apoptosis

IKKb IKKa
NEMO + Akt
JUNK, p38
nucleus NIK+ MAPK
A20 P
P P
lkB
p65
p50 p50 p65 P

Figure 5.3 TNF signaling pathways. TNF binds as a trimer to its receptor to activate several intracellular pathways. Aggregation of
a protein complex, including TRAF2, transduces the signal along the IkB pathway leading to phosphorylation of IkB with liberation
of the transcription factor Nf-kB for nuclear entry and regulation of gene transcription. Additional pathways are activated via TRAF,
including JNK and p38 MAPK. The apoptotic pathway is simultaneously activated by TNF through FADD and the caspase cascade.
Modulator proteins include inhibitory SODD at the TNF receptor, cIAP, which attenuates the apoptotic response, RIP, and NEMO.
Reprinted from Gene, vol. 321, Nanes MS. Tumor necrosis factor-alpha: molecular and cellular mechanisms in skeletal pathology,
115 (2003), with permission from Elsevier.

R1+/+R2-/- knockout cells respond like the wild- Once the TNF trimer is bound, the receptor
type cells in vitro, whereas R1-/-/R2-/- cells are associates with cytosolic proteins that couple
resistant to TNF action, as expected. Similarly, to downstream signals. One of the rst asso-
TNF inhibition of osteoblastogenesis in bone ciations is with the mediator TNF receptor-
marrow stromal cells requires the TNFR1. associated death domain (TRADD), a cytosolic
Although not required for action, TNFR2 may protein that directs the ow of information
still contribute to the sensitivity toward TNF along two well-known pathways: The rst of
[62]. In addition, stimulation of TNFR2 could these activates TRAFs 1 and 2. These regulate a
activate additional paracrine factors that affect large cytosolic complex that contains the I
the osteoblast; thus, a role for TNFR2 in vivo kappa B kinases (a and b), IkB (a, b, and e), and
cannot be excluded. The skeletal phenotype of the transcription factor NfkB. The specic
R1-/- mice is normal [219]. R2-/- mice are also TRAF family members activated by TNF stimu-
grossly normal, although they have not been lation provide selectivity through preferential
evaluated by histomorphometry. TNF therefore activation by TNF family members. TNF-a sig-
does not seems to be involved in the regulation naling is mediated by TRAF 2 in many nonskele-
of early skeletal differentiation, notwithstanding tal cells and possibly in osteoblasts [30]. TRAF
its role in the pathophysiology of osteoporosis 6, which is activated by the RANK and CD40
and inammatory joint disease. receptors, stimulates osteoclastogenesis [41, 59,
Inflammatory Cytokines 73

96]. The association of TRAF6 with the RANK both osteoblasts and osteoclasts, IL-1-stimu-
receptor, rather than the TNFR1, is conferred by lated Nf-kB is translocated to the nucleus as
a unique TRAF6 sequence [41]. Interestingly, IL- described for TNF stimulation [93, 94]. Jeffries
1 stimulation of osteoclastogenesis also requires et al. investigated the interactions of intracellu-
TRAF6 association with the IL1R-stimulated lar proteins in the IL-1 signal cascade and
mediator IRAK [29]. More work will be needed showed that transfection of IRAK-1, TRAF-6, or
to determine if the dissimilar effects of TNF as MyD88 all caused Nf-kB translocation and
compared with RANKL are the result of selec- transactivation of a p65-dependent transcrip-
tive selection of TRAFs. In addition to induc- tional response [90]. Transfection of dominant
ing NfkB activation, TRAFs signal via JNK, negatives of any one of these proteins blocks IL-
p38 kinase, and protein kinase C (PKC). p38 1 action, suggesting that all proteins may be
enhances the transcriptional potency of Nf-kB needed for activation. This conclusion is also
[16, 214]. Indeed, inhibitors of p38 MAPK supported by the lack of an IL-1 response in
reduce TNF stimulation of osteoclastogenesis MyD88- or IRAK-1-decient cells. The small G
[117]. protein Rac-1 may also function downstream of
On the basis of electrophoretic mobility shift the IRAK-1/TRAF-6/MyD88 signal, as shown in
assays (EMSA), TNF robustly and rapidly acti- experiments that utilize dominant negative or
vates Nf-kB and AP-1 proteins in osteoblasts. constitutively active mutants of Rac-1 in the
The proteins that compose the Nf-kB and AP-1 presence of the three upstream proteins. Rac1
complexes in osteoblasts can be shown by anti- interacts directly with MyD88 and the IL-1
body supershift on EMSA to be predominantly receptor accessory protein. MyD88 acts to facil-
p65/p50 (not c-Rel or Rel-B) and predominantly itate inclusion of IRAK-4 in the complex. Unlike
JunD/Fra-2, respectively (Nanes, unpublished). the other IRAKS, which have dispensable kinase
The second pathway that spirals off from activity, IRAK-4 has an essential kinase that
TRADD is induced when the fas-activated death phosphorylates IRAK-1. Exactly which IRAK
domain (FADD) is activated. FADD is a protein functions in bone cells is unknown. Further
that triggers the pro-apoptotic caspases, leading investigation reveals that the cytosolic protein
to death in most cells, including osteoblasts. Tollip, which anchors the IL-1R to MyD88 and
These pathways are not exclusive, inasmuch as other adapters, is an active component of the
TRAF-mediated activation of the anti-apoptotic complex [25]. Once IL-1 stimulates an active
protein, c-IAP, tempers the TNF apoptotic cytosolic complex, the signal is transduced
signal. Thus, gene regulation in osteoblasts by inward. An IRAK-1-activated and mobile
TNF via Nf-kB may in one view, be limited to complex includes Tab1, Tab2, and Tak1, which
apoptotic suicide, or in another, the apoptotic associate with TRAF6. This is followed by the
response may be tempered by Nf-kB activation. expected phosphorylation of the IkB kinases,
Activation of these two major pathways has IkB, and liberation of Nf-kB for nuclear entry
made the study of TNF action difcult, because [91, 175, 201]. Following activation, membrane-
knockout or suppression of Nf-kB is often lethal associated IRAK is degraded [235].
in vivo and in vitro. TRAF-6 interaction with IRAK-1 depends
on a Pro-X-Glu-XX-(aromatic/acidic residue)
sequence shared by RANK [238]. Thus, IkB is
Unique and TNF-Redundant ultimately phosphorylated and Nf-kB enters
Signaling Pathways of IL-1 the nucleus through this double-barreled IL-1
and TNF intracellular signal pathway. As with
IL-1a and b bind receptors of the IL-1R/Toll-like TNF stimulation, IL-1 may also signal via acti-
receptor family to initiate a cascade that culmi- vation of p38 MAPK [117]. The TRAF6/Tab/
nates in end points similar to that of TNF Takk1 cytoplasmic complex directly phospho-
(Figure 5.4). IL-1 binding to its receptor pre- rylates MKK 3, 4, and 6. This leads to the down-
ferentially activates interleukin receptor- stream activation of JNK and p38. The IL-1 and
associated kinase-1 (IRAK-1), a member of a TNF pathways may also cross-stimulate via
family of intracellular proteins that possess IRAK-1, as shown in IRAK-1-decient cells.
active serine-threonine kinase activity. Surpris- In this model, TNF activity was lost but
ingly, IRAK-1 can activate Nf-kB independently was restored through expression of IRAK-1
of its kinase activity (for review, see [88]). In [206, 222].
74 Bone Resorption

IL-1a

IL1R1 IL1R-AcP

MyD88
cytoplasm

To IRAK4

Tab2
llip P Tak1
IRAK1 IRAK1
1
TRAF6 TRAF6 Tab
IKKb
IKKa
P P
lkB
nucleus p50 p56 P MKK

JNK
P
P
p56 N JNK
p50 JU Fra p38

Figure 5.4 IL-1 signaling pathway. IL-1 binds its Toll-like receptor to activate IRAK-1. Successful activation of NFkB downstream
requires a complex that includes TRAF-6 and MyD88. IRAK-4 has an essential kinase activity that phosphorylates IRAK-1. The protein,
Tollip, serves to anchor the IL-1R to MyD88 and other members of the signal complex. An active complex becomes mobile, incorpo-
rates Tab1, Tab2, and Tak1, and transduces the signal inward. TRAF6 now associates with the IKKa and b to phosphorylate IkB and
release NFkB similarly to TNF stimulation. The mobile TRAF6/Tak/Tab complex also phosphorylates MKK and activates JNK and p38
as described in the text. Reprinted from Gene, vol. 321, Nanes MS. Tumor necrosis factor-alpha: molecular and cellular mechanisms
in skeletal pathology, 115 (2003), with permission from Elsevier.

stimulus to osteoclastogenesis [19]. Because


Cytokine Effects on Osteoclast p50/p65 knockout is lethal in midgestation,
Differentiation and Survival Boyce created a p50/p52 double knockout,
depriving Nf-kB of its p50 subunit, and also of
a potentially redundant p52 substitution. The
The Relationship Between TNF and double knockout animals develop osteopetrosis
RANKL in Osteoclastogenesis in the absence of osteoclasts. Interestingly, TNF-
a and TNF-b knockout animals do not exhibit
Osteoclasts are derived from myeloid progeni- the osteopetrotic phenotype; this suggests that
tors of the monocyte macrophage lineage, as additional activators of Nf-kB must exist.
shown in Figure 5.5. The role of TNF as a stim- Later work dened the regulatory pathway
ulator of osteoclastogenesis has been conrmed for osteoclast differentiation in more detail,
by numerous investigators [116, 138, 166]. As including very early steps required to launch the
discussed above, the effect of TNF may be indi- osteoclastic trajectory of differentiation from
rect via osteoblast signals, but it may also be the myelomonocytic progenitors. Mouse knock-
direct [112, 116]. The expression of a number of out models conrm a role for Mi and PU.1 in
transcription factors, including the prototypical the early stages of hematopoietic selection
p65/p50 Nf-kB, is critical to osteoclastogenesis along this lineage [53, 134, 162, 210]. The early
(for detailed review see [7, 80, 82]). The work of progenitors in the marrow then embark on a
Boyce et al. showed that Nf-kB was an essential path toward pre-osteoclasts or monocytes/
Inflammatory Cytokines 75

B cell

marrow
stem cell preosteoclast

RAN
M-CSF cfms RANKL K
NfkB M-CSF
NfkB
PU.1 MI, fos
RANKL RANK fos
+ RAN
K
+ osteoclasts
OPG

TNF-a
stromal cell

T cell apoptosis

Figure 5.5 Role of TNF in osteoclast differentiation. Hematopoietic stem cells differentiate along the myelo-monocytic pathway
with M-CSF stimulation. Differentiating cells continue along a trajectory toward the osteoclast phenotype under the influence of
RANKL, which signals through the transcription factor NfkB. RANKL continues to support survival of the mature functional osteo-
clast. TNF induces the RANKL receptor, RANK, to increase responsiveness to RANKL. Additional factors in regulation include tran-
scription factors PU.1, Mi, and fos; and the M-CSF receptor, cfms. Further details can be found in the text. Reprinted from Gene, vol.
321, Nanes MS. Tumor necrosis factor-alpha: molecular and cellular mechanisms in skeletal pathology, 115 (2003), with permission
from Elsevier.

macrophages under the inuence of M-CSF, ovariectomy-induced acute loss of skeletal


while the soluble factors TNF, IL-1, and RANKL mass. Additional studies quickly established
promote a downstream progression toward the that both OPG levels and the OPG/RANKL
functional osteoclast phenotype, signaling via ratio were regulated by estrogen or androgen. In
the receptors, c-fms, TNFR1, IL1R, and RANK, one interesting clinical trial, provision of OPG
respectively. Once the receptors are occupied, to patients with osteoporosis or rheumatoid
the transcription factors c-fos and Nf-kB enter arthritis reduced biochemical markers of bone
the nucleus to regulate gene transcription [19, resorption [11]. This conrmed a role for the
57, 224, 228, 231, 232, 239] (also see detailed RANKL system in postmenopausal osteoporo-
reviews [7, 20, 199]). In this way, genes that code sis [103, 129, 194, 196]. However, early work
for the mature osteoclast phenotype are acti- demonstrating an important role for TNF in
vated. These include tartrate- resistant acid osteoclastogenesis had to be reconciled with
phosphatase, carbonic anhydrase II, and the data that indicated the importance of the newly
receptors for calcitonin and vitronectin, to discovered RANKL system. In particular, what
name a few. are the relative contributions of TNF and
RANKL, RANK (the RANKL receptor), and RANKL in osteoclastogenesis and do these two
the soluble decoy, OPG, form a particularly signals interact?
potent system to regulate osteoclastogenesis, as Recent work suggests that the RANK/RANKL
conrmed by experiments using transgenic system and TNF expression are interdependent
knockouts. Evidence conrmed a signicant in mice. TNF stimulates expression of the RANK
role for RANKL in skeletal ontogeny and in the receptor and of RANKL. This action in turn
76 Bone Resorption

leads to reciprocal stimulation of TNF by prevented the prolongation of the osteoclast


means of feedback [81, 112, 241, 243], and may lifespan by TNF.
enhance the osteoclastogenic response after
estrogen withdrawal, at least in mice. Indeed,
even in mice, these experimental results may be
strain-dependent. Zou et al. found that the
Effect of TNF on Osteoblasts
RANKL requirement for TNF co-stimulation
was observed in Balb/c, but not in C57BL/6 The maintenance of a healthy skeleton requires
mice, strains that differ in the magnitude of a balance of bone resorption and formation
their inammatory response [243]. rates. To achieve this, bone formation by
On the whole, RANKL only weakly stimulates osteoblasts must increase in response to the
osteoclastogenesis in murine marrow cells that resorptive stimulus of TNF; however, this does
have been derived from TNFR1 knockout mice. not occur. TNF assails bone by stimulating
Similarly, TNF is only weakly osteoclastogenic resorption while simultaneously inhibiting new
in the absence of or at very low concentrations bone formation. Even though the bone forma-
of RANKL [8, 111, 123, 241]. The weight of the tion rate increases somewhat after ovariectomy,
evidence therefore suggests that synergy of the increase is inadequate to compensate for the
RANKL and TNF is required for osteoclastoge- increase in bone resorption [107]. TNF blunts
nesis to occur. Requiring both TNF and RANKL osteoblastic bone formation in four ways: (1) by
makes for more precise control of the number suppressing mature osteoblast function, such as
of osteoclasts because it involves selective sig- the production of a matrix competent for min-
naling of the precursor cells via the two differ- eralization, (2) by blocking osteoblast differen-
ent receptors, and synergism in the stimulation tiation from progenitor cells, (3) by promoting
of Nf-kB and JNK signaling. Interestingly, both apoptosis of mature osteoblasts, and (4) by
TNF and IL-1 increase the expression of OPG, inducing osteoblast resistance to 1,25-(OH)2D3.
the soluble RANKL receptor. This could be the
result of a feedback loop that tempers the potent
RANKL stimulation of osteoclastogenesis [22, Cytokine Regulation of Osteoblast
221]. Gene Expression
Table 5.1 lists some osteoblast genes subject to
Cytokines Increase cytokine regulation. As can be seen, the genes
Osteoclast Survival that are stimulated by TNF are associated with
increased bone resorption, while genes inhib-
Several investigators have shown that TNF and ited by TNF are associated with reduced bone
IL-1 have potent anti-apoptotic effects in osteo- formation. TNF and IL-1 inhibit the production
clasts. Prolongation of osteoclast lifespan may of type I collagen from cultured calvaria and
be an important contribution toward acceler- long bone explants from isolated fetal calvaria
ated bone resorption. The antiapoptotic action osteoblasts, and osteoblastic clonal cell lines [14,
of IL-1 is associated with NfkB activation and 15, 28, 34, 158, 195]. The inhibitory effect of TNF
blocked by inhibition of IkB degradation [94]. on collagen synthesis has been demonstrated by
IL-1 inhibits apoptosis induced by caspase measuring 3[H]-proline incorporation into TCA
activation [164]. precipitable protein and by northern analysis of
The exact signaling that leads from TNF or a1(I) collagen mRNA. Surprisingly, few tran-
IL-1 to apoptosis is now known. Miyazaki et al. scriptional studies on TNF regulation of colla-
have shown that blockade of Nf-kB prevented gen gene transcription (COL1A1 or COL1A2)
bone-resorbing activity but did not prevent IL- have been performed in osteoblasts. However,
1 induced apoptosis, for which ERK was a detailed analysis TNF regulation of the colla-
required [148]. Lee et al. have conrmed that gen promoter has been reported, using transient
ERK is required for TNF- or IL-1-stimulated OC transfection of dermal broblasts or hepatic
survival [124, 126]. These authors have also sug- stellate cells [73, 74]. These studies have
gested that AKT is involved, because inhibition identied putative regulatory regions of the
of the PI3kinase, an upstream activator of AKT, COL1A1 and COL1A2 promoters that confer
Inflammatory Cytokines 77

Table 5.1. Inhibitory and stimulatory effects of cytokines on genes in skeletal cells.
TNF-regulated genes
TNF-inhibited mRNA: TNF-stimulated mRNA:
Inhibition of Bone Formation Stimulation of Bone Resorption
aI collagen [28, 34, 158, 169, 195] Interleukin-6 [37, 77, 120, 130, 154, 172]
Osteocalcin [70, 118, 119, 127, 159] Inducible nitric oxide synthase [83]
Alkaline phosphatase [28, 34, 70, 114, 119, Tissue plasminogen activator/ uPA [132, 169, 170]
155, 198, 240] Plasminogen activator inhibitor-1,
Gelatinases
Insulin-like growth [63, 190, 236] Matrix metalloproteinases [23, 31, 215]
factor-1 Tissue inhibitor of matrix
metalloproteinases
Vitamin D receptor [140] Receptor activator of NfkB [112, 241, 243]
Parathyroid hormone [100, 192] ICAM-1 [120]
receptor
PDGFR [114] Colony-stimulating factors: [37, 52, 86, 98, 131, 237]
CSF-1, GM-CSF, G-CSF
N-cadherin [213] Cyclooxygenase 2 [174, 223]
RUNX2 [64] Interleukin-11 [184]
Osterix [65]
IL-1-regulated genes
IL-1-inhibited mRNA: IL-1-stimulated mRNA:
Inhibition of Bone Formation Stimulation of Bone Resorption
aI collagen [73] Interleukin-6 [21, 122]
Platelet derived growth [114, 230] ICAM-1 [163]
factor receptor
Matrix metalloproteinases 2,3,9,13 [121, 145]
Cyclooxygenase 2 [174, 223]
Phospholipase A2 [174]

TNF inhibitory action. For the COL1A1 pro- IL-1 also inhibits type 1 collagen synthesis
moter, Mori identied two TNF-responsive and transcription of the Col1A1 promoter.
regions in the proximal promoter that bind Studies in mice bearing a collagen promoter-
nuclear proteins and inhibit transcription [147]. chloramphenicol acetyltransferase transgene
Additional studies have shown that the TNF- suggest that IL-1 inhibition of transcription
stimulated transcription factor cEBPb is bound requires new protein synthesis, and may be
to the proximal promoter and that this effect is partly mediated by prostaglandins [73].
blocked by the naturally occurring dominant Osteocalcin is another skeletal matrix pro-
negatives cEBPd and p20cEBPb [75, 76]. TNF tein, second in abundance to type 1 collagen.
also reduces the binding of Sp-1 to the 5- Osteocalcin is 1,25-(OH)2D3-responsive and is
untranslated region of COL1A1 [75, 76, 85]. In produced almost exclusively by osteoblasts. The
the COL1A2 promoter, TNF similarly down-reg- skeletal specicity of osteocalcin and its use
ulates Sp1 binding while stimulating AP-1 and as a marker of the osteoblast phenotype has
Nf-kB (p65/p50) binding to a downstream focused attention on the regulation of its pro-
region, that has been identied as a TGFb moter. TNF decreases the expression of osteo-
responsive site [40, 84, 95, 115]. Mutational calcin mRNA in primary cultures of osteoblasts
analysis of these promoters conrms the role of and in clonal osteoblastic cell lines, an action
Nf-kB as a trans-acting regulator. due to transcriptional repression by Nf-kB [70,
78 Bone Resorption

118, 127, 157, 159]. In contrast to TNF inhibition [213]. The cytokine targets described above all
of type I collagen expression, TNF inhibition contribute to osteoblast bone formation; thus,
of osteocalcin does not involve Nf-kB binding the function of the mature osteoblast is com-
to the osteocalcin promoter [157]. Here the promised in the TNF excess state.
inhibitory mechanism is indirect with TNF An increase in the expression of genes that
causing resistance to 1,25-(OH)2D3 stimulation contribute to the activation of resorption occurs
of the promoter (discussed in detail below). This simultaneously with cytokine inhibition of
mechanism may explain why some investigators bone-forming osteoblast genes. Both IL-1 and
report that TNF only weakly inhibits osteocal- TNF stimulate the activation phase of the bone
cin secretion, whereas TNF inhibition of 1,25- remodeling cycle by stimulating expression
(OH)2D3-stimulated secretion is relatively more of proteolytic enzymes that degrade surface
potent [70, 119, 127, 159, 200]. IL-1 has been osteoid. The degradation of osteoid is quickly
reported to inhibit constitutive and vitamin followed by attachment of bone-resorbing
D-stimulated osteocalcin secretion by tra- osteoclasts. These proteolytic enzymes include
becular osteoblasts, but not of periosteal- matrix metalloproteinases (MMP-1 through 9)
derived osteoblasts [14, 119, 198]. and their inhibitors (TIMPs) [42, 45, 121, 132,
Alkaline phosphatase, which is required for 145, 169, 170, 179, 181, 193, 215]. Stimulation of
normal mineralization, is inhibited by TNF by a metalloproteinase genes by TNF and IL-1 could
complex mechanism [28, 34, 70, 119, 155, 169, increase the frequency of activation of bone
198, 240]. TNF regulation of alkaline phos- remodeling units in the skeleton, thereby con-
phatase occurs at several levels. Although TNF tributing to a net increase in resorption [216].
will decrease alkaline phosphatase mRNA and Synthesis of prostaglandin and of the key
protein, TNF can also acutely stimulate alkaline prostaglandin synthetic enzyme, cyclooxyge-
phosphatase release. TNF-induced apoptosis of nase 2 (COX2), is increased by IL-1 and TNF and
osteoblasts is associated with an immediate may contribute to bone resorption in some
release of soluble alkaline phosphatase [49]. species [44, 48, 74, 101, 104, 139, 146, 174, 202,
TNF may also stimulate retinoic acid-mediated 204, 205, 223, 233].
increases in alkaline phosphatase gene tran- Many signals that promote the differentiation
scription, but simultaneously causes an accu- of osteoclasts are, in fact, osteoblast and marrow
mulation of mature mRNA in the cytoplasm, stromal cell products. These include the TNF-
possibly with translational suppression [135]. induced IL-6, MCSF (CSF-1), RANK, and
Nevertheless, the net effect of TNF is inhibition RANKL [81, 112, 241]. TNF potently stimulates
of skeletal alkaline phosphatase activity. Simi- IL-6 expression by stromal cells and osteoblasts
larly, IL-1, but not IL-6, inhibits alkaline phos- through a transcriptional Nf-kB-dependent
phatase activity in human periosteal osteoblasts mechanism [37, 66, 67, 77, 87, 99, 120, 130, 182,
[119]. 218]. A TNF-stimulated pathway that includes
TNF also suppresses IGF-1 mRNA and secre- Nf-kB activation via TRAF2 has been reported
tion [190]. The effect of TNF on IGF-1 expres- for dendritic cells [137]. Studies in bone and
sion is post-transcriptional, since basal and dendritic cells have provided additional evi-
PTH-stimulated activity of the IGF upstream dence to support a role for p38 kinase and PKC
promoter is not reduced by TNF. In addition, in TNF stimulation of the IL-6 promoter [117,
IGF-1 mRNA stability is not changed by TNF 154]. The way in which TNF stimulates MCSF is
[236]. A role as a potential TNF target for the more controversial [52]. Kaplan et al. [98], have
second downstream IGF-1 promoter has not reported a transcriptional effect of TNF on
been published. MCSF expression. Furthermore, a putative Nf-
Additional TNF-regulated skeletal genes, kB binding site in the CSF-1 promoter can be
shown in Table 5.1, include nuclear receptors for shown to bind the p50 (but not the transactiva-
vitamin D (VDR), which are post-transcription- tional p65) subunit of Nf-kB [237]. However,
ally decreased by TNF [140]; PDGFR-a, inhibition of Nf-kB activation by N-acetyl cys-
decreased by a pre-translational mechanism teine fails to prevent TNF stimulation of both
[114]; and PTHR, decreased by an unknown membrane and soluble forms of MCSF, and TNF
mechanism [71, 100, 192]. TNF decreases cell does not regulate activity of a large MCSF pro-
surface N-cadherins, which may compromise moter-reporter construct; this adds uncertainty
cell-cell communication among osteoblasts to understanding the mechanism [86]. Deletion
Inflammatory Cytokines 79

of the homologous Nf-kB consensus region in (IGF-1), bone morphogenic proteins (BMPs),
the MCSF promoter does not change transcrip- and the transcription factors RUNX2 (AML-3,
tional rate [185]. TNF also stimulates secretion Pepb2aA) and osterix (Osx). Gilbert et al. [63]
of another member of the CSF factors, GM-CSF, have shown that TNF blocks osteoblast differ-
which, in turn, increases secretion of TNF in an entiation in three experimental models: fetal
autocrine manner [37, 51, 69]. calvaria precursor cells, bone marrow stromal
cells, and in the clonal MC3T3-E1 pre-osteoblast
cell line. The critical period of sensitivity to TNF
Effects of Cytokines on Osteoblast occurs when precursor cells commit to differ-
Differentiation entiate along an osteoblastic trajectory. The
block to differentiation involves inhibition of
The steady-state number of osteoblasts results IGF-1, RUNX2 (or Cbfa-1), and Osx, but not
from a balance between their rate of differenti- of BMPs-2, 4, or 6 [63, 65]. IGF-1 may act by
ation and their rate of disappearance (Figure expanding the pre-osteoblast pool through a
5.6). Disappearance from the osteoblast pool mitogenic effect and by providing an anti-
may occur by apoptosis or progression to the apoptotic stimulus. However, treatment with
osteocyte phenotype. TNF affects both the dif- IGF-1 does not overcome the inhibitory effect of
ferentiation rate of osteoblasts from mesenchy- TNF. These results suggest that TNF acts down-
mal precursor cells and also their apoptosis. stream of the IGF-1 stimulus. TNF inhibits
Osteoblasts are derived from pluripotent stem RUNX2 [64], a transcription factor that binds
cells capable of differentiating to adipocytes, skeletal-specic enhancers in the osteocalcin
chondrocytes, broblasts, skeletal muscle cells, promoter [10, 46, 113]. RUNX2 plays a critical
or tendon cells. Factors that stimulate differen- role in skeletal development, because in the
tiation in an osteoblastic direction, include the transgenic knockout there is no bone that min-
secreted proteins, insulin-like growth factor 1 eralizes, only a cartilaginous skeletal pattern

osteoid

PTH BMPs
osteocyte

precursor pre-osteoblast osteoblast + apoptosis

IGF-1 RUNX2, Osx


TNF-a
Figure 5.6 Role of TNF in osteoblast differentiation and survival. PTH and IGF-1 increase the pool of pluripotent precursor cells of
mesenchymal origin. BMP signals the differentiation along an osteoblast/chondrocyte direction. The expression of the transcription
factors RUNX2 and Osx is required for commitment to the osteoblast phenotype. TNF inhibits osteoblast differentiation by suppres-
sion of IGF-1, RUNX2, and Osx expression. Apoptosis of mature osteoblasts is stimulated. Functional consequences of TNF in mature
osteoblasts include inhibition of bone-forming genes, increased expression of genes contributing to bone resorption, and vitamin D
resistance. Reprinted from Gene, vol. 321, Nanes MS. Tumor necrosis factor-alpha: molecular and cellular mechanisms in skeletal
pathology, 115 (2003), with permission from Elsevier.
80 Bone Resorption

devoid of marrow cavity. Many skeletal genes and IL-1 induce apoptosis in osteoblasts. As
require RUNX2 for expression. These include noted above, a receptor death domain, as found
alkaline phosphatase, osteopontin, bone sialo- in TNFR1, must signal through FADD to acti-
protein, and COL1A1, and the transcription vate the caspase cascade. TNF (and IL-1) can
factor cEBPd, all factors important for new bone also augment more powerful apoptotic stimuli
formation [10, 13, 89, 102, 141, 189]. Interest- that are induced by Fas ligand or cycloheximide
ingly, TNF inhibition of RUNX2 is isoform-spe- [212]. Pascher et al. [171] did not nd an effect
cic. TNF almost completely inhibits mRNA and of TNF alone on apoptosis of human osteo-
nuclear protein levels of the ubiquitous short blasts; however, pretreatment of cells with 1,25-
form of RUNX2 (type 1), while inhibition of the (OH)2D3 rendered the cells TNF-sensitive. In an
longer osteoblastic-specic form (type 2, Til-1) interesting report by Bellido et al. [12], cal-
is only 50% [64]. These results suggest that addi- bindin-D28k, a vitamin D-stimulated intracellu-
tional targets of TNF may exist that contribute lar protein, was found to attenuate TNF-induced
to the blockade of differentiation. The TNF apoptosis by directly binding caspase-3. The
effect on RUNX2 includes both transcriptional experimental designs used by Pascher et al. and
suppression and mRNA destabilization [62, 64]. Bellido were quite different, with a very pro-
Although the RUNX2 distal promoter contains longed exposure to vitamin D in the former.
an Nf-kB homologous binding sequence, dele- Additional factors that block or attenuate TNF-
tion of this sequence does not abolish TNF induced apoptosis include dexamethasone, uid
action. An area of the proximal promoter is shear stress, BMP-2 and BMP4, and transform-
required for the inhibition of RUNX2 transcrip- ing growth factor b (TGFb) [36, 38, 39, 173]. The
tion by TNF and also by Nf-kB [64]. Osx, an Sp- relative contribution of apoptosis and rate of
1 family protein that is up-regulated by BMP-2, differentiation in regulating the pool of func-
is required downstream of RUNX2 for skeletal tioning osteoblasts remains controversial. Little
development, as knockout of Osx results in a is known about the effect of cytokines, or other
phenotype similar to that of RUNX2 knockout factors, on further differentiation of osteoblasts
despite RUNX2 expression [156, 234]. The to osteocytes.
mechanism of Osx inhibition by TNF has not
been determined.
Although TNF inhibits osteoblast differentia- TNF Induction of Vitamin
tion, the inuence of this action on the bone for- D Resistance
mation rate in the postmenopausal skeleton
has not been evaluated by histomorphometry. The active metabolite of vitamin D3, 1,25-
Gerstenfeld et al. found that fracture healing was (OH)2D3, contributes to skeletal health by stim-
delayed in TNF receptor-decient mice [61]. In ulating intestinal calcium absorption and also
this model, the inammatory action of TNF may by regulating gene transcription in bone cells.
be needed to replace fracture callus with new Aging and the postmenopausal state are asso-
bone. Uy et al. did not nd an effect of TNF on ciated with some resistance to 1,25-(OH)2D3
bone formation as measured by histomor- action. This can be attributed in part to a reduc-
phometry of nude mice bearing TNF-producing tion in VDR number [5, 128, 229]. A rightward
CHO cell tumors [216]. Neither of these models shift in the vitamin D dose-response has been
is representative of bone in the estrogen- reported in aged or osteoporotic patients with
decient state, but the evidence suggests that the respect to calbindin-9k expression, lymphocyte
physiologic effect of TNF on bone formation mitogenesis, and intestinal calcium absorption.
may be complex. A lack of effect of TNF on basal This shift may not be entirely due to a decrease
bone formation rate would not preclude a pos- in VDR number [128, 160]. TNF has been
sible inhibitory action in the postmenopausal shown to decrease vitamin D receptor number
skeleton in the estrogen-decient state. post-transcriptionally, and to decrease 1,25-
(OH)2D3-stimulated receptor transactivation in
osteoblastic cells [140, 159]. Interestingly, TNF
Effects of Cytokines on treatment reduced the ligand-stimulated
Osteoblast Survival binding of the VDR/RXR (retinoid X receptor)
heterodimer to the VDRE of the osteocalcin
Most studies [78, 92, 109] (see also references in promoter. Deletion analysis of the osteocalcin
Hock et al. [79]) support the notion that TNF promoter revealed that the vitamin D-
Inflammatory Cytokines 81

responsive element (VDRE) conferred TNF sup- occupied by the steroid receptor and Nf-kB and
pression, even when the VDRE was cloned into as a result blunts transcriptional activation.
a heterologous viral promoter [118, 157]. Tran- The p65 subunit of Nf-kB does not directly
scriptional activation by a liganded VDR can bind the VDR (Nanes, unpublished). Nf-kB
therefore be suppressed by TNF-stimulated repression of VDR has recently been shown to
signals. Subsequently, Farmer et al. have shown require the Rel homology domain of p65, which
that transcriptional suppression can be induced includes one of the CBP binding regions [133].
by forced expression of the p65 subunit of Nf- Thus, involvement of coactivator interface for
kB, but not by the p50 subunit [50]. A recipro- both p65 and steroid receptors is required for
cal suppression of Nf-kB transactivation of the transrepression. Lu et al. [133] have identied an
IL-8 promoter by 1,25(OH)2D3 may involve a additional midmolecular region of p65 that is
mechanism that inhibits Nf-kB binding to its also required for transrepression of VDR. This
cognate response element [72]. region includes a number of serine and tyrosine
The regulation of VDR function by Nf-kB is phosphorylation targets previously found to be
not surprising, inasmuch as a similar trans- required for p65 transcriptional activity. Dele-
repression of glucocorticoid receptors (GR) and tion of this region or of individual serine or
retinoid X receptor (RXR) transactivation has tyrosines reverses most of the p65 repression of
been reported [3, 27, 54, 191]. The interaction VDR. These results add support to the concept
between the GR and Nf-kB is reciprocal. Steroid that p65 interacts with nuclear proteins that are
receptors1 that repress Nf-kB transactivation shared by the VDR transcriptional complex so
include the GR, progesterone receptor (PR), and as to reduce transactivational potency of the
the VDR [27, 72]. The mechanism of Nf-kB reg- VDR. What coactivators are involved has not
ulation of steroid receptor function is contro- been determined.
versial. An Nf-kB response element in the target
gene is not required for Nf-kB repression of
either VDR or GR transactivation. The repres-
sion mechanism therefore may involve a
Conclusion
protein-protein interaction. Further support for
this concept is that GR repression of Nf-kB is Although many cytokines contribute to the
limited to the a isoform of the GR and localizes pathophysiology of bone loss, TNF has an
to a region of the GR that encompasses the coac- important and central role. A normally coupled
tivator interface [24, 217]. Possible mechanisms resorption/formation system, important for the
for Nf-kB trans-repression include (1) direct homeostasis of bone, is disturbed in the pres-
binding of Nf-kB to steroid receptors; (2) Nf-kB ence of elevated TNF. Such elevations may be
competition or co-occupancy of general tran- acute, as in a are of inammatory arthritis, or
scription proteins, including CBP or SRC-1; (3) insidious, as with estrogen loss and aging.
competition for other nuclear proteins that Increased production of TNF damages bone by
bridge the steroid receptor-DNA complex to increasing bone resorption while simultane-
polymerase II; or (4) direct modication of ously inhibiting bone-forming osteoblasts. This
polymerase II activity by TNF or Nf-kB. De dual effect of TNF is a potent stimulus to skele-
Bosscher et al. have reported that GR repression tal loss that ultimately leads to microarchitec-
of Nf-kB, or the reciprocal p65 repression of GR, tural deterioration and increased fracture risk.
occurs regardless of the level of CBP or SRC-1 As discussed above, no single mechanism is
in the cell, a nding that does not exclude poten- responsible for TNF action on bone cells; rather,
tial competition for other nuclear coactivators activation of a complex signal pathway drives
[43]. Nissen et al. [161] have reported that the gene transcription toward a skeletal catabolic
GR zinc nger domain (including the coactiva- state. TNF cooperates with other signal path-
tor interface) can bind directly to the coactiva- ways activated by IL-1 and additional cytokines
tor interface of p65. From the available data it and the RANKL system, and also suppresses
seems that the transcriptional complex is co- vitamin D action to impair bone at the cell,
tissue, and physiologic levels. The accumulated
evidence suggests that targeting of cytokine
1
The term steroid receptor is used here to represent the action by interfering with signal pathways from
nuclear receptors of the thyroid/retinoid/estrogen super- the TNFR1 or IL-1 receptors, through TRAFs,
family including the VDR. IRAKs, and nally Nf-kB, remains an important
82 Bone Resorption

goal for the treatment of osteoporosis. Many 11. Bekker PJ, Holloway D, Nakanishi A, Arrighi M, Leese
questions remain to be answered about PT, Dunstan CR (2001) The effect of a single dose of
osteoprotegerin in postmenopausal women. J Bone
cytokine action in bone. What are the key early Miner Res 16:34860.
genes that are activated (or suppressed) by TNF 12. Bellido T, Huening M, Raval-Pandya M, Manolagas SC,
and IL-1? How can we capitalize on the mecha- Christakos S (2000) Calbindin-D28k is expressed in
nism of TNF stimulation of RANK/RANKL to osteoblastic cells and suppresses their apoptosis by
inhibiting caspase-3 activity. J Biol Chem 275:26328
block osteoclastogenesis? Can we circumvent 32.
TNF suppression of osteoblastogenesis to 13. Benson MD, Aubin JE, Xiao G, Thomas PE, Franceschi
promote increased bone formation rates? RT (1999) Cloning of a 2.5 kb murine bone sialoprotein
Further investigation of cytokine action in bone promoter fragment and functional analysis of putative
is needed to answer these questions and provide Osf2 binding sites. J Bone Min Res 14:396405.
14. Beresford JN, Gallagher JA, Gowen M, Couch M, Poser
interventions for bone loss in the aging J, Wood DD, et al. (1984) The effects of monocyte-
skeleton. conditioned medium and interleukin 1 on the synthe-
sis of collagenous and non-collagenous proteins by
mouse bone and human bone cells in vitro. Biochim
Biophys Acta 801:5865.
References 15. Bertolini DR, Nedwin GE, Bringman TS, Smith DD,
Mundy GR (1986) Stimulation of bone resorption and
inhibition of bone formation in vitro by human
1. Abrahamsen B, Shalhoub V, Larson EK, Eriksen EF, tumour necrosis factors. Nature 319:51618.
Beck-Nielsen H, Marks SC, Jr. (2000) Cytokine RNA 16. Birkenkamp KU, Tuyt LM, Lummen C, Wierenga AT,
levels in transiliac bone biopsies from healthy early Kruijer W, Vellenga E (2000) The p38 MAP kinase
postmenopausal women. Bone 26:13745. inhibitor SB203580 enhances nuclear factor-kappa B
2. Abu-Amer Y, Erdmann J, Alexopoulou L, Kollias G, transcriptional activity by a non-specic effect upon
Ross FP, Teitelbaum SL (2000) Tumor necrosis factor the ERK pathway. Br J Pharmacol 131:99107.
receptors types 1 and 2 differentially regulate osteo- 17. Bodine PV,Vernon SK, Komm BS (1996) Establishment
clastogenesis. J Biol Chem 275:2730710. and hormonal regulation of a conditionally trans-
3. Adcock IM, Shirasaki H, Gelder CM, Peters MJ, Brown formed preosteocytic cell line from adult human bone.
CR, Barnes PJ (1994) The effects of glucocorticoids on Endocrinology 137:4592604.
phorbol ester and cytokine stimulated transcription 18. Boss JM, Jensen PE (2003) Transcriptional regulation
factor activation in human lung. Life Sci 55:1147 of the MHC class II antigen presentation pathway. Curr
53. Opin Immunol 15:10511.
4. An J, Ribeiro RC, Webb P, Gustafsson JA, Kushner PJ, 19. Boyce BF, Xing L, Franzoso G, Siebenlist U (1999)
Baxter JD, et al. (1999) Estradiol repression of tumor Required and nonessential functions of nuclear factor-
necrosis factor-a transcription requires estrogen kappa B in bone cells. Bone 25:1379.
receptor activation function-2 and is enhanced by 20. Boyle WJ, Simonet WS, Lacey DL (2003) Osteoclast dif-
coactivators. Proc Natl Acad Sci USA 96:151616. ferentiation and activation. Nature 423:33742.
5. Armbrecht HJ, Boltz MA, Christakos S, Bruns ME 21. Bradford PG, Maglich JM, Kirkwood KL (2000) IL-1
(1998) Capacity of 1,25-dihydroxyvitamin D to stimu- beta increases type 1 inositol trisphosphate receptor
late expression of calbindin D changes with age in the expression and IL-6 secretory capacity in osteoblastic
rat. Arch Biochem Biophys 352:15964. cell cultures. Mol Cell Biol Res Commun 3:735.
6. Atkins GJ, Haynes DR, Geary SM, Loric M, Crotti TN, 22. Brandstrom H, Jonsson KB, Vidal O, Ljunghall S,
Findlay DM (2000) Coordinated cytokine expression Ohlsson C, Ljunggren O (1998) Tumor necrosis factor-
by stromal and hematopoietic cells during human a and -beta upregulate the levels of osteoprotegerin
osteoclast formation. Bone 26:65361. mRNA in human osteosarcoma MG-63 cells. Biochem
7. Aubin JE, Bonnelye E (2000) Osteoprotegerin and its Biophys Res Commun 248:4547.
ligand: a new paradigm for regulation of osteoclasto- 23. Brennan FM, Browne KA, Green PA, Jaspar JM, Maini
genesis and bone resorption. Osteoporos Int 11:905 RN, Feldmann M (1997) Reduction of serum matrix
13. metalloproteinase 1 and matrix metalloproteinase 3 in
8. Azuma Y, Kaji K, Katogi R, Takeshita S, Kudo A (2000) rheumatoid arthritis patients following anti-tumour
Tumor necrosis factor-a induces differentiation of necrosis factor-a (cA2) therapy. Br J Rheumatol 36:
and bone resorption by osteoclasts. J Biol Chem 275: 64350.
485864. 24. Brogan IJ, Murray IA, Cerillo G, Needham M, White A,
9. Baker PJ, Dixon M, Evans RT, Dufour L, Johnson E, Davis JR (1999) Interaction of glucocorticoid receptor
Roopenian DC (1999) CD4(+) T cells and the proin- isoforms with transcription factors AP-1 and Nf-kB:
ammatory cytokines gamma interferon and inter- lack of effect of glucocorticoid receptor beta. Mol Cell
leukin-6 contribute to alveolar bone loss in mice. Infect Endocrinol 157:95104.
Immun 67:28049. 25. Burns K, Clatworthy J, Martin L, Martinon F, Plumpton
10. Banerjee C, McCabe LR, Choi JY, Hiebert SW, Stein JL, C, Maschera B, et al. (2000) Tollip, a new component of
Stein GS, et al. (1997) Runt homology domain proteins the IL-1RI pathway, links IRAK to the IL-1 receptor. Nat
in osteoblast differentiation: AML3/CBFA1 is a major Cell Biol 2:34651.
component of a bone-specic complex. J Cell Biochem 26. Burow ME, Weldon CB, Tang Y, McLachlan JA,
66:18. Beckman BS (2001) Oestrogen-mediated suppression
Inflammatory Cytokines 83

of tumour necrosis factor a -induced apoptosis in 41. Darnay BG, Ni J, Moore PA, Aggarwal BB (1999) Acti-
MCF-7 cells: subversion of Bcl-2 by anti-oestrogens. J vation of Nf-kB by RANK requires tumor necrosis
Steroid Biochem Mol Biol 78:40918. factor receptor-associated factor (TRAF) 6 and Nf-kB
27. Caldenhoven E, Liden J, Wissink S, Van de Stolpe A, -inducing kinase. Identication of a novel TRAF6
Raaijmakers J, Koenderman L, et al. (1995) Negative interaction motif. J Biol Chem 274:772431.
cross-talk between RelA and the glucocorticoid 42. de Bart AC, Quax PH, Lowik CW, Verheijen JH (1995)
receptor: a possible mechanism for the antiinamma- Regulation of plasminogen activation, matrix
tory action of glucocorticoids. Mol Endocrinol 9: metalloproteinases and urokinase-type plasminogen
40112. activator-mediated extracellular matrix degradation in
28. Canalis E (1987) Effects of tumor necrosis factor on human osteosarcoma cell line MG63 by interleukin-1
bone formation in vitro. Endocrinology. 121:1596604. a. J Bone Miner Res 10:137484.
29. Cao Z, Xiong J, Takeuchi M, Kurama T, Goeddel DV 43. De Bosscher K, Vanden Berghe W, Vermeulen L,
(1996) TRAF6 is a signal transducer for interleukin-1. Plaisance S, Boone E, Haegeman G (2000) Glucocorti-
Nature 383:4436. coids repress Nf-kB -driven genes by disturbing the
30. Carpentier I, Beyaert R (1999) TRAF1 is a TNF interaction of p65 with the basal transcription machin-
inducible regulator of Nf-kB activation. FEBS Lett ery, irrespective of coactivator levels in the cell. Proc
460:24650. Natl Acad Sci USA 97:391924.
31. Catrina AI, Lampa J, Ernestam S, af Klint E, Bratt J, 44. de Brum-Fernandes AJ, Laporte S, Heroux M, Lora M,
Klareskog L, et al. (2002) Anti-tumour necrosis factor Patry C, Menard HA, et al. (1994) Expression of pro-
(TNF)-_a therapy (etanercept) down-regulates serum staglandin endoperoxide synthase-1 and prostag-
matrix metalloproteinase (MMP)-3 and MMP-1 in landin endoperoxide synthase-2 in human osteoblasts.
rheumatoid arthritis. Rheumatology (Oxford) 41: Biochem Biophys Res Commun 198:95560.
4849. 45. Delaisse JM, Eeckhout Y,Vaes G (1988) Bone-resorbing
32. Cenci S, Toraldo G, Weitzmann MN, Roggia C, Gao Y, agents affect the production and distribution of pro-
Qian WP, et al. (2003) Estrogen deciency induces bone collagenase as well as the activity of collagenase in
loss by increasing T cell proliferation and lifespan bone tissue. Endocrinology 123:26476.
through IFN-gamma-induced class II transactivator. 46. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G
Proc Natl Acad Sci USA 100:1040510. (1997) Osf2/Cbfa1: a transcriptional activator of
33. Cenci S, Weitzmann MN, Roggia C, Namba N, Novack osteoblast differentiation Cell 89:74754.
D, Woodring J, et al. (2000) Estrogen deciency induces 47. Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ,
bone loss by enhancing T-cell production of TNF-a. Lacey DL, Riggs BL (2003) Role of RANK ligand in
J Clin Invest 106:122937. mediating increased bone resorption in early post-
34. Centrella M, McCarthy TL, Canalis E (1988) Tumor menopausal women. J Clin Invest 111:122130.
necrosis factor-a inhibits collagen synthesis and alka- 48. Evans DB, Bunning RA, Russell RG (1990) The effects
line phosphatase activity independently of its effect on of recombinant human interleukin-1 beta on cellular
deoxyribonucleic acid synthesis in osteoblast- proliferation and the production of prostaglandin
enriched bone cell cultures. Endocrinology 123:1442 E2, plasminogen activator, osteocalcin and alkaline
8. phosphatase by osteoblast-like cells derived from
35. Chabaud M, Miossec P (2001) The combination of human bone. Biochem Biophys Res Commun 166:208
tumor necrosis factor a blockade with interleukin-1 16.
and interleukin-17 blockade is more effective for con- 49. Farley JR, Stilt-Cofng B (2001) Apoptosis may deter-
trolling synovial inammation and bone resorption in mine the release of skeletal alkaline phosphatase activ-
an ex vivo model. Arthritis Rheum 44:1293303. ity from human osteoblast-line cells. Calcif Tissue Int
36. Chae HJ, Chae SW, Kang JS, Bang BG, Cho SB, Park RK, 68:4352.
et al. (2000) Dexamethasone suppresses tumor necro- 50. Farmer PK, He X, Schmitz ML, Rubin J, Nanes MS
sis factor-a induced apoptosis in osteoblasts: possi- (2000) Inhibitory effect of Nf-kB on 1,25-dihydroxyvi-
ble role for ceramide. Endocrinology 141:290413. tamin D(3) and retinoid X receptor function. Am J
37. Chaudhary LR, Spelsberg TC, Riggs BL (1992) Produc- Physiol Endocrinol Metab 279:E21320.
tion of various cytokines by normal human osteoblast- 51. Felix R, Cecchini MG, Hofstetter W, Guenther HL,
like cells in response to interleukin-1 beta and tumor Fleisch H (1991) Production of granulocyte-
necrosis factor-a: lack of regulation by 17 beta- macrophage (GM-CSF) and granulocyte colony-
estradiol. Endocrinology 130:252834. stimulating factor (G-CSF) by rat clonal osteoblastic
38. Chen S, Guttridge DC, Tang E, Shi S, Guan K, Wang CY cell population CRP 10/30 and the immortalized cell
(2001) Suppression of tumor necrosis factor-mediated line IRC10/30-myc1 stimulated by tumor necrosis
apoptosis by nuclear factor kappaB-independent bone factor a. Endocrinology 128:6617.
morphogenetic protein/Smad signaling. J Biol Chem 52. Felix R, Fleisch H, Elford PR (1989) Bone-resorbing
276:3925963. cytokines enhance release of macrophage colony-
39. Chua CC, Chua BH, Chen Z, Landy C, Hamdy RC (2002) stimulating activity by the osteoblastic cell MC3T3-E1.
TGF-beta1 inhibits multiple caspases induced by TNF- Calcif Tissue Int 44:35660.
a in murine osteoblastic MC3T3-E1 cells. Biochim 53. Feng X, Teitelbaum SL, Quiroz ME, Cheng SL, Lai CF,
Biophys Acta 1593:18. Avioli LV, et al. (2000) Sp1/Sp3 and PU.1 differentially
40. Chung KY, Agarwal A, Uitto J, Mauviel A (1996) An AP- regulate beta(5) integrin gene expression in macro-
1 binding sequence is essential for regulation of the phages and osteoblasts. J Biol Chem 275:8331
human a2(I) collagen (COL1A2) promoter activity by 40.
transforming growth factor-beta. J Biol Chem 271: 54. Fernandez-Martin JL, Kurian S, Farmer P, Nanes MS
32728. (1998) Tumor necrosis factor activates a nuclear
84 Bone Resorption

inhibitor of vitamin D and retinoid-X receptors. Mol 70. Gowen M, MacDonald BR, Russell RG (1988) Actions
Cell Endocrinol 141:6572. of recombinant human gamma-interferon and tumor
55. Fox HS, Bond BL, Parslow TG (1991) Estrogen regu- necrosis factor a on the proliferation and osteoblastic
lates the IFN-gamma promoter. J Immunol 146:43627. characteristics of human trabecular bone cells in vitro.
56. Fox SW, Chambers TJ (2000) Interferon-gamma Arthritis Rheum 31:15007.
directly inhibits TRANCE-induced osteoclastogenesis. 71. Hanevold CD, Yamaguchi DT, Jordan SC (1993) Tumor
Biochem Biophys Res Commun 276:86872. necrosis factor a modulates parathyroid hormone
57. Franzoso G, Carlson L, Xing L, Poljak L, Shores EW, action in UMR-106-01 osteoblastic cells. J Bone Miner
Brown KD, et al. (1997) Requirement for Nf-kB in Res 8:1191200.
osteoclast and B-cell development. Genes Dev 11: 72. Harant H, Andrew PJ, Reddy GS, Foglar E, Lindley IJ
348296. (1997) 1 a,25-dihydroxyvitamin D3 and a variety of its
58. Fuller K, Murphy C, Kirstein B, Fox SW, Chambers TJ natural metabolites transcriptionally repress nuclear-
(2002) TNF a potently activates osteoclasts, through a factor-kappaB-mediated interleukin-8 gene expres-
direct action independent of and strongly synergistic sion. Eur J Biochem 250:6371.
with RANKL. Endocrinology 143:110818. 73. Harrison JR, Kleinert LM, Kelly PL, Krebsbach PH,
59. Galibert L, Tometsko ME, Anderson DM, Cosman D, Woody C, Clark S, et al. (1998) Interleukin-1 represses
Dougall WC (1998) The involvement of multiple tumor COLIA1 promoter activity in calvarial bones of trans-
necrosis factor receptor (TNFR)-associated factors in genic ColCAT mice in vitro and in vivo. J Bone Miner
the signaling mechanisms of receptor activator of Nf- Res 13:107683.
kB, a member of the TNFR superfamily. J Biol Chem. 74. Harrison JR, Lorenzo JA, Kawaguchi H, Raisz LG,
273:341207. Pilbeam C (1994) Stimulation of prostaglandin E2 pro-
60. Galien R, Garcia T (1997) Estrogen receptor impairs duction by interleukin-1a and transforming growth
interleukin-6 expression by preventing protein bind- factor a in osteoblastic MC3T3-E1 cells. J Bone Miner
ing on the Nf-kB site. Nucleic Acids Res 25:2424 Res 9:81723.
9. 75. Hatamochi A, Mori K, Ueki H (1994) Role of cytokines
61. Gerstenfeld LC, Cho TJ, Kon T, Aizawa T, Cruceta J, in controlling connective tissue gene expression. Arch
Graves BD, et al. (2001) Impaired intramembranous Dermatol Res 287:11521.
bone formation during bone repair in the absence of 76. Hernandez I, de la Torre P, Rey-Campos J, Garcia I,
tumor necrosis factor-a signaling. Cells Tissues Sanchez JA, Munoz R, et al. (2000) Collagen a1(I) gene
Organs 169:28594. contains an element responsive to tumor necrosis
62. Gilbert L, Brantley A, Rubin J, Nanes MS (2002) The p55 factor-a located in the 5 untranslated region of its rst
TNF receptor mediates TNF inhibition of osteoblast exon. DNA Cell Biol 19:34152.
differentiation. J Bone Miner Res 17:SU205. 77. Hierl T, Borcsok I, Sommer U, Ziegler R, Kasperk C
63. Gilbert L, He X, Farmer P, Boden S, Kozlowski M, Rubin (1998) Regulation of interleukin-6 expression in
J, et al. (2000) Inhibition of osteoblast differentiation human osteoblastic cells in vitro. Exp Clin Endocrinol
by tumor necrosis factor-a. Endocrinology 141:3956 Diabetes 106:32433.
64. 78. Hill PA, Tumber A, Meikle MC (1997) Multiple extra-
64. Gilbert L, He X, Farmer P, Rubin J, Drissi H, van WAJ, cellular signals promote osteoblast survival and apop-
et al. (2002) Expression of the osteoblast differentia- tosis. Endocrinology 138:384958.
tion factor RUNX2 (Cbfa1/AML3/Pebp2 aA) is inhib- 79. Hock JM, Krishnan V, Onyia JE, Bidwell JP, Milas J,
ited by tumor necrosis factor-a. J Biol Chem 277:2695 Stanislaus D (2001) Osteoblast apoptosis and bone
701. turnover. J Bone Miner Res 16:97584.
65. Gilbert LC, He, X., Kauppi, A., Rubin, J., Nanes, M.S. 80. Hofbauer LC, Heufelder AE (2001) Role of receptor
(2003) Inhibition of osteoblast differentiation by TNF- activator of nuclear factor-kappaB ligand and osteo-
a is associated with suppression of osterix expression. protegerin in bone cell biology. J Mol Med 79:243
J Bone Mineral Res 18:SU177. 53.
66. Gimble JM, Hudson J, Henthorn J, Hua XX, Burstein 81. Hofbauer LC, Lacey DL, Dunstan CR, Spelsberg TC,
SA (1991) Regulation of interleukin 6 expression in Riggs BL, Khosla S (1999) Interleukin-1beta and tumor
murine bone marrow stromal cells. Exp Hematol 19: necrosis factor-a, but not interleukin-6, stimulate
105560. osteoprotegerin ligand gene expression in human
67. Girasole G, Jilka RL, Passeri G, Boswell S, Boder G, osteoblastic cells. Bone 25:2559.
Williams DC, et al. (1992) 17 beta-estradiol inhibits 82. Horowitz MC, Xi Y, Wilson K, Kacena MA (2001)
interleukin-6 production by bone marrow-derived Control of osteoclastogenesis and bone resorption by
stromal cells and osteoblasts in vitro: a potential mech- members of the TNF family of receptors and ligands.
anism for the antiosteoporotic effect of estrogens. J Cytokine Growth Factor Rev 12:918.
Clin Invest 89:88391. 83. Hukkanen M, Hughes FJ, Buttery LD, Gross SS, Evans
68. Gourley T, Roys S, Lukacs NW, Kunkel SL, Flavell RA, TJ, Seddon S, et al. (1995) Cytokine-stimulated expres-
Chang CH (1999) A novel role for the major histo- sion of inducible nitric oxide synthase by mouse, rat,
compatibility complex class II transactivator CIITA in and human osteoblast-like cells and its functional role
the repression of IL-4 production. Immunity 10: in osteoblast metabolic activity. Endocrinology 136:
37786. 544553.
69. Gowen M, Chapman K, Littlewood A, Hughes D, Evans 84. Inagaki Y, Truter S, Tanaka S, Di Liberto M, Ramirez F
D, Russell G (1990) Production of tumor necrosis (1995) Overlapping pathways mediate the opposing
factor by human osteoblasts is modulated by other actions of tumor necrosis factor-a and transforming
cytokines, but not by osteotropic hormones. Endo- growth factor-beta on a2(I) collagen gene transcrip-
crinology 126:12505. tion. J Biol Chem 270:33538.
Inflammatory Cytokines 85

85. Iraburu MJ, Dominguez-Rosales JA, Fontana L, Auster scription of the colony-stimulating factor-1 gene in
A, Garcia-Trevijano ER, Covarrubias-Pinedo A, et al. murine osteoblasts. J Cell Physiol 168:199208.
(2000) Tumor necrosis factor a down-regulates expres- 99. Kassem M, Harris SA, Spelsberg TC, Riggs BL (1996)
sion of the a1(I) collagen gene in rat hepatic stellate Estrogen inhibits interleukin-6 production and gene
cells through a p20C/EBPbeta- and C/EBPdelta- expression in a human osteoblastic cell line with high
dependent mechanism. Hepatology 31:108693. levels of estrogen receptors. J Bone Miner Res 11:
86. Isaacs SD, Fan X, Fan D, Gewant H, Murphy TC, Farmer 1939.
P, et al. (1999) Role of NFkappaB in the regulation of 100. Katz MS, Gutierrez GE, Mundy GR, Hymer TK,
macrophage colony stimulating factor by tumor necro- Cauleld MP, McKee RL (1992) Tumor necrosis factor
sis factor-a in ST2 bone stromal cells. J Cell Physiol and interleukin 1 inhibit parathyroid hormone-
179:193200. responsive adenylate cyclase in clonal osteoblast-like
87. Ishimi Y, Miyaura C, Jin CH, Akatsu T, Abe E, cells by down-regulating parathyroid hormone recep-
Nakamura Y, et al. (1990) IL-6 is produced by tors. J Cell Physiol 153:20613.
osteoblasts and induces bone resorption. J Immunol 101. Kawaguchi H,Yavari R, Stover ML, Rowe DW, Raisz LG,
145:3297303. Pilbeam CC (1994) Measurement of interleukin-1
88. Janssens S, Beyaert R (2003) Functional diversity stimulated constitutive prostaglandin G/H synthase
and regulation of different interleukin-1 receptor- (cyclooxygenase) mRNA levels in osteoblastic MC3T3-
associated kinase (IRAK) family members. Mol Cell E1 cells using competitive reverse transcriptase poly-
11:293302. merase chain reaction. Endocr Res 20:21933.
89. Javed A, Barnes GL, Jasanya BO, Stein JL, Gerstenfeld L, 102. Kern B, Shen J, Starbuck M, Karsenty G (2001) Cbfa1
Lian JB, et al. (2001) runt homology domain tran- contributes to the osteoblast-specic expression of
scription factors (Runx, Cbfa, and AML) mediate type I collagen genes. J Biol Chem 276:71017.
repression of the bone sialoprotein promoter: evidence 103. Khosla S, Atkinson EJ, Dunstan CR, OFallon WM
for promoter context-dependent activity of Cbfa pro- (2002) Effect of estrogen versus testosterone on cir-
teins. Mol Cell Biol 21:2891905. culating osteoprotegerin and other cytokine levels
90. Jefferies C, Bowie A, Brady G, Cooke EL, Li X, ONeill in normal elderly men. J Clin Endocrinol Metab
LA (2001) Transactivation by the p65 subunit of Nf-kB 87:15504.
in response to interleukin-1 (IL-1) involves MyD88, IL- 104. Kim CH, Kang BS, Lee TK, Park WH, Kim JK, Park
1 receptor-associated kinase 1, TRAF-6, and Rac1. Mol YG, et al. (2002) IL-1beta regulates cellular prolifera-
Cell Biol 21:454452. tion, prostaglandin E2 synthesis, plasminogen
91. Jiang Z, Ninomiya-Tsuji J, Qian Y, Matsumoto K, Li X activator activity, osteocalcin production, and bone
(2002) Interleukin-1 (IL-1) receptor-associated kinase- resorptive activity of the mouse calvarial bone cells.
dependent IL-1-induced signaling complexes phos- Immunopharmacol Immunotoxicol 24:395407.
phorylate TAK1 and TAB2 at the plasma membrane 105. Kim YM, Im JY, Han SH, Kang HS, Choi I (2000) IFN-
and activate TAK1 in the cytosol. Mol Cell Biol 22: gamma up-regulates IL-18 gene expression via IFN
715867. consensus sequence-binding protein and activator
92. Jilka RL, Weinstein RS, Bellido T, Partt AM, protein-1 elements in macrophages. J Immunol 165:
Manolagas SC (1998) Osteoblast programmed cell 3198205.
death (apoptosis): modulation by growth factors and 106. Kimble RB, Bain S, Pacici R (1997) The functional
cytokines. J Bone Miner Res 13:793802. block of TNF but not of IL-6 prevents bone loss
93. Jimi E, Ikebe T, Takahashi N, Hirata M, Suda T, Koga T in ovariectomized mice. J Bone Miner Res 12:935
(1996) Interleukin-1a activates all NF-kappa-b-like 41.
factor in osteoclast-like cells. Journal of Biological 107. Kimble RB, Matayoshi AB, Vannice JL, Kung VT,
Chemistry 271:46054608. Williams C, Pacici R (1995) Simultaneous block of
94. Jimi E, Nakamura I, Ikebe T, Akiyama S, Takahashi N, interleukin-1 and tumor necrosis factor is required to
Suda T (1998) Activation of Nf-kB is involved in the completely prevent bone loss in the early postovariec-
survival of osteoclasts promoted by interleukin-1. J tomy period. Endocrinology 136:305461.
Biol Chem 273:8799805. 108. Kimble RB, Srivastava S, Ross FP, Matayoshi A, Pacici
95. Kahari VM, Chen YQ, Su MW, Ramirez F, Uitto J (1990) R (1996) Estrogen deciency increases the ability of
Tumor necrosis factor-a and interferon-gamma sup- stromal cells to support murine osteoclastogenesis via
press the activation of human type I collagen gene an interleukin-1and tumor necrosis factor-mediated
expression by transforming growth factor-beta 1. Evi- stimulation of macrophage colony-stimulating factor
dence for two distinct mechanisms of inhibition at the production. J Biol Chem 271:288907.
transcriptional and posttranscriptional levels. J Clin 109. Kitajima I, Soejima Y, Takasaki I, Beppu H, Tokioka T,
Invest 86:148995. Maruyama I (1996) Ceramide-induced nuclear trans-
96. Kaji K, Katogi R, Azuma Y, Naito A, Inoue JI, Kudo A location of NF-kappa B is a potential mediator of the
(2001) Tumor necrosis factor a -induced osteoclasto- apoptotic response to TNF-a in murine clonal
genesis requires tumor necrosis factor receptor- osteoblasts. Bone 19:26370.
associated factor 6. J Bone Miner Res 16:15939. 110. Kitazawa R, Kimble RB, Vannice JL, Kung VT, Pacici R
97. Kamada M, Irahara M, Maegawa M, Ohmoto Y, Murata (1994) Interleukin-1 receptor antagonist and tumor
K, Yasui T, et al. (2001) Transient increase in the levels necrosis factor binding protein decrease osteoclast for-
of T-helper 1 cytokines in postmenopausal women and mation and bone resorption in ovariectomized mice. J
the effects of hormone replacement therapy. Gynecol Clin Invest 94:2397406.
Obstet Invest 52:828. 111. Kobayashi K, Takahashi N, Jimi E, Udagawa N, Takami
98. Kaplan DL, Eielson CM, Horowitz MC, Insogna KL, M, Kotake S, et al. (2000) Tumor necrosis factor a stim-
Weir EC (1996) Tumor necrosis factor-a induces tran- ulates osteoclast differentiation by a mechanism inde-
86 Bone Resorption

pendent of the ODF/RANKL-RANK interaction. J Exp in vitro osteoclastogenesis. Endocrinology 144:3524


Med 191:27586. 31.
112. Komine M, Kukita A, Kukita T, Ogata Y, Hotokebuchi T, 126. Lee ZH, Lee SE, Kim CW, Lee SH, Kim SW, Kwack K,
Kohashi O (2001) Tumor necrosis factor-a cooperates et al. (2002) IL-1a stimulation of osteoclast survival
with receptor activator of nuclear factor kappaB ligand through the PI 3-kinase/Akt and ERK pathways. J
in generation of osteoclasts in stromal cell-depleted rat Biochem (Tokyo) 131:1616.
bone marrow cell culture. Bone 28:47483. 127. Li YP, Stashenko P (1992) Proinammatory cytokines
113. Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, tumor necrosis factor-a and IL-6, but not IL-1, down-
Deguchi K, et al. (1997) Targeted disruption of Cbfa1 regulate the osteocalcin gene promoter. J Immunol
results in a complete lack of bone formation owing to 148:78894.
maturational arrest of osteoblasts. Cell 89:75564. 128. Liel Y, Shany S, Smirnoff P, Schwartz B (1999) Estrogen
114. Kose KN, Xie JF, Carnes DL, Graves DT (1996) Pro- increases 1,25-dihydroxyvitamin D receptors expres-
inammatory cytokines downregulate platelet derived sion and bioresponse in the rat duodenal mucosa.
growth factor-a receptor gene expression in human Endocrinology 140:2805.
osteoblastic cells. J Cell Physiol 166:18897. 129. Lindberg MK, Erlandsson M, Alatalo SL, Windahl
115. Kouba DJ, Chung KY, Nishiyama T, Vindevoghel L, S, Andersson G, Halleen JM, et al. (2001) Estrogen
Kon A, Klement JF, et al. (1999) Nuclear factor-kappa B receptor alpha, but not estrogen receptor beta, is
mediates TNF-a inhibitory effect on a2(I) collagen involved in the regulation of the OPG/RANKL
(COL1A2) gene transcription in human dermal brob- (osteoprotegerin/receptor activator of NF-kappa B
lasts. J Immunol 162:422634. ligand) ratio and serum interleukin-6 in male mice. J
116. Kudo O, Fujikawa Y, Itonaga I, Sabokbar A, Torisu T, Endocrinol 171:42533.
Athanasou NA (2002) Proinammatory cytokine 130. Littlewood AJ, Russell J, Harvey GR, Hughes DE,
(TNFa /IL-1a) induction of human osteoclast forma- Russell RG, Gowen M (1991) The modulation of the
tion. J Pathol 198:2207. expression of IL-6 and its receptor in human osteo-
117. Kumar S, Votta BJ, Rieman DJ, Badger AM, Gowen M, blasts in vitro. Endocrinology 129:151320.
Lee JC (2001) IL-1- and TNF-induced bone resorption 131. Logan TF, Gooding W, Kirkwood JM, Shadduck RK
is mediated by p38 mitogen activated protein kinase. (1996) Tumor necrosis factor administration is associ-
J Cell Physiol 187:294303. ated with increased endogenous production of M-CSF
118. Kuno H, Kurian SM, Hendy GN, White J, deLuca HF, and G-CSF but not GM-CSF in human cancer patients.
Evans CO, et al. (1994) Inhibition of 1,25-dihydroxyvi- Exp Hematol 24:4953.
tamin D3 stimulated osteocalcin gene transcription by 132. Lorenzo JA, Pilbeam CC, Kalinowski JF, Hibbs MS
tumor necrosis factor-a: structural determinants (1992) Production of both 92- and 72-kDa gelatinases
within the vitamin D response element. Endocrinology by bone cells. Matrix 12:28290.
134:252431. 133. Lu X, Fiester, P., Rubin, J., Nanes M.S. (2003) NfkB sup-
119. Kuroki T, Shingu M, Koshihara Y, Nobunaga M (1994) pression of VDR transactivaation requires the Rel
Effects of cytokines on alkaline phosphatase and Homology Domain of p65. J Bone Mineral Res 18:F459.
osteocalcin production, calcication and calcium 134. Luchin A, Suchting S, Merson T, Rosol TJ, Hume DA,
release by human osteoblastic cells. Br J Rheumatol Cassady AI, et al. (2001) Genetic and physical interac-
33:22430. tions between Microphthalmia transcription factor
120. Kurokouchi K, Kambe F,Yasukawa K, Izumi R, Ishiguro and PU.1 are necessary for osteoclast gene expression
N, Iwata H, et al. (1998) TNF-a increases expression and differentiation. J Biol Chem 276:3670310.
of IL-6 and ICAM-1 genes through activation of Nf-kB 135. Manji SS, Zhou H, Findlay DM, Martin TJ, Ng KW
in osteoblast-like ROS17/2.8 cells. J Bone Miner Res (1995) Tumor necrosis factor-a facilitates nuclear
13:12909. actions of retinoic acid to regulate expression of the
121. Kusano K, Miyaura C, Inada M, Tamura T, Ito A, Nagase alkaline phosphatase gene in preosteoblasts. J Biol
H, et al. (1998) Regulation of matrix metallopro- Chem 270:895862.
teinases (MMP-2, -3, -9, and -13) by interleukin-1 and 136. Mann GN, Jacobs TW, Buchinsky FJ, Armstrong EC,
interleukin-6 in mouse calvaria: association of MMP Li M, Ke HZ, et al. (1994) Interferon-gamma causes
induction with bone resorption. Endocrinology 139: loss of bone volume in vivo and fails to ameliorate
133845. cyclosporin A-induced osteopenia. Endocrinology
122. Lacey DL, Grosso LE, Moser SA, Erdmann J, Tan HL, 135:10771083.
Pacici R, et al. (1993) IL-1-induced murine osteoblast 137. Mann J, Oakley F, Johnson PW, Mann DA (2002) CD40
IL-6 production is mediated by the type 1 IL-1 recep- induces interleukin-6 gene transcription in dendritic
tor and is increased by 1,25 dihydroxyvitamin D3. J Clin cells: regulation by TRAF2, AP-1, NF-kappa B, AND
Invest 91:173142. CBF1. J Biol Chem 277:1712538.
123. Lam J, Takeshita S, Barker JE, Kanagawa O, Ross FP, 138. Manolagas SC (1995) Role of cytokines in bone resorp-
Teitelbaum SL (2000) TNF-a induces osteoclastogene- tion. Bone 17:63S67S.
sis by direct stimulation of macrophages exposed to 139. Marusic A, Kalinowski JF, Harrison JR, Centrella M,
permissive levels of RANK ligand. J Clin Invest 106: Raisz LG, Lorenzo JA (1991) Effects of transforming
14818. growth factor-beta and IL-1a on prostaglandin syn-
124. Lee SE, Chung WJ, Kwak HB, Chung CH, Kwack KB, Lee thesis in serum-deprived osteoblastic cells. J Immunol
ZH, et al. (2001) Tumor necrosis factor-a supports the 146:26338.
survival of osteoclasts through the activation of Akt 140. Mayur N, Lewis S, Catherwood BD, Nanes MS (1993)
and ERK. J Biol Chem 276:493439. Tumor necrosis factor-a decreases 1,25-dihydroxyvit-
125. Lee SK, Kalinowski JF, Jastrzebski SL, Puddington L, amin D3 receptors in osteoblastic ROS 17/2.8 cells. J
Lorenzo JA (2003) Interleukin-7 is a direct inhibitor of Bone Miner Res 8:9971003.
Inflammatory Cytokines 87

141. McCarthy TL, Ji C, Chen Y, Kim KK, Imagawa M, Ito Y, 156. Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM,
et al. (2000) Runt domain factor (Runx)-dependent Behringer RR, et al. (2002) The novel zinc nger-
effects on CCAAT/ enhancer-binding protein delta containing transcription factor osterix is required for
expression and activity in osteoblasts. J Biol Chem 275: osteoblast differentiation and bone formation. Cell
2174653. 108:1729.
142. McSheehy PM, Chambers TJ (1986) Osteoblastic cells 157. Nanes MS, Kuno H, Demay MB, Kurian M, Hendy GN,
mediate osteoclastic responsiveness to parathyroid DeLuca HF, et al. (1994) A single up-stream element
hormone. Endocrinology 118:8248. confers responsiveness to 1,25-dihydroxyvitamin D3
143. McSheehy PM, Chambers TJ (1986) Osteoblast-like and tumor necrosis factor-a in the rat osteocalcin
cells in the presence of parathyroid hormone release gene. Endocrinology 134:111320.
soluble factor that stimulates osteoclastic bone resorp- 158. Nanes MS, McKoy WM, Marx SJ (1989) Inhibitory
tion. Endocrinology 119:16549. effects of tumor necrosis factor-a and interferon-
144. McSheehy PM, Chambers TJ (1987) 1,25-Dihydroxyvi- gamma on deoxyribonucleic acid and collagen syn-
tamin D3 stimulates rat osteoblastic cells to release a thesis by rat osteosarcoma cells (ROS 17/2.8).
soluble factor that increases osteoclastic bone resorp- Endocrinology 124:33945.
tion. J Clin Invest 80:4259. 159. Nanes MS, Rubin J, Titus L, Hendy GN, Catherwood B
145. Meikle MC, Bord S, Hembry RM, Reynolds JJ (1995) (1991) Tumor necrosis factor-a inhibits 1,25-
The synthesis of collagenase, gelatinase-A (72 kDa) dihydroxyvitamin D3-stimulated bone Gla protein
and -B (95 kDa), and TIMP-1 and -2 by human synthesis in rat osteosarcoma cells (ROS 17/2.8) by a
osteoblasts from normal and arthritic bone. Bone 17: pretranslational mechanism. Endocrinology 128:2577
25560. 82.
146. Min YK, Rao Y, Okada Y, Raisz LG, Pilbeam CC (1998) 160. Need AG, Morris HA, Horowitz M, Scopacasa E, Nordin
Regulation of prostaglandin G/H synthase-2 expres- BE (1998) Intestinal calcium absorption in men with
sion by interleukin-1 in human osteoblast-like cells. spinal osteoporosis. Clin Endocrinol (Oxf) 48:1638.
J Bone Miner Res 13:106675. 161. Nissen RM, Yamamoto KR (2000) The glucocorticoid
147. Mori K, Hatamochi A, Ueki H, Olsen A, Jimenez SA receptor inhibits NFkappaB by interfering with serine-
(1996) The transcription of human a1(I) procollagen 2 phosphorylation of the RNA polymerase II carboxy-
gene (COL1A1) is suppressed by tumour necrosis terminal domain. Genes Dev 14:231429.
factor-a through proximal short promoter elements: 162. Nomura S, Sakuma T, Higashibata Y, Oboki K, Sato M
evidence for suppression mechanisms mediated by (2001) Molecular cause of the severe functional de-
two nuclear-factorbinding sites. Biochem J 319 (Pt 3): ciency in osteoclasts by an arginine deletion in the
8116. basic domain of Mi transcription factor. J Bone Miner
148. Muegge K, Durum S (1990) Cytokines and Transcrip- Metab 19:1837.
tion Factors. [Review]. Cytokine 2:18. 163. Okada Y, Morimoto I, Ura K, Watanabe K, Eto S,
149. Muhlethaler-Mottet A, Di Berardino W, Otten LA, Mach Kumegawa M, et al. (2002) Cell-to-Cell adhesion via
B (1998) Activation of the MHC class II transactivator intercellular adhesion molecule-1 and leukocyte func-
CIITA by interferon-gamma requires cooperative tion-associated antigen-1 pathway is involved in
interaction between Stat1 and USF-1. Immunity 8: 1a,25(OH)2D3, PTH and IL-1a -induced osteoclast dif-
15766. ferentiation and bone resorption. Endocr J 49:48395.
150. Muhlethaler-Mottet A, Otten LA, Steimle V, Mach B 164. Okahashi N, Koide M, Jimi E, Suda T, Nishihara T (1998)
(1997) Expression of MHC class II molecules in differ- Caspases (interleukin-1beta-converting enzyme family
ent cellular and functional compartments is controlled proteases) are involved in the regulation of the survival
by differential usage of multiple promoters of the of osteoclasts. Bone 23:3341.
transactivator CIITA. Embo J 16:285160. 165. OKeefe GM, Nguyen VT, Ping Tang LL, Benveniste EN
151. Murphy KM, Ouyang W, Farrar JD, Yang J, Ranganath (2001) IFN-gamma regulation of class II transactivator
S, Asnagli H, et al. (2000) Signaling and transcription promoter IV in macrophages and microglia: involve-
in T helper development. Annu Rev Immunol 18: ment of the suppressors of cytokine signaling-1
45194. protein. J Immunol 166:22609.
152. Nagarajan UM, Bushey A, Boss JM (2002) Modulation 166. Pacici R (1996) Estrogen, cytokines, and pathogene-
of gene expression by the MHC class II transactivator. sis of postmenopausal osteoporosis. J Bone Miner Res
J Immunol 169:507888. 11:104351.
153. Nagarajan UM, Lochamy J, Chen X, Beresford GW, 167. Pacici R, Brown C, Puscheck E, Friedrich E,
Nilsen R, Jensen PE, et al. (2002) Class II transactiva- Slatopolsky E, Maggio D, et al. (1991) Effect of surgical
tor is required for maximal expression of HLA-DOB in menopause and estrogen replacement on cytokine
B cells. J Immunol 168:17806. release from human blood mononuclear cells. Proc
154. Nagy Z, Radeff J, Stern PH (2001) Stimulation of inter- Natl Acad Sci USA 88:51348.
leukin-6 promoter by parathyroid hormone, tumor 168. Pai RK, Askew D, Boom WH, Harding CV (2002) Reg-
necrosis factor -a, and interleukin-1beta in UMR-106 ulation of class II MHC expression in APCs: roles of
osteoblastic cells is inhibited by protein kinase C types I, III, and IV class II transactivator. J Immunol
antagonists. J Bone Miner Res 16:12207. 169:132633.
155. Nakase T, Takaoka K, Masuhara K, Shimizu K, 169. Panagakos FS, Kumar S (1994) Modulation of proteases
Yoshikawa H, Ochi T (1997) Interleukin-1 beta and their inhibitors in immortal human osteoblast-like
enhances and tumor necrosis factor-a inhibits bone cells by tumor necrosis factor-a in vitro. Inammation
morphogenetic protein-2-induced alkaline phos- 18:24365.
phatase activity in MC3T3-E1 osteoblastic cells. Bone 170. Panagakos FS, Kumar S (1995) Differentiation of
21:1721. human osteoblastic cells in culture: modulation of pro-
88 Bone Resorption

teases by extracellular matrix and tumor necrosis receptor in bone marrow cultures. J Exp Med 183:
factor-a. Inammation 19:42343. 258191.
171. Pascher E, Perniok A, Becker A, Feldkamp J (1999) 185. Rubin J, Fan D, Wade A, Murphy TC, Gewant H, Nanes
Effect of 1a,25(OH)2-vitamin D3 on TNF -a -mediated MS, et al. (2000) Transcriptional regulation of the
apoptosis of human primary osteoblast-like cells in expression of macrophage colony stimulating factor.
vitro. Horm Metab Res 31:6536. Mol Cell Endocrinol 160:193202.
172. Passeri G, Girasole G, Manolagas SC, Jilka RL (1994) 186. Salamone LM, Whiteside T, Friberg D, Epstein RS,
Endogenous production of tumor necrosis factor by Kuller LH, Cauley JA (1998) Cytokine production and
primary cultures of murine calvarial cells: inuence on bone mineral density at the lumbar spine and femoral
IL-6 production and osteoclast development. Bone neck in premenopausal women. Calcif Tissue Int 63:
Miner 24:10926. 46670.
173. Pavalko FM, Gerard RL, Ponik SM, Gallagher PJ, Jin Y, 187. Salem ML, Hossain MS, Nomoto K (2000) Mediation of
Norvell SM (2003) Fluid shear stress inhibits TNF-a the immunomodulatory effect of beta-estradiol on
induced apoptosis in osteoblasts: a role for uid shear inammatory responses by inhibition of recruitment
stress-induced activation of PI3-kinase and inhibition and activation of inammatory cells and their gene
of caspase-3. J Cell Physiol 194:194205. expression of TNF-a and IFN-gamma. Int Arch Allergy
174. Pruzanski W, Stefanski E,Vadas P, Kennedy BP, van den Immunol 121:23545.
Bosch H (1998) Regulation of the cellular expression 188. Sato K, Satoh T, Shizume K, Yamakawa Y, Ono Y,
of secretory and cytosolic phospholipases A2, and Demura H, et al. (1992) Prolonged decrease of serum
cyclooxygenase-2 by peptide growth factors. Biochim calcium concentration by murine gamma-interferon in
Biophys Acta 1403:4756. hypercalcemic, human tumor (EC-GI)-bearing nude
175. Qian Y, Commane M, Ninomiya-Tsuji J, Matsumoto K, mice. Cancer Res 52:4449.
Li X (2001) IRAK-mediated translocation of TRAF6 189. Sato M, Morii E, Komori T, Kawahata H, Sugimoto M,
and TAB2 in the interleukin-1-induced activation of Terai K, et al. (1998) Transcriptional regulation of
NFkappa B. J Biol Chem 276:416617. osteopontin gene in vivo by PEBP2aA/CBFA1 and
176. Ralston SH (1994) Analysis of gene expression in ETS1 in the skeletal tissues. Oncogene 17:151725.
human bone biopsies by polymerase chain reaction: 190. Scharla SH, Strong DD, Mohan S, Chevalley T, Linkhart
evidence for enhanced cytokine expression in post- TA (1994) Effect of tumor necrosis factor-a on the
menopausal osteoporosis. J Bone Miner Res 9:88390. expression of insulin-like growth factor I and insulin-
177. Ralston SH, Russell RG, Gowen M (1990) Estrogen like growth factor binding protein 4 in mouse osteo-
inhibits release of tumor necrosis factor from periph- blasts. Eur J Endocrinol 131:293301.
eral blood mononuclear cells in postmenopausal 191. Scheinman RI, Gualberto A, Jewell CM, Cidlowski JA,
women. J Bone Miner Res 5:9838. Baldwin AS, Jr. (1995) Characterization of mechanisms
178. Ramalho AC, Jullienne A, Couttet P, Graulet AM, involved in transrepression of NF-kappa B by activated
Morieux C, de Vernejoul MC, et al. (2001) Effect of glucocorticoid receptors. Mol Cell Biol 15:94353.
oestradiol on cytokine production in immortalized 192. Schneider HG, Allan EH, Moseley JM, Martin TJ,
human marrow stromal cell lines. Cytokine 16:12630. Findlay DM (1991) Specic down-regulation of
179. Rao VH, Singh RK, Delimont DC, Finnell RH, Bridge parathyroid hormone (PTH) receptors and responses
JA, Neff JR, et al. (1999) Transcriptional regulation of to PTH by tumour necrosis factor -a and retinoic acid
MMP-9 expression in stromal cells of human giant cell in UMR 10606 osteoblast-like osteosarcoma cells.
tumor of bone by tumor necrosis factor-a. Int J Oncol Biochem J 280 ( Pt 2):4517.
14:291300. 193. Shen V, Kohler G, Jeffrey JJ, Peck WA (1988) Bone-
180. Rickard D, Russell G, Gowen M (1992) Oestradiol resorbing agents promote and interferon-gamma
inhibits the release of tumour necrosis factor but not inhibits bone cell collagenase production. J Bone
interleukin 6 from adult human osteoblasts in vitro. Miner Res 3:65766.
Osteoporos Int 2:94102. 194. Shevde NK, Bendixen AC, Dienger KM, Pike JW (2000)
181. Rifas L, Halstead LR, Peck WA, Avioli LV, Welgus HG Estrogens suppress RANK ligand-induced osteoclast
(1989) Human osteoblasts in vitro secrete tissue differentiation via a stromal cell independent mecha-
inhibitor of metalloproteinases and gelatinase but not nism involving c-Jun repression. Proc Natl Acad Sci
interstitial collagenase as major cellular products. J USA 97:782934.
Clin Invest 84:68694. 195. Smith DD, Gowen M, Mundy GR (1987) Effects of
182. Rifas L, Kenney JS, Marcelli M, Pacici R, Cheng SL, interferon-gamma and other cytokines on collagen
Dawson LL, et al. (1995) Production of interleukin-6 in synthesis in fetal rat bone cultures. Endocrinology
human osteoblasts and human bone marrow stromal 120:24949.
cells: evidence that induction by interleukin-1 and 196. Srivastava S, Toraldo G, Weitzmann MN, Cenci S, Ross
tumor necrosis factor-a is not regulated by ovarian FP, Pacici R (2001) Estrogen decreases osteoclast
steroids. Endocrinology 136:405667. formation by down-regulating receptor activator of
183. Roggia C, Gao Y, Cenci S, Weitzmann MN, Toraldo G, NF-kappa B ligand (RANKL)-induced JNK activation.
Isaia G, et al. (2001) Up-regulation of TNF-producing J Biol Chem 276:883640.
T cells in the bone marrow: a key mechanism by which 197. Srivastava S, Weitzmann MN, Cenci S, Ross FP, Adler S,
estrogen deciency induces bone loss in vivo. Proc Natl Pacici R (1999) Estrogen decreases TNF gene expres-
Acad Sci USA 98:139605. sion by blocking JNK activity and the resulting pro-
184. Romas E, Udagawa N, Zhou H, Tamura T, Saito M, Taga duction of c-Jun and JunD. J Clin Invest 104:50313.
T, et al. (1996) The role of gp130-mediated signals in 198. Stashenko P, Obernesser MS, Dewhirst FE (1989) Effect
osteoclast development: regulation of interleukin 11 of immune cytokines on bone. Immunol Invest 18:
production by osteoblasts and distribution of its 23949.
Inflammatory Cytokines 89

199. Suda T, Kobayashi K, Jimi E, Udagawa N, Takahashi N regulated kinase 1/2, Jun N-terminal kinase, and c-Jun
(2001) The molecular basis of osteoclast differentia- are involved in NF-kappa B-dependent IL-6 expression
tion and activation. Novartis Found Symp 232:23547; in human monocytes. J Immunol 162:4893902.
discussion 24750. 215. Uchida M, Shima M, Shimoaka T, Fujieda A, Obara K,
200. Taichman RS, Hauschka PV (1992) Effects of inter- Suzuki H, et al. (2000) Regulation of matrix metallo-
leukin-1 beta and tumor necrosis factor-a on osteo- proteinases (MMPs) and tissue inhibitors of metallo-
blastic expression of osteocalcin and mineralized proteinases (TIMPs) by bone resorptive factors in
extracellular matrix in vitro. Inammation 16:587601. osteoblastic cells. J Cell Physiol 185:20714.
201. Takaesu G, Ninomiya-Tsuji J, Kishida S, Li X, Stark GR, 216. Uy HL, Mundy GR, Boyce BF, Story BM, Dunstan CR,
Matsumoto K (2001) Interleukin-1 (IL-1) receptor- Yin JJ, et al. (1997) Tumor necrosis factor enhances
associated kinase leads to activation of TAK1 by induc- parathyroid hormone-related protein-induced hyper-
ing TAB2 translocation in the IL-1 signaling pathway. calcemia and bone resorption without inhibiting bone
Mol Cell Biol 21:247584. formation in vivo. Cancer Res 57:31949.
202. Takaoka Y, Niwa S, Nagai H (1999) Interleukin-1beta 217. van der Saag PT, Caldenhoven E, van de Stolpe A (1996)
induces interleukin-6 production through the produc- Molecular mechanisms of steroid action: a novel type
tion of prostaglandin E(2) in human osteoblasts, MG- of cross-talk between glucocorticoids and NF-kappa B
63 cells. J Biochem (Tokyo) 126:5538. transcription factors. Eur Respir J Suppl 22:146s153s.
203. Takayanagi H, Ogasawara K, Hida S, Chiba T, Murata S, 218. Vargas SJ, Naprta A, Glaccum M, Lee SK, Kalinowski J,
Sato K, et al. (2000) T-cell-mediated regulation of Lorenzo JA (1996) Interleukin-6 expression and histo-
osteoclastogenesis by signalling cross-talk between morphometry of bones from mice decient in recep-
RANKL and IFN-gamma. Nature 408:6005. tors for interleukin-1 or tumor necrosis factor. J Bone
204. Tatakis DN, Schneeberger G, Dziak R (1988) Recombi- Miner Res 11:173644.
nant interleukin-1 stimulates prostaglandin E2 pro- 219. Vargas SJ, Naprta A, Lee SK, Kalinowski J, Kawaguchi
duction by osteoblastic cells: synergy with parathyroid H, Pilbeam CC, et al. (1996) Lack of evidence for an
hormone. Calcif Tissue Int 42:35862. increase in interleukin-6 expression in adult murine
205. Tatakis DN, Schneeberger G, Dziak R (1991) Recombi- bone, bone marrow, and marrow stromal cell cultures
nant interleukin-1 (IL-1) stimulates prostaglandin after ovariectomy. J Bone Miner Res 11:192634.
E2 production by osteoblastic cells: role of calcium, 220. Verthelyi D, Klinman DM (2000) Sex hormone levels
calmodulin, and cAMP. Lymphokine Cytokine Res correlate with the activity of cytokine-secreting cells in
10:959. vivo. Immunology 100:38490.
206. Thomas JA,Allen JL, Tsen M, Dubnicoff T, Danao J, Liao 221. Vidal ON, Sjogren K, Eriksson BI, Ljunggren O,
XC, et al. (1999) Impaired cytokine signaling in mice Ohlsson C (1998) Osteoprotegerin mRNA is increased
lacking the IL-1 receptor-associated kinase. J Immunol by interleukin-1a in the human osteosarcoma cell line
163:97884. MG-63 and in human osteoblast-like cells. Biochem
207. Thomson BM, Mundy GR, Chambers TJ (1987) Tumor Biophys Res Commun 248:696700.
necrosis factors -a and b induce osteoblastic cells to 222. Vig E, Green M, Liu Y, Donner DB, Mukaida N,
stimulate osteoclastic bone resorption. J Immunol 138: Goebl MG, et al. (1999) Modulation of tumor necrosis
7759. factor and interleukin-1-dependent Nf-kB activity by
208. Thomson BM, Saklatvala J, Chambers TJ (1986) mPLK/IRAK. J Biol Chem 274:1307784.
Osteoblasts mediate interleukin 1 stimulation of bone 223. Wadleigh DJ, Herschman HR (1999) Transcriptional
resorption by rat osteoclasts. J Exp Med. 164:10412. regulation of the cyclooxygenase-2 gene by diverse
209. Tohkin M, Kakudo S, Kasai H, Arita H (1994) Compar- ligands in murine osteoblasts. Biochem Biophys Res
ative study of inhibitory effects by murine interferon Commun 264:86570.
gamma and a new bisphosphonate (alendronate) in 224. Wagner EF (2002) Functions of AP1 (Fos/Jun) in bone
hypercalcemic, nude mice bearing human tumor (LJC- development. Ann Rheum Dis 61 Suppl 2:11402.
1-JCK). Cancer Immunol Immunother 39:15560. 225. Waldburger JM, Suter T, Fontana A, Acha-Orbea H,
210. Tondravi MM, McKercher SR, Anderson K, Erdmann Reith W (2001) Selective abrogation of major histo-
JM, Quiroz M, Maki R, et al. (1997) Osteopetrosis in compatibility complex class II expression on extrahe-
mice lacking haematopoietic transcription factor PU.1. matopoietic cells in mice lacking promoter IV of the
Nature 386:814. class II transactivator gene. J Exp Med 194:393406.
211. Toraldo G, Roggia C, Qian WP, Pacici R, Weitzmann 226. Weitzmann MN, Cenci S, Rifas L, Brown C, Pacici R
MN (2003) IL-7 induces bone loss in vivo by induction (2000) Interleukin-7 stimulates osteoclast formation
of receptor activator of nuclear factor kappa B ligand by up-regulating the T-cell production of soluble
and tumor necrosis factor -a from T cells. Proc Natl osteoclastogenic cytokines. Blood 96:18738.
Acad Sci USA 100:125130. 227. Weitzmann MN, Roggia C, Toraldo G, Weitzmann L,
212. Tsuboi M, Kawakami A, Nakashima T, Matsuoka N, Pacici R (2002) Increased production of IL-7 uncou-
Urayama S, Kawabe Y, et al. (1999) Tumor necrosis ples bone formation from bone resorption during
factor-a and interleukin-1beta increase the Fas- estrogen deciency. J Clin Invest 110:164350.
mediated apoptosis of human osteoblasts. J Lab Clin 228. Wiktor-Jedrzejczak W, Urbanowska E, Aukerman SL,
Med 134:22231. Pollard JW, Stanley ER, Ralph P, et al. (1991) Correction
213. Tsutsumimoto T, Kawasaki S, Ebara S, Takaoka K by CSF-1 of defects in the osteopetrotic op/op mouse
(1999) TNF-a and IL-1beta suppress N-cadherin suggests local, developmental, and humoral require-
expression in MC3T3-E1 cells. J Bone Miner Res ments for this growth factor. Exp Hematol 19:1049
14:175160. 54.
214. Tuyt LM, Dokter WH, Birkenkamp K, Koopmans SB, 229. Wood RJ, Fleet JC, Cashman K, Bruns ME, Deluca HF
Lummen C, Kruijer W, et al. (1999) Extracellular- (1998) Intestinal calcium absorption in the aged rat:
90 Bone Resorption

evidence of intestinal resistance to 1,25(OH)2 vitamin 237. Yao GQ, Sun BH, Insogna KL, Weir EC (2000) Nuclear
D. Endocrinology 139:38438. factor-kappaB p50 is required for tumor necrosis
230. Xie JF, Stroumza J, Graves DT (1994) IL-1 down- factor-a -induced colony-stimulating factor-1 gene
regulates platelet-derived growth factor-a receptor expression in osteoblasts. Endocrinology 141:291422.
gene expression at the transcriptional level in human 238. Ye H, Arron JR, Lamothe B, Cirilli M, Kobayashi T,
osteoblastic cells. J Immunol 153:37883. Shevde NK, et al. (2002) Distinct molecular mechanism
231. Xing L, Bushnell TP, Carlson L, Tai Z, Tondravi M, for initiating TRAF6 signalling. Nature 418:4437.
Siebenlist U, et al. (2002) Nf-kB p50 and p52 expres- 239. Yoshida H, Hayashi S, Kunisada T, Ogawa M,
sion is not required for RANK-expressing osteoclast Nishikawa S, Okamura H, et al. (1990) The murine
progenitor formation but is essential for RANK- and mutation osteopetrosis is in the coding region of the
cytokine-mediated osteoclastogenesis. J Bone Miner macrophage colony stimulating factor gene. Nature
Res 17:120010. 345:4424.
232. Xing L, Carlson L, Story B, Tai Z, Keng P, Siebenlist U, 240. Yoshihara R, Shiozawa S, Imai Y, Fujita T (1990) Tumor
et al. (2003) Expression of either Nf-kB p50 or p52 in necrosis factor -a and interferon gamma inhibit pro-
osteoclast precursors is required for IL-1-induced liferation and alkaline phosphatase activity of human
bone resorption. J Bone Miner Res 18:2609. osteoblastic SaOS-2 cell line. Lymphokine Res 9:59
233. Xu J, Cissel DS, Varghese S, Whipkey DL, Blaha JD, 66.
Graeber GM, et al. (1997) Cytokine regulation of adult 241. Zhang YH, Heulsmann A, Tondravi MM, Mukherjee A,
human osteoblast-like cell prostaglandin biosynthesis. Abu-Amer Y (2001) Tumor necrosis factor-a (TNF)
J Cell Biochem 64:61831. stimulates RANKL-induced osteoclastogenesis via
234. Yagi K, Tsuji K, Nifuji A, Shinomiya K, Nakashima K, coupling of TNF type 1 receptor and RANK signaling
DeCrombrugghe B, et al. (2003) Bone morphogenetic pathways. J Biol Chem 276:5638.
protein-2 enhances osterix gene expression in chon- 242. Zheng SX, Vrindts Y, Lopez M, De Groote D, Zangerle
drocytes. J Cell Biochem 88:107783. PF, Collette J, et al. (1997) Increase in cytokine pro-
235. Yamin TT, Miller DK (1997) The interleukin-1 recep- duction (IL-1 beta, IL-6, TNF-a but not IFN-gamma,
tor-associated kinase is degraded by proteasomes fol- GM-CSF or LIF) by stimulated whole blood cells in
lowing its phosphorylation. J Biol Chem 272:215407. postmenopausal osteoporosis. Maturitas 26:6371.
236. Yang C PC, Phillips LS, Rubin J, Nanes MS (1998) Para- 243. Zou W, Hakim I, Tschoep K, Endres S, Bar-Shavit Z
doxical enhancement of PTH-stimulated IGF-1 gene (2001) Tumor necrosis factor-a mediates RANK
transcription by TNF-a despite inhibition of IGF-1 ligand stimulation of osteoclast differentiation by an
mRNA. Bone 23:F041. autocrine mechanism. J Cell Biochem 83:7083.
6.
Genetics and Mutations Affecting
Osteoclast Development and Function
Mark C. Horowitz, Melissa A. Kacena, and
Joseph A. Lorenzo

source of osteoclast precursors. It was possible


Introduction to culture these cells with stromal cell lines or
primary osteoblasts to induce the formation
To understand the manner in which genetic of multinucleated, TRAP+ bone-resorbing
background or DNA mutation affects osteoclast osteoclast-like cells [177]. These authors also
formation and function, it is necessary to demonstrated that cell contact between the
possess a basic understanding of the cell biology hematopoietic osteoclast precursor and the
of osteoclastogenesis. Osteoclasts are large, mesenchymal stromal/osteoblastic cell was
multinucleated, tartrate-resistant, acid phos- required for osteoclast formation. These exper-
phatase (TRAP)-positive cells that are respon- iments formed the basis for the seminal work
sible for bone resorption. They arise from that demonstrated the requirement for interac-
hematopoietic stem cells and, like other tion between the receptor activator of nuclear
hematopoietic cells, are thought to pass through factor (NF)-kB (RANK)-receptor and the acti-
a series of differentiation stages that culminate vator of NF-kB ligand (RANKL) for osteoclast
in large, multinucleated, bone-resorbing cells. precursors to differentiate terminally into func-
Osteoclasts are the only true bone-resorbing tional osteoclast-like cells, a phenomenon that
cells. However, the specic stages and the is aided by the presence of macrophage colony-
regulation of their pathway development have stimulating factor (M-CSF) [5, 97, 200]. RANK is
not been completely elucidated. Osteoclasts a member of the superfamily of TNF receptors;
arise from a bifurcation of the monocyte/ it is expressed on B and T cells, dendritic cells,
macrophage lineage and thus evolve from the osteoclast precursors, and mature osteoclasts
common myeloid progenitor. Using cells iso- [5]. RANKL is a type II transmembrane protein,
lated from human bone marrow, Kurihara puri- expressed on stromal cells, osteoblasts, and T
ed CD34+ (a cell-surface marker found on cells [97, 194, 200].
myeloid progenitor cells, CFU-GMs) and treated It has taken more than 30 years for bone biol-
them with granulocyte-macrophage colony- ogists to understand how osteoclasts develop,
stimulating factor and 1,25-(OH)2D3. This become active and function. Modern molecular
resulted in the formation of osteoclasts, a result biology has made it possible to manipulate
that suggests that the osteoclast precursor was specic genes. The identication of spontaneous
the granulocyte-macrophage progenitor [95, mutations and the ability to engineer overexpres-
150]. However, Hattersley and Lee reported that sion or deletion of specic genes have brought
a more primitive cell than the CFU-GM differ- valuable insights into how those genes affect the
entiated into osteoclasts [61, 101]. Udagawa organism. This genetic approach has been used
showed that spleen or bone marrow cells are a successfully in dening osteoclast biology.
91
92 Bone Resorption

macrophage lineage progenitors [6, 33]. PU.1-/-


In the Nucleus mice, in addition to having no B cells, fail to
Transcriptional Regulation develop osteoclasts and macrophages [176].
PU.1 regulates these progenitors by controlling
the expression of the IL-7 receptor and c-fms
The molecular dissection of the differentiation (the M-CSF receptor) [34, 33]. Low concentra-
pathway of hematopoietic cell lineages has been tions of PU.1 protein induce B cell differen-
greatly facilitated by the identication of tran- tiation, while high concentrations promote
scription factors required for successful matu- macrophage development and inhibit B cell pro-
ration of a cell.An example of this occurs during gression [35]. In keeping with this hierarchical
B lymphopoiesis. In this process, numerous expression of PU.1 RNA, osteoclasts express
transcription factors, as well as specic protein higher levels of PU.1 mRNA than BM
protein interactions that dene their activities, macrophage precursor cells [176]. PU.1 exerts
are required for the progression of B cell differ- additional control over osteoclast differentia-
entiation to occur (Figure 6.1). Osteoclasts likely tion through its interaction with the micro-
follow a similar, staged development. However, phthalmia transcription factor (MITF) [116].
the transcription factors which control this pro- This well-characterized locus has at least 16 dif-
gression have only been partially identied. ferent mutant alleles. Defects in the mi, or and
This is particularly true for the early stages of crc alleles of MITF produce osteopetrosis [129,
development. Loss of these specic factors pre- 167]. OC can be detected in mi/mi mice,
cludes cells from continued maturation, and however they fail to fuse and form multinucle-
causes cells at the latest stage of differentiation ated cells [54, 174]. These data indicate that
to accumulate prior to the arrest. PU.1 is one of defects in osteoclast formation in mi/mi mice
the rst in a series of transcription factors, occur later than in PU.1-decient mice. In addi-
which include E2A, Early B Cell Factor (EBF), tion to the osteoclast defect, mi/mi mice are
and Pax5, that function in a specic maturation- decient in B cell precursors [169]. Co-culture
dependent sequence that is required for B cell of BM cells from mi/mi mice with stromal cells
development. E2A appears to regulate tran- in the presence of IL-7 (the major growth and
scription of the EBF gene and in turn EBF has differentiation factor for B cells) resulted in B-
been implicated in activating Pax5 [87, 135]. cell differentiation. These results suggest that
PU.1, a member of the ETS domain transcrip- the defect in B-cell development in mi/mi mice
tion factors, is required to regulate the prolifer- was not in the precursors, but rather in the local
ation and differentiation of B cell and microenvironment.

Figure 6.1 B-cell differentiation from hematopoietic stem cells is a multistep process requiring a program of transcription factors.
Genetics and Mutations Affecting Osteoclast Development and Function 93

Two different, basic helix-loop-helix proteins mice are enriched in osteoclast precursors.
encoded by the E2A and EBF genes play a criti- These data identify Pax5 as a transcription
cal role at the start of B cell differentiation. factor that is required for the normal regulation
Absence of either of these proteins halts B cell of osteoclast development. In addition, Pax5-/-
development at the earliest stage, before the DH- mice represent a novel system in which to
JH rearrangement occurs [10, 108]. Although the study the relationship of osteoclasts and B cell
skeletons of these mutant mice have not been differentiation.
extensively analyzed, EBF-decient mice are Recently, it was shown that loss or overex-
runted (M. Horowitz, Hesslein, Lorenzo unpub- pression of certain transcription factors repro-
lished). This implies that their bones are grams the fate of progenitor/precursor cells
pathologic. [196]. As an example, enforced expression in B
Pax5 is a member of the multigene family that cells of C/EBPa and C/EBPb, a bZip family tran-
encodes the paired box transcription factors. scription factor, reprograms the cells into
The Pax5 gene encodes for the B cell lineage spe- macrophages [196]. Conversely, macrophages
cic activation factor (BSAP). Pax5 is expressed from mice decient in C/EBPb exhibit func-
exclusively in the B-lymphocyte lineage that tional defects [161]. At present it is unclear how
leads from B220+ pro-B cells to mature B cells. changes in the expression of these transcription
However, Pax5 is not present in terminally dif- factors relates specically to osteoclasts.
ferentiated plasma cells [179]. When the Pax5 However, it is not unreasonable to predict that
gene is deleted in mice, the most striking result, these alterations may affect osteoclast differen-
in addition to early postnatal lethality, is the loss tiation. These ndings are consistent with the
of all B cell development beyond the early pro- idea that there exist networks of transcription
B cell stage [134]. This developmental arrest of factors, which together function as both positive
the B cell lineage leaves normal numbers of and negative regulators of differentiation [139].
B220+ pro-B cells in the BM. Pro-B cells isolated It is now known that NF-kB is a critical medi-
from Pax5-/- BM can be grown in vitro on ator of RANK signaling in most responsive cells,
stromal/osteoblast feeder cells in the presence including osteoclasts and osteoclast precursors
of IL-7, a condition that maintains their undif- [5, 17, 195]. RANKL-RANK interaction signals
ferentiated phenotype [8]. With removal of IL-7 TRAF-6, which activates NF-kB. NF-kB is also
and stimulation with the appropriate cytokines, required for osteoclastic responses to IL-1 and
these cells can differentiate into multiple TNF [197]. NF-kB transcription factors are
hematopoietic lineages [134]. Similarly, in dimers composed of various combinations of
normal BM, an early osteoclast progenitor the structurally related proteins p50 (NFKB1),
(Mac-1+ c-fms-) can be induced to express a B- p52 (NFKB2), p65 (RelA), c-Rel (Rel), and RelB
cell-like phenotype (B220+) when cultured with [49, 75]. Unlike the single knockout of either p50
a stromal/osteoblast cell line and treated with or p52, which have no skeletal phenotype,
IL-7 [8]. Treatment of the Pax5-/- pro-B cells p50/p52 double knockout mice fail to generate
with M-CSF and RANKL induced the formation mature osteoclasts or B cells and have severe
of osteoclast-like cells; this raises the possibility osteopetrosis. These mice are runted and their
that Pax5 is involved in the regulation of osteo- incisor teeth fail to erupt, both of which are
clast development [134]. Because cloned pro-B- additional signs of an osteoclast defect. The
cell lines can be used, these ndings are the p50/p52 double-knockout mice also have
clearest indication that pro-B cells can be markedly impaired thymic and splenic architec-
induced to form osteoclasts in the absence of tures and macrophage activity. Co-culture
Pax5. These observations and the severe runting experiments show that the defect in these
of the homozygous mutant mice prompted us to animals is in osteoclast precursors rather than
examine Pax5-/- mice to determine whether in osteoclast induction. Hence, in this regard
they had a bone phenotype. Pax5-/- mice are they resemble RANK- or RANKL-decient
severely osteopenic, missing more than 60% of mice.
their bone mass [67]. This condition is the result c-fos is a proto-oncogene that is a component
of a 35-fold increase in the number of osteo- of the AP-1 transcription factor complex [7].
clasts in bone, even though the number of Overexpression of c-fos transforms chondrob-
osteoblasts in Pax5-/- mice is the same as in lasts and osteoblasts, but not osteoclasts/
control mice. In vitro, spleen cells from Pax5-/- hematopoietic cells [58, 155, 186, 187]. There-
94 Bone Resorption

fore, the fact that c-fos -/- mice develop sis. Hence, it appears that independent actions
osteopetrosis as a result of impaired osteoclast of both NFATc1 and c-fos are critical elements
differentiation is unexpected [59]. In addition to of normal osteoclast formation.
having osteopetrosis, c-fos -/- mice are smaller,
and have reduced body weight, presumably
because lack of tooth eruption causes dimin-
ished food intake. These mice have a shorter
At the Cell Surface and
snout and shorter limbs [188]. Interestingly, Beyond Cytokines,
these mice also have a marked reduction (75%)
in B220+ B cells in their spleen. This is another
Receptors and Signaling
example of a defect in osteoclast development
that is paralleled by defects in B-cell develop- M-CSF, also known as colony-stimulating
ment. Further investigation into the mechanism factor-1 (CSF-1), was originally found to be a
associated with the osteopetrotic phenotype of regulator of macrophage formation [153].
c-fos-/- mice has shown that there are no However, a spontaneous mouse mutant with a
TRAP+ cells in the long bones [59]. Osteoclast phenotype of absent osteoclasts and defective
progenitors, but no differentiated multinucle- macrophage-monocyte formation (the op/op
ated osteoclasts, were seen [59]. Interestingly, c- mouse) was discovered to have a point mutation
fos was found to cause a lineage shift between in the coding region of the M-CSF gene [46, 193,
osteoclasts and macrophages, which resulted 202]. This mutation introduced a stop codon
in an increase in the number of tissue and prevented normal expression of a func-
macrophages. These studies claried the role of tional M-CSF protein. Injection of M-CSF into
c-fos as a transcriptional regulator of osteoclast op/op mice corrected the defect in osteoclast
and macrophage differentiation [59]. formation and bone resorption [47], a result
Nuclear factor of activated T cells-c1 that conrmed the importance of this protein
(NFATc1) has recently been shown to be a crit- for osteoclast development. Recently, a mouse
ical signaling molecule in osteoclastogenic that lacked the receptor for M-CSF (c-fms a
responses [76, 170]. NFATs are a family of tran- receptor tyrosine kinase) was generated and
scription factors that shuttle between the cyto- found to have a phenotype, that is similar to that
plasm and the nucleus. Activation, which allows of the op/op mouse [31].
entry to the nucleus [27], occurs when calcium Stimulators of bone resorption increase pro-
activates the phosphatase calcineurin. This in duction of M-CSF in bone [45, 156, 191], with
turn, dephosphorylates cytoplasmic NFAT, and multiple transcripts of M-CSF produced by
exposes a nuclear localization sequence in the alternate splicing [29, 28]. The membrane-bound
molecule. This allows NFAT to be transported to or cell-surface form of M-CSF differs from the
the nucleus, where it binds DNA and activates soluble form because it lacks a cleavage site. Only
transcription. Using embryonic stem cells to forms with a cleavage site are subject to rapid
generate osteoclasts, Takayanagi et al. [170] proteolytic release of the ectodomain as a
demonstrated that ES cells from NFATc1- soluble protein. The membrane-bound form of
decient mice, which have an embryonic lethal M-CSF is regulated by stimulators of resorption
mutation, did not form osteoclasts when treated and facilitates the differentiation of osteoclasts
with M-CSF and RANKL. In contrast, M-CSF from precursor cells [156, 198].
and RANKL stimulated abundant osteoclast for- The effect of M-CSF on the function of
mation in cultures of wild-type ES cells. Retro- mature osteoclasts is controversial. Osteoclasts
viral overexpression of NFATc1 in bone marrow express c-fms [65] and initial reports concluded
cells induced osteoclastogenesis in the absence that M-CSF inhibited bone resorption in mature
of RANKL stimulation [170]. This nding con- osteoclasts [62]. Subsequent studies have found
rms the critical role of NFATc1 induction in that it increased cytoplasmic spreading, fusion
RANKL responses. It also appears that NFAT is of mononuclear osteoclast precursors into
a downstream target of c-fos in RANK-medi- osteoclasts, and led to osteoclastic resorption [3,
ated osteoclastogenesis [124]. Interestingly, in 102, 159]. M-CSF also induces proliferation of
the absence of RANKL, stimulation of c- osteoclast precursors. While these precursor
fos-decient osteoclast precursor cells were cells are proliferating in response to M-CSF, they
insensitive to NFATc1-induced osteoclastogene- do not differentiate into multinucleated bone-
Genetics and Mutations Affecting Osteoclast Development and Function 95

resorbing cells. This is one reason that osteo- cells [8]. Once activated, RANK initiates a series
clastic bone resorption is inhibited by M-CSF. of intracellular second messengers, including
The ability of M-CSF to stimulate cytoplasmic TNF-receptor associated factors (TRAF) family
spreading in osteoclasts appears to involve c-src members, NF-kB, mitogen-activated protein
because src kinase activity increased threefold kinases (MAPK), activator protein-1 (AP-1),
after M-CSF treatment. Moreover, osteoclastic NFATc1, and the src-AKT-phosphatidylinositol-
cytoplasmic spreading did not occur in c- 3-OH kinase (PI(3)K) system [17].
src-decient mice [72, 73]. M-CSF also appears Perhaps the most convincing evidence that
to down-regulate the expression of c-fms. RANKL and RANK are critical regulators of
The role of M-CSF in regulating osteoclast osteoclastogenesis comes from mice lacking
apoptosis has recently been examined. Addition these proteins. RANKL-decient mice have sig-
of M-CSF to mature osteoclast cultures prolongs nicant osteopetrosis, no osteoclasts, a normal
their survival [50, 80]. This response may be number of monocyte-macrophages, and extra-
important for the development of the medullary hematopoiesis [93]. They also have
osteopetrotic phenotype in op/op mice because signicant defects in B-cell lymphopoiesis, with
transgenic expression of bcl-2, which blocks markedly reduced numbers of pre-B-lympho-
apoptosis in myeloid cells, partially reversed cytes in their marrow. Remarkably, RANKL
the defects in osteoclast and macrophage knockout mice lack lymph nodes and their T
development [99]. cells secrete less cytokines. These ndings
The role of osteoprotegerin (OPG) in bone demonstrate that, in addition to its effects on
was rst established in studies with transgenic osteoclastogenesis, RANKL is an important
animals [164]. It was demonstrated that global regulator of lymphocyte development.
overexpression of the protein in mice produced RANK-decient mice also have no osteoclasts
animals with markedly decreased bone resorp- and develop osteopetrosis and extramedullary
tion, increased bone mass, and few osteoclasts. hematopoiesis [38, 103]. In addition, like
OPG is a member of the tumor necrosis factor RANKL-decient mice, they lack mature B-
receptor superfamily (TNFRSF11b). It is a decoy lymphocytes and have defects in lymph node
receptor for RANKL [98, 201]. OPG inhibits OC development. Overexpression of a soluble form
formation and bone resorption by binding of RANK in transgenic mice produced a
RANKL and preventing its interaction with phenotype that was similar to that of OPG-
RANK, its cognate receptor, on the surface of overexpressing transgenic mice [68] and was
osteoclast precursor cells. Mice lacking OPG characterized by osteopetrosis and a decreased
have severe osteoporosis, an increase in the number of osteoclasts in bone.
number of osteoclasts, and, importantly, exhibit Our understanding of the signaling pathways
arterial calcication [24, 127]. Studies with utilized by RANK in the activation of osteo-
OPG-decient mice have shown that the active clasts has been greatly enhanced by the devel-
resorption of bone by osteoclasts is an ongoing opment of knockout mouse models that lack
process, which must be overtly held in check to specic components of this signaling pathway.
maintain bone mass. One role of M-CSF in facil- TRAFs 2, 5, and 6 all bind RANK, but only
itating osteoclastogenesis is its ability to stimu- TRAF6 deciency has been associated with
late RANK expression on osteoclast precursor osteopetrosis in mice [111, 133]. TRAF proteins

Table 6.1. Transgenic or Spontaneous Mutations in Mice with Alterations in Osteoclast Number/Physiology
Cathepsin k Ion channels Prostaglandin receptors
c-fos IRAK Pu.1
Cyclooxygenase LIF receptor RANKL
DAP-12 M-CSF RANK
FcRg Micropthalmia transcription factors SHP-1
gp130 NFAT Src
IL-1 receptors NFkB TNF receptors
Intergins (avb3) OPG TRAF5 and 6
Interferons Pax 5
96 Bone Resorption

are believed to be involved in mediating with an almost fourfold increase in osteoclast


responses of TNF superfamily receptors like number, has been reported [104]. IRAKM
RANK. Two murine models of TRAF6 de- appears to down-regulate osteoclast differentia-
ciency have now been produced [111, 133]. Curi- tion and activation through NF-kB and MAPK
ously, in one mouse model, there was an almost signaling pathways. IL-1 receptor type II does
complete absence of osteoclasts in bone [133], not appear to have agonist activity. Rather, there
while in the other, osteoclasts were abundant in is convincing evidence that it is a decoy recep-
bone but appeared to function poorly and tor, which prevents activation of IL-1 type I
lacked contact with the bone surface [111]. Mice receptors [30]. IL-1 receptor type II can also
lacking TRAF6 also have impaired CD40 and synergize with IL-1ra to inhibit activation of the
lipopolysaccharide (LPS)-induced proliferation IL-1 receptor type I [25]. IL-1 may play a role in
in their B cells, impaired nitric oxide induction estrogen-mediated bone loss, because the IL-1
in macrophages and impaired interleukin-1 antagonist IL-1ra blocks ovariectomy-induced
(IL-1)-, CD40-, and LPS-induced activation of bone loss [140]. This has been conrmed by
NF-kB and JNK. In addition, in vitro osteoclas- studies demonstrating that mice lacking the IL-
togenesis from spleen cell precursors obtained 1 receptor type I (IL-1R1 -/-) did not lose bone
from these mice was impaired [111, 133]. mass following ovariectomy [113].
TRAF5-decient mice demonstrate normal Like IL-1, tumor necrosis factor (TNF) con-
bone mass but delayed osteoclastogenic sists of two related polypeptides (a and b) that
responses to RANKL and TNF in vitro and a are separate gene products [13, 138, 164]. TNF a
delayed hypercalcemic response to PTH in vivo and b have similar biologic activities, and both
[84]. This nding demonstrates that TRAF5, are potent stimulators of bone resorption [12,
while not critical for osteoclastogenic res- 36]. TNF is similar to IL-1 in its potency to
ponses, is part of the signaling pathway in osteo- induce bone resorption. Like IL-1, TNF also
clast precursor cells. enhances the formation of osteoclast-like cells
IL-1 is encoded by two separate gene prod- in bone marrow culture [143], and thus appears
ucts, a and b, which have identical activities to regulate osteoclast precursor cell differentia-
[36]. IL-1 was the rst polypeptide mediator of tion. TNFa is produced by human osteoblast-
immune cell function that was found to regulate like cell cultures [55] and its production can be
bone resorption [112]. It also increased stimulated by IL-1, GM-CSF, and LPS, but not by
prostaglandin synthesis in bone [112], an action PTH, 1,25-(OH)2 vitamin D, or calcitonin.
that may account for some of its resorptive Recently, TNF was shown to stimulate osteoclast
activity. IL-1 is the most potent known peptide formation directly in an in vitro culture system.
stimulator of in vitro bone resorption [112]; it This effect was independent of the actions of
also has potent in vivo effects [18]. RANK, because it occurred in cells from RANK-
Two known receptors for IL-1, type I and type decient mice [9, 90]. However, the functional
II, have been identied [37]. All known biologic signicance of this nding is questionable
responses to IL-1 appear to be mediated exclu- because in vivo administration of TNF to
sively through the type I receptor [165]. Signal- RANK-decient mice caused only an occasional
ing through type I receptors involves activation osteoclast to form [81].
of NF-kB and specic TRAFs [121, 41]. IL-1 Like IL-1, TNF binds to two cell surface recep-
receptor type I requires interaction with a tors, the TNF receptor 1 or p55 and the TNF
second protein, interleukin-1 receptor accessory receptor 2 or p75 [48]. In contrast to IL-1, both
protein (IRAcP), to generate post-receptor receptors transmit biologic responses. Mice
signals [69, 94, 192]. IRAcP induces activation decient in either or both TNF receptors have
of interleukin-1 receptor-associated kinase been engineered [142, 154]. All of these animals
(IRAK) [192]. Phosphorylated IRAK binds appear healthy and breed normally, but lack
TRAF6 with subsequent downstream signaling. selective immune responses. Bones from mice
IRAKM, unlike other members of the family, that lack either IL-1 receptor type I or TNF
lacks kinase activity and is a negative regulator receptor 1 are similar to those of wild-type
of IL-1R signaling. IRAKM-decient mice mice, except for a 36% increase in the total bone
develop lower total bone mineral density than area of calvaria from IL-1 receptor type I-
do wild-type controls; a 60% reduction in tra- decient mice [182]. Mice decient in TNF
becular bone, as measured by micro-CT, along receptors (TNF -/-) fail to lose bone mass after
Genetics and Mutations Affecting Osteoclast Development and Function 97

ovariectomy [149]. This nding appears signi- cic binding. Besides IL-6, the cytokines of this
cant because production of TNF by T lympho- family include interleukin 11 (IL-11), leukemia
cytes appears responsible for some of the bone inhibitory factor (LIF), oncostatin M (OSM),
loss that occurs in mice after ovariectomy [149]. ciliary neurotrophic factor (CNTF), and car-
IL-6, like IL-1 and TNF, modulates immune diotrophin-1 (CT-1). Except for CT-1, receptors
cell function in many ways. It also affects the for all other IL-6 family members have been
replication and differentiation of other cell detected in bone marrow stromal or osteoblas-
types [2, 63]. The receptor for IL-6 is composed tic cells [11]. In addition, IL-11-specic
of two parts: a specic IL-6-binding protein (IL- receptors are present in OC-like cells that are
6 receptor), which can be either membrane generated in vitro. Signal transduction through
bound or soluble, and gp-130, an activator these receptors utilizes the JAK/STAT pathway
protein that is common to a number of cytokine [88].
receptors [88]. Production of the specic IL-6 Leukemia inhibitory factor (LIF) is produced
receptor may be regulated, because bone cells by bone cells in response to a number of resorp-
do not always express it [171]. tion stimuli [57, 78, 122]. However, production
The ability of IL-6 to stimulate bone resorp- can be variable and depends on the model
tion in vitro is variable and depends on the system that is being studied. The effects of
assay system [1, 77, 109, 115]. The reasons for LIF on bone resorption also are variable. In a
this variability are not known. A major function number of in vitro model systems LIF stimu-
of IL-6 is to regulate the differentiation of osteo- lates resorption by a mechanism that is
clast progenitor cells into mature osteoclasts prostaglandin dependent [148], while in other
[118, 151]. In an in vitro model of osteoclast dif- in vitro assays, it has shown inhibitory effects
ferentiation from human bone marrow, IL-6 was [114, 180]. Up-regulation of RANKL production
found to be a potent stimulus of new osteoclast- seems to be the mechanism by which LIF stim-
like cell development by a mechanism that also ulates bone resorption in organ cultures, but the
was dependent on IL-1 [96]. In contrast, in a role of prostaglandins in this model remains
murine in vitro system, IL-6 was only effective unknown [141]. Mice that lack the specic LIF
when soluble IL-6 receptor was added to the cul- receptor and hence, cannot respond to LIF,
tures [171]. Implantation of Chinese hamster exhibit reduced bone volume and a sixfold
ovary (CHO) cells, which were genetically increase in osteoclast number [190].
engineered to express murine IL-6, into nude The role of IL-6 family members in osteoclast
mice produced a syndrome of hypercalcemia, formation recently has been reexamined in light
cachexia, leukocytosis, and thrombocytosis of ndings that demonstrated that, compared to
[14]. The effects of IL-6 on resorption in this wild-type animals, mice lacking the gp-130
model were weak, but were potentiated signi- activator protein have an increased number of
cantly when the animals also were treated with osteoclasts [85]. Because gp-130 is an activator
PTHrP [32]. A major effect of IL-6 in these in of signal transduction for all members of the IL-
vivo models was to increase the number of early 6 family, this result implies that one or more IL-
osteoclast precursors (CFU-GM) in the bone 6 family members inhibit osteoclast formation
marrow. IL-6 action may contribute to some of and bone resorption.
the increased bone resorption and bone pathol- Prostaglandins are potent stimulators of bone
ogy that are associated with the clinical syn- resorption [145]. Curiously, they directly inhibit
dromes of Pagets disease [152], hypercalcemia the resorbing activity of mature osteoclasts [26,
of malignancy [60], brous dysplasia [199], 51]. However, they stimulate RANKL and inhibit
giant cell tumors of bone [147], and OPG production by osteoblasts [20, 64, 184,
GorhamStout disease [55]. 201]. This leads to increases in the number
Ovariectomy-induced bone loss in mice has of osteoclasts and to a net increase in bone
been associated with increased IL-6 production resorption.
[79] and mice decient in IL-6 failed to lose Prostaglandins synergize with RANK-L and
bone after ovariectomy [44]. M-CSF to stimulate both the formation of osteo-
IL-6 is a member of a group of cytokines that clasts from hematopoietic progenitors and the
share the gp-130 activator protein in their bone-resorbing activity of mature osteoclasts
receptor complex [119]. Each family member [189]. In mice that lack PGHS2, the inducible
utilizes unique ligand receptors to generate spe- enzyme, which produces prostaglandins from
98 Bone Resorption

arachidonic acid, there was a 6070% decrease defect in osteoclast differentiation as opposed
in the ability of PTH or 1,25-(OH)2D3 to stimu- to a defect in activity. In vitro, few osteoclasts
late an increase in osteoclast-like cell formation formed when precursors were stimulated
in bone marrow cultures [136]. Hence, with RANKL and M-CSF. Further, unlike in
prostaglandins appear to be involved in the DAP12 -/- mice, co-culturing precursors with
resorptive responses of a number of agents. osteoblasts did not signicantly elevate the
Prostaglandin receptors are present on bone number of osteoclasts formed in vitro [92, 128].
cells and of these the EP2 and EP4 receptors, Two signaling pathways have been implicated
which stimulate cyclic AMP, appear to mediate as responsible for the severe inhibition of osteo-
resorptive responses [168]. Interestingly, the clastogenesis in these mice. Mocsai et al. [128]
enhancement of RANKL-mediated effects on demonstrated that during osteoclastogenesis,
osteoclast numbers exclusively involves interac- ITAM-signaling was dependent on Syk kinase.
tions with EP2, because these effects did not Further, Cbl and Pyk2 (signals downstream of
occur in cells from EP2-decient mice [105]. Syk) previously had been implicated in osteo-
Prostaglandins are anabolic when given inter- clast formation and/or function [43, 100, 158,
mittently in vivo [86]. Signicantly, the in vivo 172]. In other studies, Koga et al. [92] conrmed
bone anabolic effects of prostaglandins appear that NFATc1 is a downstream target of FcRg-
to be mediated by the EP4 receptor, because and DAP12- ITAM-mediated signaling and that
these responses are absent in EP4-decient mice the induction of NFATc1 is required for RANK-
[203]. induced osteoclast differentiation. These
Similar to other osteopetrotic mice such as ndings suggest that other ITAM-containing
the op/op mice [46, 91, 193], DAP12 -/- mice signaling adapters, or their downstream targets,
and DAP12 -/-FcRg -/- double-knockout also can play a role in regulating osteoclastoge-
mice had osteopetrosis or severe osteopetrosis, nesis (Figure 6.2).
respectively, as a result of impaired osteoclast As described earlier, deciencies in osteoclast
differentiation [70, 83, 92, 128]. DAP12 and FcRg differentiation are responsible for the osteopet-
(Fc receptor g-chain) are both transmembrane rosis seen in the op/op mice [46, 91, 193], PU.1
proteins that contain an immunoreceptor tyro- [14], NFkB [49, 197], and c-fos [59, 188] null
sine-based activation motif (ITAM). mice. However, like the cathepsin K decient
DAP12 -/- mice exhibited mild osteopetrosis mice [56, 157], c-src -/- mice have osteopetro-
as evidenced by micro-CT and histomorphome- sis that has resulted from impaired osteoclast
tric analysis, but were normal in terms of function [4, 19, 66, 74, 166, 175]. The c-src proto-
growth and fertility [70, 83, 92, 128]. In vivo oncogene is a tyrosine kinase [23] that is highly
osteoclast number was normal as was osteoclast expressed in osteoclasts and is used as a marker
morphology; however, in vitro the osteoclast for the osteoclast phenotype [66]. Initially, c-src
number was signicantly reduced when bone -/- mice died within the rst few weeks after
marrow cells were cultured in the presence of birth because of the failure of their incisors to
RANKL and M-CSF. The osteoclast morphology erupt. Feeding mice ground-up chow and baby
also was different, and the ability to resorb bone formula allowed them to survive and made
was severely reduced [70, 83, 92, 128]. Osteoclast possible long-term follow-up [19]. Osteopetro-
formation can be rescued with retroviral sis (up to a fourfold increase in unremodeled
expression of DAP12. In addition, when bone osteocartilaginous matrix in the vertebrae)
marrow cells were co-cultured with osteoblasts, resulted from the failure of osteoclasts to form
osteoclastogenesis was partially restored. This rufed borders [19, 175]. Similarly, studies show
suggests that osteoblasts can compensate, at that c-src -/- osteoclasts did not exhibit a well-
least in part, for the loss of DAP12-mediated dened attachment ring [74]. Other studies
signaling. demonstrated that pp60c-src (the gene product)
A shorter stature and a more rounded face was expressed in osteoclasts and even though
characterized the more severe osteopetrosis other src family members (c-fyn, c-yes, and c-
seen in DAP12 -/-FcRg -/- mice. Interestingly, lyn) were similarly expressed in osteoclasts, this
unlike what occurs is src -/- or RANKL -/- did not compensate for the absence of pp60c-src
mice, teeth erupted normally in DAP12 -/- FcRg [66].
-/- mice [93, 128, 166]. The osteoclast number In other studies, investigators developed c-
was signicantly reduced in vivo, indicating a src -/- mice that expressed either wild-type or
Genetics and Mutations Affecting Osteoclast Development and Function 99

Figure 6.2 Immunoreceptor tyrosine-based activation motifs (ITAMs) are part of the signaling pathway leading to osteoclast
formation.

mutant versions of src. Expression of the wild- formation, which likely also contribute to the
type transgene rescued the osteopetrotic phe- osteopetrotic phenotype [177]. In addition to
notype; osteoclasts from these mice exhibited the in vivo increase in osteoblast number
rufed borders and produced normal bone his- described in c-src -/- mice, in vitro results have
tology. Expression of a src allele that reduced shown that incubation of osteoblasts with src-
activation of the kinase several fold also fully antisense oligodeoxynucleotides (~60% reduc-
restored the normal bone phenotype in src-/- tion in src levels) reduced cell proliferation
mice. However, expression of an src allele that [123]. Overall, these data suggest that c-src plays
severely impaired kinase activity only partially a dual role, regulating both bone formation and
restored the bone phenotype. Osteoclast rufed resorption.
borders were formed in these mice, but fewer Mice expressing the autosomal recessive
osteoclasts displayed them. These results motheaten (me/me) mutation are decient in
suggest that in vivo c-src has important kinase- SHP-1 protein tyrosine phosphatase [163].
independent functions [160]. In mice that lack These mice have markedly reduced bone
c-src, src inhibitors inhibit bone resorption in mineral density. Their trabecular and cortical
vitro and in vivo [126]. On the other hand, c-src thicknesses are decreased as a result of a 14%
-/- mice also display enhanced indices of bone increase in osteoclast number [178]. Co-culture
100 Bone Resorption

of normal osteoblasts with me/me BM cells (as osteoclast function. Saftig et al. [157] reported a
a source of osteoclast precursors) led to an signicant, sixfold reduction in the number of
increase in osteoclasts as compared to controls. resorption pits formed by cathepsin K -/-
Notably, B cells from me/me mice are hyperre- osteoclasts in vitro. The perimeter of these pits
sponsive and polyclonaly activated. Whether was signicantly greater than in controls, but
these B cells participate in the increased osteo- the volume and depth were signicantly
clast formation in these animals remains to be reduced. Gowen et al. [56] reported visualizing
shown. distinct zones of demineralization (osteoclast-
To resorb bone, osteoclasts must solubilize bone interface) that were not apparent in
both the inorganic mineral and the organic control mice. This suggests that demineraliza-
proteins of bone [15]. Inorganic mineral is tion of the bone was normal, but that removal
efciently removed with acid secretion, and of the organic bone matrix was impaired. Con-
lysosomal proteinases degrade the bone matrix versely, Okaji et al. [137] visualized indistinct
proteins [131]. One of the lysosomal pro- zones of demineralization at the osteoclast-
teinases, cathepsin K, efciently degrades bone interface on alveolar bone surfaces, and on
several bone matrix proteins including type I that basis argued that alveolar bone resorption
collagen, type II collagen, osteopontin, and of both inorganic and organic bone was
osteonectin [16, 22, 52, 82]. unaltered. The latter authors also reported that
Cathepsin K is a cysteine proteinase that is matrix metalloproteinase-9 levels were signi-
highly and predominantly expressed in cells of cantly elevated in the absence of cathepsin K,
the osteoclast lineage [21, 39, 106, 110, 162, 173]. and thus may act as a compensatory proteinase.
Cathepsin B, cathepsin L, and cathepsin O also Further, osteoclast morphology was signi-
are cysteine proteinases, but they are expressed cantly different in cathepsin K -/- mice. In these
at low levels or are absent in osteoclasts. Cathep- mice the rufed border was irregular, adhesion
sin K has been cloned from rabbit, human, and sites in the sealing zone were open and
murine osteoclasts and osteoclastomas [21, 40, detached, and ne collagen brils were present
71, 106, 107, 146, 162, 173, 183]. Taken together, in the resorption lacunae [157]; all of these
these observations suggest that cathepsin K observations are consistent with altered
plays a major role in osteoclast-mediated bone functionality.
resorption. This idea was further supported by Finally, the studies of Motyckova et al. [130]
the discovery that mutations in the cathepsin K show that cathepsin K is a transcriptional target
gene were responsible for the skeletal disorder, of MITF. As described above, mi/mi MITF mice
pycnodysostosis. The clinical presentation of are osteopetrotic. Osteoclasts from mi/mi mice
that disease includes osteopetrosis, bone are mononuclear, do not form rufed-borders,
fragility, short stature, and multiple cranial exhibit impaired bone resorption, and contain
facial and dental abnormalities [42, 53, 81, 120]. decreased levels of TRAP [117, 132, 174, 185].
The role of cathepsin K was further eluci- The latter observation is consistent with the
dated by studies that examined cathepsin K report that Mif regulates osteoclast TRAP
knockout mice [56, 137, 157] and mice overex- expression. Interestingly, these mice also have a
pressing cathepsin K [89]. Overexpression of signicant reduction in cathepsin K mRNA and
cathepsin K resulted in a 36% reduction in tra- protein expression. On the other hand, overex-
becular bone volume. In these mice, osteoclast pression of MITF up-regulated expression of
number and osteoclast surface were unchanged, cathepsin K in osteoclasts that formed in vitro
but their functionality was not assessed either [130]. These ndings indicate that cathepsin K
in vivo or in vitro [89]. Therefore, enhanced is a transcriptional target of MITF. In cathepsin
osteoclast function could provide an explana- K -/- mice, only bones formed by endochondral
tion for the reduction in bone volume that was ossication were affected; in c-fos -/- mice, on
observed. Conversely, cathepsin K null (-/-) the other hand, osteopetrosis was observed in
mice developed osteopetrosis [56, 157]. Both all bones examined, including the skull [188].
cathepsin K -/- mouse models had a similar Integrins are heterodimeric, transmembrane,
phenotype with a signicant increase in trabec- adhesion receptors that mediate cellmatrix
ular (~3.5-fold increase) and cortical bone and cellcell interactions [181]. The avb3 inte-
volume, and no alteration in osteoclast number. grin is expressed in osteoclasts and plays an
However, both articles report alterations in important role in bone resorption [125]. Mice
Genetics and Mutations Affecting Osteoclast Development and Function 101

decient in the b3 subunit (b3 -/- mice) devel- ing of osteoclast biology, the consequence of
oped osteosclerosis and were hypocalcemic. which will be the development of new thera-
Interestingly, the osteoclasts in these mice were peutics that regulate the skeleton.
normal in appearance, but their numbers had
increased 3.5-fold. Despite their normal appear-
ance, these osteoclasts were dysfunctional in
vitro and ex vivo by a number of criteria. For
Acknowledgments
example, they displayed a signicant reduction
in resorption potential, they failed to spread This work was supported by grants from the
normally, and they showed a general inability to National Institutes of Health to J.A.L (AR38933,
form normal rufed borders or actin rings. AR48714, AR49190) and to M.C.H (AR49109,
Osteoclast number also was decreased in vitro, AR47342) and the Yale Core Center for Muscu-
suggesting arrested differentiation [125]. loskeletal Disorders (AR46032).
Importantly, in vitro studies showed that addi-
tion of high-doses of M-CSF rescued the dys-
functions of osteoclasts generated from b3 -/-
mice [44]. In vivo, these mice produce elevated
References
levels of M-CSF. This may account for the
elevated osteoclast number seen in vivo 1. Al-Humidan A, Ralston SH, Hughes DE, et al. (1991)
Interleukin-6 does not stimulate bone resorption in
versus the diminished osteoclast number seen neonatal mouse calvariae. J Bone Miner Res 6:38.
in vitro [44]. It also was shown that bone 2. Akira S, Hirano T, Taga T, Kishimoto T (1990) Biology
marrow macrophages derived from b3 -/- mice of multifunctional cytokines: IL-6 and related mole-
expressed decreased levels of c-fos, which may, cules (IL-1 and TNF). FASEB J 4:28607.
in turn, account for the arrested differentiation 3. Amano H, Yamada S, Felix R (1998) Colony-
stimulating factor-1 stimulates the fusion process in
of osteoclasts in vitro and possibly in vivo [44]. osteoclasts. J Bone Miner Res 13:84653.
While overexpression of c-fos in osteoclasts 4. Amling M, Neff L, Priemel M, Schilling AF, Rueger JM,
generated from b3 -/- mice also was able to nor- Baron R (2000) Progressive increase in bone mass and
malize osteoclast number, these cells were development of odontomas in aging osteopetrotic c-
src-decient mice. Bone 27:60310.
unable to resorb bone. This showed that the avb3 5. Anderson DM, Maraskovsky E, Billingsley, et al. (1997)
integrin is required for bone resorption to take A homologue of the TNF receptor and its ligand
place [44]. enhance T-cell growth and dendritic-cell function.
Nature 390:1759.
6. Anderson KL, Smith KA, Pio F, et al. (1998) Neutrophils
decient in PU.1 do not terminally differentiate or
Conclusion become functionally competent. Blood 92:157685.
7. Angel P, Karin M (1991) The role of Jun, Fos and the
AP-1 complex in cell-proliferation and transformation.
It is clear from the work presented here that Biochem Biophys Acta 1072, 12957.
8. Arai F, Miyamoto T, Ohneda O, et al. (1999) Commit-
formation of bone-resorbing osteoclasts from ment and differentiation of osteoclast precursor cells
progenitors is a highly ordered multi-step by the sequential expression of c-Fms and receptor
process. The identication of spontaneous activator of nuclear factor KB (RANK) receptors. J Exp
genetic mutations and our ability to engineer Med 12:174154.
gene expression has resulted in a quantum leap 9. Azuma Y, Kaji K, Katogi R, Takeshita S, Kudo A (2000)
Tumor necrosis factor-alpha induces differentiation
forward in our understanding of this process. of and bone resorption by osteoclasts. J Biol Chem
Regulation of osteoclast formation can occur at 275:485864.
many steps along the pathway leading to func- 10. Bain G, Robanus Maandag EC, Izon DJ, et al. (1994)
tional osteoclasts. This high degree of regula- E2A proteins are required for proper B cell develop-
ment and initiation of immunoglobulin gene
tion suggests that osteoclast formation, and rearrangements. Cell 79:88592.
thereby the function that osteoclasts carry out, 11. Bellido T, Stahl N, Farruggella TJ, Borba V,
is important to the survival of the animal. Yancopoulos GD, Manolagas SC (1996) Detection of
Although much is now known about osteoclast receptors for interleukin-6, interleukin-11, leukemia
formation and function, the ability to use inhibitory factor, oncostatin M, and ciliary neu-
rotrophic factor in bone marrow stromal osteoblastic
genetic tools will, without question, allow cells. J Clin Invest 97:4317.
identication of new regulatory molecules. Such 12. Bertolini DR, Nedwin GE, Bringman TS, Smith DD,
discoveries will result in a detailed understand- Mundy GR (1986) Stimulation of bone resorption and
102 Bone Resorption

inhibition of bone formation in vitro by human 31. Dai XM, Ryan GR, Hapel AJ, et al. (2002) Targeted dis-
tumour necrosis factors. Nature 319:51618. ruption of the mouse colony-stimulating factor 1
13. Beutler B, Cerami A (1987) Cachectin: More than a receptor gene results in osteopetrosis, mononuclear
tumor necrosis factor. N Eng J Med 316:37985. phagocyte deciency, increased primitive progenitor
14. Black K, Garrett IR, Mundy GR (1991) Chinese hamster cell frequencies, and reproductive defects. Blood 99:
ovary cells transfected with the murine interleukin-6 11120
gene cause hypercalcemia as well as cachexia, leukocy- 32. De La Mata J, Uy HL, Guise TA, et al. (1995) Interleukin-
tosis and thrombocytosis in tumor bearing nude mice. 6 enhances hypercalcemia and bone resorption medi-
Endocrinology 128:26579. ated by parathyroid hormone-related protein in vivo.
15. Blair HC, Khan AJ, Crouch EC, Jeffrey JJ, Teitelbaum SL J Clin Invest 95:284652.
(1986) Isolated osteoclasts resorb the organic and inor- 33. DeKoter RP, Walsh JC, Singh H (1998) PU.1 regulates
ganic components of bone. J Cell Biol 102:116472. both cytokine-dependent proliferation and differenti-
16. Bossard MJ, Tomaszek TA, Thompson SK, et al. (1996) ation of granulocyte/macrophage progenitors. EMBO
Proteolytic activity of human osteoclast cathepsin I: J 17:445668.
Expression, purication, activation, and substrate 34. DeKoter RP, Lee H-J, Singh H (2002) PU.1 regulates
identication. J Biol Chem 271:1251724. expression of the interleukin-7 receptor in lymphoid
17. Boyle WJ, Simonet WS, Lacey DL (2003) Osteoclast dif- progenitors. Immunity 16:297309.
ferentiation and activation. Nature 423:33742. 35. DeKoter RP, Singh H (2000) Regulation of B lympho-
18. Boyce BF, Aufdemorte TB, Garrett R, Yates AJP, Mundy cyte and macrophage development by graded expres-
GR (1989) Effects of interleukin-1 on bone turnover in sion of PU.1. Science 288:143941.
normal mice. Endocrinology 125:1142. 36. Dinarello CA (1991) Interleukin-1 and interleukin-1
19. Boyce BF, Chen H, Soriano P, Mundy GR (1993) Histo- antagonism. Blood 77:162752.
morphometric and immunocytochemical studies of 37. Dinarello CA (1993) Blocking interleukin-1 in disease.
src-related osteopetrosis. Bone 14:33540. Blood Purif 11:11827.
20. Brndstrm H, Jonsson KB, Vidal O, Ljunghall S, 38. Dougall WC, Glaccum M, Charrier K, et al. (1999)
Ohlsson C, Ljunggren (1998) Tumor necrosis factor- RANK is essential for osteoclast and lymph node
a and -b upregulate the levels of osteoprotegerin development. Genes Dev 13:241224.
mRNA in human osteosarcoma MG-63 cells. Biochem 39. Drake FH, Dodds RA, James IR, et al. (1996) Cathepsin
Biophys Res Commun 248:4547. K, but not cathepsins B, L, or S, is abundantly expressed
21. Bromme D, Okamoto K (1995) The baculovirus in human osteoclasts. J Biol Chem 271:1251116.
cysteine protease has a cathepsin B-like S2-subsite 40. Drake FH, James IE, Connor JR, et al. (1994) Identi-
specicity. Biol Chem 376:61115. cation of a novel osteoclast-selective human cysteine
22. Bromme D, Okamoto K, Wang W, Biroc S (1996) proteinase. J Bone Miner Res 9:S177.
Human cathepsin 02, a matrix protein-degrading 41. Eder J (1997) Tumour necrosis factor alpha and inter-
cysteine protease expressed in osteoclasts: Functional leukin 1 signalling: do MAPKK kinases connect it all?
expression of human cathepsin 02 in spodoptera Trends Pharmacol Sci 18:31922.
frugiperda and characterization of the enzyme. J Biol 42. Edelson JG, Obad S, Geiger R, On A, Artul HJ (1992)
Chem 271:212632. Pycnodysostosis. Orthopedic aspects with a descrip-
23. Brown MT, Cooper JA (1996) Regulation, substrates, tion of 14 new cases. Clin Orthop Relat Res 280:26476.
and functions of Src. BBA Rev. Cancer 1287:12149. 43. Faccio R, Novack DV, Zallone A, Ross FP, Teitelbaum SL
24. Bucay N, Sarosi I, Dunstan CR, et al. (1998) Osteopro- (2003) Dynamic changes in the osteoclast cytoskeleton
tegerin-decient mice develop early onset osteoporo- in response to growth factors and cell attachment are
sis and arterial calcication. Genes Dev 12:12608. controlled by b3 integrin. J Cell Biol 162:499509.
25. Burger D, Chicheportiche R, Giri JG, Dayer J-M (1995) 44. Faccio R, Takeshita S, Zallone A, Ross FP, Teitelbaum
The inhibitory activity of human interleukin-1 recep- SL (2003) C-Fms and the av-b3 integrin collaborate
tor antagonist is enhanced by type II interleukin-1 during osteoclast differentiation. J Clin Invest 111:
soluble receptor and hindered by type 1 interleukin-1 74958.
soluble receptor. J Clin Invest 96:3841. 45. Felix R, Fleisch H, Elford PR (1989) Bone-resorbing
26. Chambers TJ, Fox S, Jagger CJ, Lean JM, Chow JWM cytokines enhance release of macrophage colony-
(1999) The role of prostaglandins and nitric oxide stimulating activity by the osteoblastic cell MC3T3- E1.
in the response of bone to mechanical forces. Calcif Tissue Int 44:35660.
Osteoarthritis Cartilage 7:4223. 46. Felix R, Cecchini MG, Hofstetter W, Elford PR, Stutzer
27. Crabtree GR, Olson EN (2002) NFAT signaling: A, Fleisch H (1990) Impairment of macrophage colony-
Choreographing the social lives of cells. Cell 109 stimulating factor production and lack of resident
Suppl:S6779. bone marrow macrophages in the osteopetrotic op/op
28. Cerretti DP, Wignall J, Anderson D, Tushinski RJ, Gallis mouse. J Bone Miner Res 5:7819.
B, Cosman D (1990) Membrane bound forms of human 47. Felix R, Cecchini MG, Fleisch H (1990) Macrophage
macrophage colony stimulating factor (M-CSF, CSF-1). colony stimulating factor restores in vivo bone resorp-
Prog Clin Biol Res 352:6370. tion in the op/op osteopetrotic mouse. Endocrinology
29. Cerretti DP, Wignall J, Anderson D, et al. (1988) Human 127:25924.
macrophage-colony stimulating factor: alternative 48. Fiers W (1993) Tumor Necrosis Factor. In: Sim E (ed)
RNA and protein processing from a single gene. Mol The Natural Immune System: Humoral Factors. IRL
Immunol 25:76170. Press at Oxford University Press, Oxford, pp 65119.
30. Colotta F, Re F, Muzio M, et al. (1993) Interleukin-1 type 49. Franzoso G, Carlson L, Xing L, et al. (1997) Require-
II receptor: A decoy target for IL-1 that is regulated by ment for NF-kappaB in osteoclast and B-cell develop-
IL-4. Science 261:4725. ment. Genes Dev 11:348296.
Genetics and Mutations Affecting Osteoclast Development and Function 103

50. Fuller K, Owens JM, Jagger CJ, Wilson A, Moss R, 67. Horowitz MC, Xi Y, Pugh DL, et al. (2002) Severe
Chambers TJ (1993) Macrophage colony-stimulating osteopenia with increased osteoclast progenitors in
factor stimulates survival and chemotactic behavior in Pax5 decient mice. JBMR #1098:S148 (abst).
isolated osteoclasts. J Exp Med 178:173344. 68. Hsu H, Lacey DL, Dunstan CR, et al. (1999) Tumor
51. Fuller K, Chambers TJ (1989) Effect of arachidonic acid necrosis factor receptor family member RANK medi-
metabolites on bone resorption by isolated rat osteo- ates osteoclast differentiation and activation induced
clasts. J Bone Miner Res 4:20915. by osteoprotegerin ligand. Proc Natl Acad Sci USA
52. Garnero PO, Borel IB, Ferreras M, et al. (1998) The 96:35405.
collagenolytic activity of cathepsin K is unique 69. Huang J, Gao X, Li S, Cao Z (1997) Recruitment of
among mammalian proteinases. J Biol Chem 273: IRAK to the interleukin 1 receptor complex requires
3234752. interleukin 1 receptor accessory protein. Proc Natl
53. Gelb BD, Shi G-P, Chapman HA, Desnick RJ (1996) Pyc- Acad Sci USA 94:1282932.
nodysostosis, a lysosomal disease caused by cathepsin 70. Humphrey MB, Ogasawara K, Yao W, et al. (2004) The
K deciency. Science 273:12368. signaling adapter protein DAP12 regulates multinucle-
54. Glowacki J, Cox KA, Wilcon S (1989) Impaired osteo- ation during osteoclast development. J Bone Miner Res
clast differentiation in subcutaneous implants of bone 19:22434.
particles in osteopetrotic mutants. J Bone Miner Res 71. Inaoka T, Bilbe G, Ishibashi O, Tezuka K, Kumegawa M,
5:2718. Kokubo T (1995) Molecular cloning of human cDNA
55. Gowen M, Chapman K, Littlewood A, Hughes D, Evans for cathepsin K: Novel cysteine proteinase predomi-
D, Russell RGG (1990) Production of tumor necrosis nately expressed in bone. Biochem Biophys Res
factor by human osteoblasts is modulated by other Commun 206:8996.
cytokines, but not by osteotropic hormones. 72. Insogna K, Tanaka S, Neff L, Horne W, Levy J, Baron R
Endocrinology 126:12505. (1997) Role of c-Src in cellular events associated with
56. Gowen M, Lazner F, Dodds R, et al. (1999) Cathepsin K colony-stimulating factor-1-induced spreading in
knockout mice develop osteopetrosis due to a decit in osteoclasts. Mol Reprod Dev 46:1048.
matrix degradation but not demineralization. J Bone 73. Insogna KL, Sahni M, Grey AB, et al. (1997) Colony-
Miner Res 14:165463. stimulating factor-1 induces cytoskeletal reorganiza-
57. Greeneld EM, Horowitz MC, Lavish SA (1996) Stimu- tion and c- src-dependent tyrosine phosphorylation of
lation by parathyroid hormone of interleukin-6 and selected cellular proteins in rodent osteoclasts. J Clin
leukemia inhibitory factor expression in osteoblasts is Invest 100:247685.
an immediate-early gene response induced by cAMP 74. Insogna K, Tanaka S, Neff L, Horne W, Levy J, Baron R
signal transduction. J Biol Chem 271:109849. (1997) Role of c-Src in cellular events associated with
58. Grigoriadis AE, Schellander K, Wang Z-Q, Wagner colony-stimulating factor-1-induced spreading in
EF (1993) Osteoblasts are target cells for transforma- osteoclasts. Molecular Reproduction & Development
tion in C-fos transgenic mice. J Cell Biol 122:685 46:1048.
701. 75. Iotsova V, Caamano J, Loy J, Yang Y, Lewin A, Bravo R
59. Grigoriadis AE, Wang Z-Q, Cecchini MG, et al. (1994) (1997) Osteopetrosis in mice lacking NF-kappaB1 and
c-FOS: a key regulator of osteoclast-macrophage line- NF-kappaB2. Nat Med 3:12859.
age determination and bone remodeling. Science 266: 76. Ishida N, Hayashi K, Hoshijima M, et al. (2002) Large
4436. scale gene expression analysis of osteoclastogenesis in
60. Guise TA, Mundy GR (1998) Cancer and bone. vitro and elucidation of NFAT2 as a key regulator. J Biol
Endocrine Reviews 19:1854. Chem 277:4114756.
61. Hattersley G, Kerby JA, Chambers TJ (1991) Gen- 77. Ishimi Y, Miyaura C, Jin CH, et al. (1990) IL-6 is pro-
eration of osteoclast precursors in multilineage duced by osteoblasts and induces bone resorption.
hematopoietic colonies. Endocrinology 128:25962. J Immunol 145:3297303.
62. Hattersley G, Dorey E, Horton MA, Chambers TJ (1988) 78. Ishimi Y, Abe E, Jin CH, et al. (1992) Leukemia
Human macrophage colony-stimulating factor inhibits inhibitory factor/differentiation-stimulating factor
bone resorption by osteoclasts disaggregated from rat (LIF/D-factor): Regulation of its production and pos-
bone. J Cell Physiol 137:199203. sible roles in bone metabolism. J Cell Physiol 152:718.
63. Hirano T, Akira S, Taga T, Kishimoto T (1990) Biolog- 79. Jilka RL, Hangoc G, Girasole G, et al. (1992) Increased
ical and clinical aspects of interleukin 6. Immunol osteoclast development after estrogen loss: mediation
Today 11:4439. by interleukin-6. Science 257:8891.
64. Hofbauer LC, Dunstan CR, Spelsberg TC, Riggs BL, 80. Jimi E, Shuto T, Koga T (1995) Macrophage colony-
Khosla S (1998) Osteoprotegerin production by human stimulating factor and interleukin-1a maintain the
osteoblast lineage cells is stimulated by vitamin D, survival of osteoclast-like cells. Endocrinology
bone morphogenetic protein-2, and cytokines. 136:80811.
Biochem Biophys Res Commun 250:77681. 81. Johnson MR, Polymeropoulos MH, Vos HL, Ortiz de
65. Hofstetter W, Wetterwald A, Cecchini MC, Felix R, Luna RI, Francomano CA (1996) A nonsense mutation
Fleisch H, Mueller C (1992) Detection of transcripts for in the cathepsin K gene observed in a family with pyc-
the receptor for macrophage colony-stimulating factor, nodysostosis. Genome Res 6:10505.
c-fms, in murine osteoclasts. Proc Natl Acad Sci USA 82. Kaenah W, Brmme D, Buttle DJ, Croucher LJ, Hol-
89:963741. lander AJ (1998) Human cathepsin K cleaves native
66. Horne WC, Neff L, Chatterjee D, Lomri A, Levy JB, type I and II collagens at the N-terminal end of the
Baron R (1992) Osteoclasts express high levels of triple helix. Biochem 331:72732.
pp60c-src in association with intracellular membranes. J 83. Kaifu T, Nakahara J, Inui M, et al. (2003) Osteopetrosis
Cell Biology 119:100313. and thalamic hypomyelinosis with synaptic degenera-
104 Bone Resorption

tion in DAP12-decient mice. J Clin Invest 111:323 101. Lee MY, Eyre DR, Osborne WR (1991) Isolation of
32. murine osteoclast colony-stimulating factor. Proc Natl
84. Kanazawa K, Azuma Y, Nakano H, Kudo A (2003) Acad Sci USA 88:85004.
TRAF5 functions in both RANKL- and TNFalpha- 102. Lees RL, Heersche JNM (1999) Macrophage colony
induced osteoclastogenesis. J Bone Miner Res 18: stimulating factor increases bone resorption in dis-
44350. persed osteoclast cultures by increasing osteoclast size.
85. Kawasaki K, Gao YH, Yokose S, et al. (1997) Osteoclasts J Bone Miner Res 14:93745.
are present in gp130-decient mice. Endocrinology 103. Li J, Sarosi I, Yan XQ, et al. (2000) RANK is the intrin-
138:495965. sic hematopoietic cell surface receptor that controls
86. Kawaguchi H, Pilbeam CC, Harrison JR, Raisz LG osteoclastogenesis and regulation of bone mass and
(1995) The role of prostaglandins in the regulation of calcium metabolism. Proc Natl Acad Sci USA 97:
bone metabolism. Clin Orthop 313:3646. 156671.
87. Kee BL, Murre C (1998) Induction of early B cell factor 104. Li H, Cuartas E, Cui W, et al. (2004) Irak-m is a nega-
(EBF) and multiple B lineage genes by the basic helix- tive regulator of osteoclast. In: Proceedings: Advances
loop-helix transcription factor E12. J Exp Medicine in Skeletal Anabolic Agents for the Treatment of Osteo-
188:699713. porosis, ASBMR 35, 23 (Abstract).
88. Kishimoto T, Taga T, Akira S (1994) Cytokine signal 105. Li X, Okada Y, Pilbeam CC, Lorenzo, et al. (2000)
transduction. Cell 76:25362. Knockout of the murine prostaglandin EP2 receptor
89. Kiviranta R, Morko J, Uusitalo H, Aro HT, Vuorio E, impairs osteoclastogenesis in vitro. Endocinology
Rantakokko J (2001) Accelerated turnover of meta- 141:205461.
physeal trabecular bone in mice overexpressing 106. Li YP, Alexander M, Wucherpfennig AL, Yelick P, Chen
cathepsin K. J Bone Miner Res 16:144452. W, Stashenko P (1995) Cloning and complete coding
90. Kobayashi K, Takahashi N, Jimi E, et al. (2000) sequence of a novel human cathepsin expressed in
Tumor Necrosis Factor alpha Stimulates Osteoclast giant cells of osteoclastomas. J Bone Miner Res
Differentiation by a Mechanism Independent of the 10:11972202.
ODF/RANKL-RANK Interaction. J Exp Med 191:275 107. Li YP, Chen W (1999) Characterization of mouse
86. cathepsin K gene, the gene promoter, and the gene
91. Kodama H, Yamasaki A, Nose M, et al. (1991) Congen- expression. J Bone Miner Res 14:48799.
ital osteoclast deciency in osteopetrotic (op/op) mice 108. Lin H, Grosschedl R (1995) Failure of B-cell differenti-
is cured by injections of macrophage colony stimulat- ation in mice lacking the transcription factor EBF.
ing factor. J Exp Med 173:26972. Nature 376:2637.
92. Koga T, Inul M, Inoue K, et al. (2004) Costimulatory 109. Linkhart TA, Linkhart SG, MacCharles DC, Long DL,
signals mediated by the ITAM motif cooperate with Strong DD (1991) Interleukin-6 messenger RNA
RANKL for bone homeostasis. Nature 428:75863. expression and interleukin-6 protein secretion in cells
93. Kong YY,Yoshida H, Sarosi I, et al. (1999) OPGL is a key isolated from normal human bone: regulation by inter-
regulator of osteoclastogenesis, lymphocyte develop- leukin-1. J Bone Miner Res 6:128594.
ment and lymph-node organogenesis. Nature 397:315 110. Littlewood EA, Kokubo T, Ishibashi O, et al. (1997)
23. Localization of cathepsin K in human osteoclasts by in
94. Korherr C, Hofmeister R, Wesche H, Falk W (1997) A situ hybridization and immunohistochemistry. Bone
critical role for interleukin-1 receptor accessory 20:816.
protein in interleukin-1 signaling. Eur J Immunol 111. Lomaga MA, Yeh WC, Sarosi I, et al. (1999) TRAF6
27:2627. deciency results in osteopetrosis and defective
95. Kurihara N, Chenu C, Civin CI, Roodman GD (1990) interleukin-1, CD40, and LPS signaling. Genes Dev
Identication of committed mononuclear precursors 13:101524.
for osteoclast-like cells formed in long-term marrow 112. Lorenzo JA, Sousa S, Alander C, Raisz LG, Dinarello CA
cultures. Endocrinology 126:273341. (1987) Comparison of the bone-resorbing activity in
96. Kurihara N, Bertolini D, Suda T, Akiyama Y, Roodman the supernatants from phytohemagglutinin-stimu-
GD (1990) IL-6 stimulates osteoclast-like multi- lated human peripheral blood mononuclear cells with
nucleated cell formation in long term human marrow that of cytokines through the use of an antiserum to
cultures by inducing IL-1 release. J Immunol 144: interleukin 1. Endocrinology 121:116470.
422630. 113. Lorenzo JA, Naprta A, Rao Y, et al. (1998) Mice lacking
97. Lacey DL, Timms E, Tan H-L, et al. (1998) Osteoprote- the type I interleukin-1 receptor do not lose bone mass
gerin (OPG) ligand is a cytokine that regulates osteo- after ovariectomy. Endocrinology 139:30225.
clast differentiation and activation. Cell 93:16576. 114. Lorenzo JA, Sousa SL, Leahy CL (1990) Leukemia
98. Lacey DL, Tan HL, Lu J, et al. (2000) Osteoprotegerin inhibitory factor (LIF) inhibits basal bone resorption
ligand modulates murine osteoclast survival in vitro in fetal rat long bone cultures. Cytokine 2:26671.
and in vivo. Am J Pathol 157:43548. 115. Lowik CW, Van der PG, Bloys H, et al. (1989) Parathy-
99. Lagasse E, Weissman IL (1997) Enforced expression of roid hormone (PTH) and PTH-like protein (PLP) stim-
Bcl-2 in monocytes rescues macrophages and partially ulate interleukin-6 production by osteogenic cells: a
reverses osteopetrosis in op/op mice. Cell 89:1021 possible role of interleukin-6 in osteoclastogeneis.
31. Biochem Biophys Res Commun 162:154652.
100. Lakkakorpi PT, Brett A, Lipfert L, Rodan GA, Duong Le 116. Luchin A, Suchting S, Merson T, et al. (2001) Genetic
T (2003) PYK2 atuophosphorylation, but not kinase and physical interactions between Microphthalmia
activity, is necessary for adhesion-induced association transcription factor and PU.1 are necessary for osteo-
with c-Src, osteoclast spreading, and bone resorption. clast gene expression and differentiation. J Biol Chem
J Biol Chem 278:1150212. 276:3670310.
Genetics and Mutations Affecting Osteoclast Development and Function 105

117. Luchin A, Purdom G, Murphy K, et al. (2000) The 135. ORiordan M, Grosschedl R (1999) Coordinate regula-
microphthalmia transcription factor regulates expres- tion of B cell differentiation by the transcription
sion of the tartrate-resistant acid phosphatase gene factors EBF and E2A. Immunity 11:2131.
during terminal differentiation of osteoclasts. J Bone 136. Okada Y, Lorenzo JA, Freeman AM, et al. (2000)
Miner Res 15:45160. Prostaglandin G/H synthase-2 is required for maximal
118. Manolagas SC, Jilka RL (1995) Mechanisms of disease: formation of osteoclast-like cells in culture. J Clin
Bone marrow, cytokines, and bone remodeling Invest 105:82332.
Emerging insights into the pathophysiology of osteo- 137. Okaji M, Sakai H, Sakai E, et al. (2003) The regulation
porosis. N Engl J Med 332:30511. of bone resorption in tooth formation and eruption
119. Manolagas SC, Bellido T, Jilka RL (1995) New insights processes in mouse alveolar crest devoid of cathepsin
into the cellular, biochemical, and molecular basis of K. J Pharmacol Sci 91:285294.
postmenopausal and senile osteoporosis: Roles of IL-6 138. Oliff A (1988) The role of tumor necrosis factor
and gp130. Intl J Immunopharm 17:10916. (cachectin) in cachexia. Cell 54:1412.
120. Maroteaux P, Lamy M (1962) Pyknodysostosis. Presse 139. Orkin SH (2000) Diversication of hematopoietic stem
Med 70:9991002. cells to specic lineages. Nat Rev Genet 1:5764.
121. Martin MU, Falk W (1997) The interleukin-1 receptor 140. Pacici R, Rifas L, McCracken R, Avioli LV (1990) The
complex and interleukin-1 signal transduction. Eur role of interleukin-1 in postmenopausal bone loss. Exp
Cytokine Netw 8:517. Gerontol 25:30916.
122. Marusic A, Kalinowski JF, Jastrzebski S, Lorenzo JA 141. Palmqvist P, Persson E, Conaway HH, Lerner UH
(1993) Production of leukemia inhibitory factor (2002) IL-6, Leukemia Inhibitory Factor, and Onco-
mRNA and protein by malignant and immortalized statin M Stimulate Bone Resorption and Regulate the
bone cells. J Bone Miner Res 8:61724. Expression of Receptor Activator of NF-kappaB
123. Marzia M, Sims NA, Voit S, et al. (2000) Decreased c- Ligand, Osteoprotegerin, and Receptor Activator of
Src expression enhances osteoblast differentiation and NF-kappaB in Mouse Calvariae. J Immunol 169:3353
bone formation. J Cell Biology 151:31120. 62.
124. Matsuo K, Galson DL, Zhao C, et al. (2004) Nuclear 142. Pfeffer K, Matsuyama T, Kndig TM, et al. (1993) Mice
Factor of Activated T-cells (NFAT) rescues osteoclas- decient for the 55 kd tumor necrosis factor receptor
togenesis in precursors lacking c-Fos. J Biol Chem are resistant to endotoxic shock, yet succumb to L.
279:2647580. monocytogenes infection. Cell 73:45767.
125. McHugh KP, Hodivala-Dilke K, Zheng M-H, et al. 143. Pfeilschifter J, Chenu C, Bird A, Mundy GR, Roodman
(2000) Mice lacking [beta]3 integrins are osteosclerotic GD (1989) Interleukin-1 and tumor necrosis factor
because of dysfunctional osteoclasts. J Clin Invest stimulate the formation of human osteoclast-like cells
105:43344. in vitro. J Bone Min Res 4:11318.
126. Missbach M, Jeschke M, Feyen J, et al. (1999) A novel 144. Poli V, Balena R, Fattori E, et al. (1994) Interleukin-6
inhibitor of the tyrosine kinase Src suppresses phos- decient mice are protected from bone loss caused by
phorylation of its major cellular substrates and estrogen depletion. EMBO Journal 13:118996.
reduces bone resorption in vitro and in rodent models 145. Raisz LG (1999) Prostaglandins and bone: physiology
in vivo. Bone 24:43749. and pathophysiology. Osteoarthritis Cartilage 7:419
127. Mizuno A, Amizuka N, Irie K, et al. (1998) Severe 21
osteoporosis in mice lacking osteoclastogenesis 146. Rantakokko J, Aro HT, Savontaus M, Vuorio E (1996)
inhibitory factor/osteoprotegerin. Biochem Biophys Mouse cathepsin K: cDNA cloning and predominant
Res Commun 247:61015. expression of the gene in osteoclasts, and in some
128. Mcsai A, Humphrey MB, Van Zife JAG, et al. (2004) hypertrophying chondrocytes during mouse develop-
The immunomodulatory adapter proteins DAP12 and ment. FEBS Lett 393:30713.
Fc receptor g-chain (FcRg) regulate development of 147. Reddy SV, Takahashi S, Dallas M, Williams RE, Neckers
functional osteoclasts through the Syk tyrosine kinase. L, Roodman GD (1994) Interleukin-6 antisense deoxy-
Proc Natl Acad Sci USA 101:615863. oligonucleotides inhibit bone resorption by giant cells
129. Moore KJ (1995) Insight into the microphthalmia gene. from human giant cell tumors of bone. J Bone Miner
Trends Genet 11:4428. Res 9:7537.
130. Motyckova G, Weilbaecher KN, Horstmann M, Rieman 148. Reid IR, Lowe C, Cornish J, et al. (1990) Leukemia
DJ, Fisher DZ, Fisher DE (2001) Linking osteopetrosis inhibitory factor: a novel bone-active cytokine. Endo-
and pycnodysostosis: regulation of cathepsin K expres- cinology 126:141620.
sion by the microphthalmia transcription factor 149. Roggia C, Gao Y, Cenci S, et al. (2001) Up-regulation
family. Proc Natl Acad Sci USA 98:5798803. of TNF-producing T cells in the bone marrow: A
131. Mundy GR (1989) Local factors in bone remodeling. key mechanism by which estrogen deciency in-
Recent Prog Horm Res 45:50727. duces bone loss in vivo. Proc Natl Acad Sci USA
132. Murphy HM (1973) The osteopetrotic syndrome in the 98:139605.
microphthalmic mutant mouse. Calcif Tissue Res 150. Roodman GD (1996) Advances in Bone Biology: The
13:1926. osteoclast. Endocrine Reviews 17:30832.
133. Naito A, Azuma S, Tanaka S, et al. (1999) Severe 151. Roodman GD (1992) Interleukin-6: an osteotropic
osteopetrosis, defective interleukin-1 signaling and factor? J Bone Miner Res 7:4758.
lymph node organogenesis in TRAF6-decient mice. 152. Roodman GD, Kurihara N, Ohsaki Y, et al. (1992) A
Genes Cells 4:35362. potential autocrine/paracrine factor in Pagets disease
134. Nutt SL, Heavey B, Rolink AG, Busslinger M (1999) of bone. J Clin Invest 89:4652.
Commitment to the B-lymphoid lineage depends on 153. Roth P, Stanley ER (1992) The biology of CSF-1 and its
the transcription factor Pax5. Nature 401:55662. receptor. Curr Top Microbiol Immunol 181:14167.
106 Bone Resorption

154. Rothe J, Lesslauer W, Lotscher H, et al. (1993) Mice mation by interleukin 6. Proc Natl Acad Sci USA
lacking the tumour necrosis factor receptor 1 are resis- 90:119248.
tant to TNF-mediated toxicity but highly susceptible 172. Tanaka S, Amling M, Neff L, et al. (1996) c-Cbl is down-
to infection by Listeria monocytogenes. Nature 364: stream of c-Src in a signaling pathway necessary for
798802. bone resorption. Nature 383:52831.
155. Rther U, Komitowski D, Schubert FR, Wagner EF 173. Tezuka K, Tezuka Y, Maijima A, et al. (1994) Molecular
(1989) c-fos expression induces bone tumors in trans- cloning of a possible cytokine proteins predominantly
genic mice. Oncogene 4:8615. expressed in osteoclasts. J Biol Chem 269:11069.
156. Rubin J, Fan X, Thornton D, Bryant R, Biskobing D 174. Thesingh CW, Scherft JP (1985) Fusion disability of
(1996) Regulation of murine osteoblast macrophage embryonic osteoclast precursor cells and macrophages
colony-stimulating factor production by 1,25(OH)2D3. in the microphthalmic osteopetrotic mouse. Bone 6:
Calcif Tissue Int 59:2916. 4352.
157. Saftig P, Hunziker E, Wehmeyer O, et al. (1998) 175. Tiffee JC, Xing L, Nilsson S, Boyce BF (1999) Dental
Impaired osteoclastic bone resorption leads to abnormalities associated with failure of tooth eruption
osteopetrosis in cathepsin-K-decient mice. Proc Natl in src knockout and op/op mice. Calcif Tissue Int
Acad Sci USA 95:134538. 65:538.
158. Sanjay A, Houghton A, Neff L, et al. (2001) Cbl Associ- 176. Tondravi MM, McKercher, Anderson K, et al. (1997)
ates with Pyk2 and Src to regulate Src kinase activity, Osteopetrosis in mice lacking haematopoietic tran-
avb3 integrin-mediated signaling, cell adhesion, and scription factor PU.1. Nature 386:814.
osteoclast motility. J Cell Biol 152:18196. 177. Udagawa N, Takahashi N, Akatsu, T, et al. (1989) The
159. Sarma U, Flanagan AM (1996) Macrophage colony- bone marrow-derived stromal cell lines MC3T3-
stimulating factor induces substantial osteoclast gen- G2/PA6 and ST2 support osteoclast-like cell differenti-
eration and bone resorption in human bone marrow ation in cocultures with mouse spleen cells.
cultures. Blood 88:253140. Endocrinology 125:180513.
160. Schwartzberg PL, Xing L, Hoffman O, et al. (1997) 178. Umeda S, Beamer WG, Takagi K, et al. (1999) Deciency
Rescue of osteoclast function by transgenic expression of SHP-1 protein-tyrosine phosphatase activity results
of kinase-decient Src in src-/- mutant mice. Genes & in heightened osteoclast function and decreased bone
Development 11:283544. density. Am J Pathol 155:22333.
161. Screpanti I, Romani L, Musiani P, et al. (1995) Lym- 179. Urbanek P,Wang Z, Fetka I, et al. (1994) Complete block
phoproliferative disorder and imbalance T-helper of early B cell differentiation and altered patterning of
response in C-EBP beta-decient mice. EMBO 14: the posterior midbrain in mice lacking Pax5/BSAP. Cell
193241. 79:90112.
162. Shi GP, Chapman HA, Bhairi SM, DeLeeuw C, Reddy 180. Van Beek E, Van der Wee-Pals L, et al. (1993) Leukemia
VY, Weiss SJ (1995) Molecular cloning of human inhibitory factor inhibits osteoclastic resorption,
cathepsin O, a novel endoproteinase and homologue of growth, mineralization, and alkaline phosphatase
rabbit OC2. FEBS Lett 357:12934. activity in fetal mouse metacarpal bones in culture. J
163. Shultz LD, Schweitzer PA, Rajan TV, et al. (1993) Muta- Bone Miner Res 8:1918.
tions at the murine motheaten locus are within the 181. van der Flier A, Sonnenberg A (2001) Function and
hematopoietic cell protein-tyrosine phosphates interactions of integrins. Cell Tissue Res 305:285
(Hcph) gene. Cell 73:144554. 98.
164. Simonet WS, Lacey DL, Dunstan CR, et al. (1997) 182. Vargas SJ, Naprta A, Glaccum M, Lee SK, Kalinowski J,
Osteoprotegerin: a novel secreted protein involved in Lorenzo JA (1996) Interleukin-6 expression and histo-
the regulation of bone density. Cell 89:30919. morphometry of bones from mice decient for recep-
165. Sims JE, Gayle MA, Slack JL, et al. (1993) Interleukin 1 tors for interleukin-1 or tumor necrosis factor. J Bone
signaling occurs exclusively via the type I receptor. Miner Res 11:173644.
Proc Natl Acad Sci USA 90:61559. 183. Velasco G, Ferrando AA, Puente XS, Sanchez LM,
166. Soriano P, Montgomery C, Geske R, Bradley A (1991) Lopez-Otin CJ (1994) Human cathepsin O. Molecular
Targeted disruption of the c-srd proto-oncogene leads cloning from a breast carcinoma, production of the
to osteopetrosis in mice. Cell 64:693702. active enzyme in Escherichia coli, and expression
167. Steingrimsson E, Moore KJ, Lamoreux ML, et al. (1994) analysis in human tissues. J Biol Chem 269:2713642.
Molecular basis of mouse microphthalmia mi muta- 184. Vidal ON, Sjgren K, Eriksson BI, Ljunggren ,
tions helps explain their developmental and pheno- Ohlsson C (1998) Osteoprotegerin mRNA is increased
typic consequences. Nat Genet. 8:25663. by interleukin-1a in the human osteosarcoma cell line
168. Suzawa T, Miyaura C, Inada M, et al. (2000) The role of MG-63 and in human osteoblast-like cells. Biochem
prostaglandin E receptor subtypes (EP1, EP2, EP3, and Biophys Res Commun 248:696700.
EP4) in bone resorption: an analysis using specic ago- 185. Walker DG (1975) Bone resorption restored in
nists for the respective EPs. Endocrinology 141:15549. osteopetrotic mice by transplants of normal bone
169. Tagaya H, Kunisada T, Yamazaki H, et al. (2000) marrow and spleen cells. Science 190:7845.
Intramedullary and extramedullary B lymphopoiesis 186. Wang Z-Q, Grigoriadis AE, Mhle-Steinlein U, Wagner
in osteopetrotic mice. Blood 95:336370. EF (1991) A novel target cell for c-fos-induced onco-
170. Takayanagi H, Kim S, Koga T, et al. (2002) Induction genesis: development of chondrogenic tumours in
and Activation of the Transcription Factor NFATc1 embryonic stem cell chimeras. EMBO 10:243750.
(NFAT2) Integrate RANKL Signaling in Terminal Dif- 187. Wang Z-Q, Grigoriadis AE, Wagner EF (1993) Stable
ferentiation of Osteoclasts. Dev Cell 3:889901. murine chondrogenic cell lines derived from c-fos-
171. Tamura T, Udagawa N, Takahashi N, et al. (1993) induced cartilage tumors. J Bone Miner Res 8:839
Soluble interleukin-6 receptor triggers osteoclast for- 47.
Genetics and Mutations Affecting Osteoclast Development and Function 107

188. Wang Z-Q, Ovitt C, Grigoriadis AE, et al. (1992) Bone ducers mediates NF-kappaB activation by the
and haematopoietic defects in mice lacking c-fos. TRANCE receptor. J Biol Chem 273:283559.
Nature 360:7415. 196. Xie H, Ye M, Feng R, Graf T (2004) Stepwise repro-
189. Wani MR, Fuller K, Kim NS, Choi YW, Chambers T gramming of B cells into macrophages. Cell 117:663
(1999) Prostaglandin E2 cooperates with TRANCE in 76.
osteoclast induction from hemopoietic precursors: 197. Xing L, Bushnell TP, Carlson L, et al. (2002) NF-kappaB
Synergistic activation of differentiation, cell spreading, p50 and p52 expression is not required for RANK-
and fusion. Endocrinology 140:192735. expressing osteoclast progenitor formation but is
190. Ware CB, Horowitz MC, Renshaw BR, et al. (1995) essential for RANK- and Cytokine-Mediated Osteo-
Targeted disruption of the low-afnity leukemia clastogenesis. J Bone Miner Res 17:120010.
inhibitory factor receptor gene causes placental, skele- 198. Yao GQ, Sun BH, Hammond EE, et al. (1998) The cell-
tal, neural and metabolic defects and results in peri- surface form of colony-stimulating factor-1 is regu-
natal death. Development 121:128399. lated by osteotropic agents and supports formation of
191. Weir EC, Horowitz MC, Baron R, Centrella M, Kacinski multinucleated osteoclast-like cells. J Biol Chem 273:
BM, Insogna KL (1993) Macrophage colony-stimulat- 411928.
ing factor release and receptor expression in bone cells. 199. Yamamoto T, Ozono K, Kasayama S, et al. (1996)
J Bone Miner Res 8:150718. Increased IL-6-production by cells isolated from
192. Wesche H, Korherr C, Kracht M, Falk W, Resch K, the brous bone dysplasia tissues in patients
Martin MU (1997) The interleukin-1 receptor acces- with McCune-Albright syndrome. J Clin Invest 98:
sory protein (IL-1RAcP) is essential for IL-1-induced 305.
activation of interleukin-1 receptor-associated kinase 200. Yasuda H, Shima N, Nakagawa N, et al. (1998) Osteo-
(IRAK) and stress-activated protein kinases (SAP clast differentiation factor is a ligand for osteoprote-
kinases). J Biol Chem 272:772731. gerin/osteoclastogenesis-inhibitory factor and is
193. Wiktor Jedrzejczak W, Bartocci A, Ferrante AJr, et al. identical to TRANCE/RANKL. Proc Natl Acad Sci USA
(1990) Total absence of colony-stimulating factor 1 in 95:3597602.
the macrophage-decient osteopetrotic (op/op) 201. Yoshida H, Hayashi S, Kunisada T, et al. (1990) The
mouse. Proc Natl Acad Sci USA 87:482832. murine mutation osteopetrosis is in the coding region
194. Wong BR, Rho J, Arron J, et al. (1997) TRANCE is a of the macrophage colony stimulating factor gene.
novel ligand of the tumor necrosis factor receptor Nature 345:4424.
family that activates c-jun N-terminal kinase in T cells. 202. Yoshida K, Oida H, Kobayashi T, et al. (2002) Stimula-
J Biol Chem 272:251904. tion of bone formation and prevention of bone loss by
195. Wong BR, Josien R, Lee SY, Vologodskaia M, Steinman prostaglandin E EP4 receptor activation. Proc Natl
RM, Choi Y (1998) The TRAF family of signal trans- Acad Sci USA 99:45805.
7.
Clinical Disorders Associated with
Alterations in Bone Resorption
Janet Rubin and Mark S. Nanes

This chapter is dedicated to Dr. Gideon Rodan, bone mineral density and increased fragility.
whose sustained and signicant contributions These include epigenetic diseases, such as
to the eld of clinical bone disease are much postmenopausal osteoporosis or hyperparathy-
admired by the authors. roidism, and diseases with a genetic basis or
predisposition such as Pagets disease. Table 7.1
lists genetic disorders of osteoclast over-
function.
Introduction In the second part of the chapter we will con-
sider the largely genetic diseases where osteo-
The osteoclasts specialized function is irre- clast function is decreased (Table 7.2). These
placeable by other cells. It is the only cell that vary in their phenotypic spectrum from posi-
resorbs bone, dentine, and mineralized cartilage; tive effects on bone as in the fracture-resistant
therefore defects in osteoclast function lead to a LRP5 families, to lethal effects that result from
wide array of disorders. In this chapter we will the absence of the marrow compartment neces-
discuss diseases of unchecked osteoclast activity, sary for hematopoiesis.
which lead to excessive bone resorption and thus Further and continually updated clinical and
result in bone fragility. We will also discuss dis- genetic information regarding those inherited
eases that result from too little osteoclast activ- disorders covered in this chapter are referred to
ity, which cause bone mineral content to increase in Online Mendelian Inheritance in Man
and lead to disordered bone structure (Figure (OMIM) and are shown as numbers in the text
7.1). The altered material properties associated and tables. The OMIM website can be accessed
with either decreased bone density (osteoporo- at: www.ncbi.nlm.nih.gov.
sis) or increased bone density (osteopetrosis or
osteosclerosis) can lead to skeletal failure and
fracture. Some of these disorders result from
causes extraneous to the osteoclast itself, such as Increased Osteoclast Activity
hormonal changes, and some reect inherited
defects in osteoclast formation or function.
Resulting in Decreased Bone
Many genetic disorders resulting in osteoclast Mass or Disordered Skeletal
dysfunction have been duplicated in transgenic
mice where alterations to a specic gene have led
Architecture
to abnormalities in bone remodeling (see
Horowitz, Chapter 6). The clinical term osteoporosis refers to condi-
We shall rst consider diseases where the tions in which a decrease in bone mineral
osteoclast is too active, resulting in decreased density leads to decreased skeletal strength and
108
Clinical Disorders Associated with Alterations in Bone Resorption 109

Osteoclast Function or Number increased risk of skeletal failure. The etiology of


osteoporosis includes a large number of factors
that result in either (a) an increase in osteoclast
activity that leads to increased bone resorption,
Too much: osteoporosis
(b) decreased bone formation resulting from
Too little: osteopetrosis insufcient osteoblast activity, or (c) alterations
in the tight coupling between the activities
of osteoclast and osteoblast that normally
permits the continual remodeling necessary
to allow skeletons to respond to functional
Figure 7.1 Bone remodeling. demands. Alterations in coupling can lead not
only to reduced bone mineral density, but also
to altered skeletal structure, as occurs in Pagets
disease. Even though it is not possible opera-
tionally to separate the effects of osteoclast and
osteoblast dysfunction when analyzing the

Table 7.1. Overactive Osteoclast Phenotype: Inherited


Disease Gene Protein affected Osteoclast phenotype OMIM#
Pagets disease SQSTM1 p62 sequestosome Increased numbers, 602080
hypernucleated
Pagets disease, early onset TNFRSF11A RANK As in PD 602080
Juvenile Pagets disease Osteoprotegerin Increased numbers, 239000
(hyperostosis corticalis hyperactive
deformans juvenalis)
Familial expansile osteolysis TNFRSF11A RANK Hyperactive, enlarged 174810
Expansile skeletal TNFRSF11A RANK Increased numbers, [118]
hyperphosphatasia enlarged
Polycystic lipomembranous DAP12 or DAP12/TREM2 Impaired 221770
osteodysplasia with sclerosing TREM2
leukoencephalopathy
(Nasu-Hakola disease)
Rett syndrome MECP2 Methyl-CpG-binding Unknown 312750
protein-2

Table 7.2. Underactive Osteoclast Phenotype Inherited


Disease Gene Protein affected Osteoclast phenotype OMIM#
Osteopetrosis, malignant TC1RG1 Vacuolar proton pump Number nl/ 259700
Osteopetrosis, malignant CLCN7 ClC-7 chloride channel Number nl/ 259700
Osteopetrosis, malignant GL Gray-lethal Not known 259700
Osteopetrosis, autosomal LRP5 Lipoprotein receptor Reduced in number and 607634
dominant, type I protein 5 size
Osteopetrosis, autosomal CLCN7 ClC-7 chloride channel Increase number and size 166600
dominant, type II
(Albers-Schnberg disease)
Pycnodysostosis CTSK Cathepsin K Normal 265800
Osteopetrosis w RTA CA2 Carbonic anhydrase II Normal 259730
CamuratiEngelmann TGFB1 TGFb1 Increased numbers in vitro 131300
110 Bone Resorption

ensuing bone phenotype (e.g., hyperparathy-


roidism), we have here selected to review those
conditions in which the initial defect can be best
ascribed directly to the osteoclast.

Endocrine Disorders
Sex Hormone Deficiency Leading to
Increased Osteoclast Formation
Adult bone remodeling occurs continuously,
permitting repair and allowing the skeleton to
meet changing functional needs, whether in
response to changes in load or to assure calcium
homeostasis. Modeling begins on the quiescent
bone surface at the time when osteoclasts are
recruited locally. The osteoclastic resorption
process forms a cutting cone in cortical bone, or
a trench in cancellous bone. In the wake of this
osteoclast activity, osteoblasts are recruited;
they induce bone matrix synthesis followed by
mineralization. This forms a packet of remod-
eled bone. The time for a packet to form from
start to nish is nearly eight months; most of
this time is spent forming new bone. The osteo-
clasts resorptive job is completed within the
rst two weeks. If the osteoblast response to
osteoclastic bone destruction is not tightly Figure 7.2 Post-menopausal osteoporosis resulting in
coupled, i.e., if the activation frequency initi- osteopenia with vertebral compression fracture.
ation of osteoclast activity is not followed by
a sufcient osteoblastic response, bone resorp-
tion outpaces bone formation and bone mineral controversy [73, 85]. There are clear data that
is lost [95]. This is, in fact, the situation that pre- show estrogen promotes osteoclast apoptosis
dominates in post-menopausal osteoporosis, [48]. There are also data that show estrogen
and probably also occurs as a result of acute limits osteoclast formation in vitro by inhibiting
testosterone loss in men. When in post- osteoclast recruitment [39, 55]. Estrogen in
menopausal osteoporosis resorption and for- addition regulates the expression of osteoactive
mation is evaluated by biochemical markers, factors such as osteoprotegerin or nitric oxide
resorption is shown to increase by 90%, whereas [91, 107]. Estrogen in vivo most probably mod-
formation markers increase by only 45% [32]. It ulates the RANKL:RANK:OPG axis. Estrogen
is the resulting deterioration in bone microar- deciency is associated with increased RANKL
chitecture and material properties that leads to expression by both stromal cells and circulating
an increased fracture risk (Figure 7.2: osteo- T cells [26]. Estrogen deciency furthermore
porotic spine). contributes to increased RANKL signaling by
In the postmenopausal woman, bone loss is increasing cytokine production [18] (see also
rapid within the rst ve years of estrogen Chapter 5). Estrogen has effects within the
deciency. In the later menopause, the rate of target cell of RANKL: after RANKL activates
bone loss approximates the slower, continuous RANK signaling (receptor activator of nuclear
decline associated with senile osteoporosis [53]. factor-kb, or RANK, is the receptor for RANKL)
Acute estrogen deciency increases the activa- in the osteoclast precursor, estrogen is able to
tion frequency so that more bone surface is mute the downstream signaling amplication,
covered by active osteoclasts. The many mecha- suggesting that estrogen deciency will en-
nisms by which estrogen deciency increases hance response to RANKL [100, 106]. Estrogen
osteoclastic activity are a topic of continued appears to increase osteoprotegerin levels, thus
Clinical Disorders Associated with Alterations in Bone Resorption 111

providing an anti-resorptive factor [46]; inter-


estingly, in the ovariectomized rat, osteoprote-
gerin levels have been shown to rise along with
RANKL [49]; this may represent the skeletons
attempt to restrain rampant osteoclastogenesis.
Estrogen deciency also seems to contribute
to an impaired mechanical response of bone
cells to signals generated during normal daily
physical loading. This loss of a remodeling
response to a load appears to be largely
processed by osteocytes and osteoblasts [27].
For example, the estrogen receptor alpha knock-
out mouse is unable to mount an anabolic
response to loading in terms of periosteal appo-
sition or osteoblast proliferation [66]. There are
also many reports that show that estrogen-de-
cient women, rats and pigs have reduced ana-
bolic responses to exercise measured in the
skeleton [5, 63, 123]. Androgen deciency may
also lead to alterations in the mechanostat
response to loading [62]. In summary, estrogen
modulates osteoclast activity indirectly through
modulation of osteoclastogenic factors, and
through regulation of the anabolic response to
exercise.

Hyperparathyroidism
Hyperparathyroidism is a frequently diagnosed
(1/25,000) endocrine disease, which presents in Figure 7.3 Hyperparathyroidism: Scintigraphy with tech-
netium 99m shows increased bone activity throughout the
most patients as hypercalcemia. The hypercal- skeleton of a hyperparathyroid patient.
cemia arises because of parathyroid hormone
(PTH)-induced osteoclastic bone resorption
(Figure 7.3) and changes in calcium absorption
and excretion. Elevations in serum calcium that increases 1,25-(OH)2D3 synthesis, which in turn
are less than 1.5 mg per deciliter above normal increases intestinal calcium absorption, and
are not usually associated with skeletal disease nally affects release of calcium from skeletal
[9]. Higher elevations of calcium, either due to stores. The target of PTH in bone is the
a parathyroid secreting adenoma or parathyroid stromal/osteoblast cell, which responds by
hyperplasia, generate a spectrum of disease increasing expression of RANKL [68], with an
from renal stones due to an increase in the l- increase in the osteoclast recruitment from the
tered calcium in the renal collecting duct hematopoietic pool.
system, to gastrointestinal complaints, to neuro- The skeletal lesion of hyperparathyroidism is
logical symptoms that can progress to stupor distinguished by osteitis brosa cystica. This
and coma. PTH synthesis and release is largely condition is seen today most commonly in
controlled by the parathyroid gland calcium the hyperparathyroidism accompanying renal
receptor sensing decreased serum calcium failure. Histopathological examination shows
levels [13]. When serum calcium rises, the intense osteoclastic resorption resulting in bone
calcium receptor generates intracellular calcium destruction that leaves a picture of peritrabec-
signals, which inhibit PTH release. Alternatively, ular brosis within an enlarged marrow space.
when serum calcium drops, intracellular The etiology of primary hyperparathyroi-
calcium also drops, causing the release of PTH. dism is unknown. In two familial syndromes
To increase extracellular calcium, released parathyroid hyperplasia is associated with
PTH raises calcium resorption in the kidney, other endocrine abnormalities. One is the
112 Bone Resorption

syndrome of multiple endocrine neoplasia I receptor may enhance osteoclast formation. The
(MEN1) (OMIM 131100), which presents as physiology of TSH control of bone cell function
parathyroid, enteropancreatic neuroendocrine, will be of great interest to endocrinologists in
anterior pituitary, or foregut carcinoid tumors. the coming years.
MEN1 is caused by a mutation in the menin
gene [20] and germline MEN1 mutations have
been identied in many MEN families [75].
Glucocorticoid Excess
Most germline and somatic mutations in MEN1 In 1932 Harvey Cushing described a connection
mutations lead to inactivating mutations of between osteoporosis and the overproduction
menin, suggesting that MEN1 may be a tumor of cortisol by the adrenal glands [24]. Corticos-
suppressor gene such that stepwise inactivation teroid therapy for inammatory diseases such
of both menin alleles will lead to cell cycle entry. as asthma has similar effects, leading to
Indeed, menin interacts with the junD tran- increased incidence of vertebral fracture. Iatro-
scription factor during cell division [75]. MEN2 genic Cushings syndrome is now thought to be
(OMIM 171400) also is associated with hyper- the most common cause of secondary osteo-
parathyroidism, and stems from a mutation porosis. Bone loss is initiated by osteoclastic
in the ret proto-oncogene, a tyrosine kinase remodeling, but is compounded by a deciency
[31]. The ret proto-oncogene mutation, besides of remodeling. The mechanisms by which
inducing parathyroid hyperplasia, also causes corticosteroids affect bone remodeling are
pheochromocytoma and medullary cancer pleiotropic, and cortisol has both direct and
of the thyroid. The skeletal lesions and indirect effects on bone cells.
hypercalcemia in both MEN syndromes are Hypercortisolism has systemic effects on
indistinguishable from those of primary other hormonal axes that indirectly modulate
hyperparathyroidism. bone turnover. Sex hormones, in particular
estradiol and testosterone, are decreased in
patients on high doses of steroids [79]. Phar-
Hyperthyroidism macological use of glucocorticosteroids also is
Hyperthyroidism triggers increased bone associated with inhibition of intestinal calcium
remodeling, producing a skeletal syndrome in absorption [2]. Decreased intestinal calcium
which the osteoclastic lesion predominates [40]. ux stimulates the parathyroid glands to release
Practicing endocrinologists know that hyper- PTH, but the role of secondary hyperparathy-
thyroidism can cause hypercalcemia in as many roidism in glucocorticoid-induced osteoporosis
as 20% of cases; indeed, von Recklinghausen appears to be minor.
recognized hyperthyroidism as a risk fracture Direct effects of glucocorticoids on bone
for osteoporosis a century ago [50]. Bone resorption and formation have been measured
mineral density is signicantly decreased in in multiple studies [17]. Glucocorticoids stimu-
patients with untreated hyperthyroidism and late osteoclast activity and recruitment in vitro:
returns to normal upon treatment [111]. in addition to increasing osteoclast activity [41],
Thyroid hormone has direct effects on bone glucocorticoids stimulate expression of RANKL
cells, and indeed thyroid hormone can induce by bone stromal cells [124]. Impairment in bone
the expression of RANKL in bone cells [78]. formation is, however, the predominant result of
However, thyroid-stimulating hormone (TSH) glucocorticoid excess. Osteoblast and osteoclast
may play the primary role in osteoclast recruit- recruitment are affected, resulting in dysfunc-
ment here. Osteoclasts and their precursors tional remodeling and downgrading the main-
have been found to express the TSH receptor. A tenance of bone strength [116]. Mice treated
50% reduction in expression produced a pro- with corticosteroids have decreased bone
found osteoporosis in heterozygote TSH-recep- density, decreased serum osteocalcin, decreased
tor knockout mice [1]. Indeed, TSH itself cancellous bone area, and trabecular narrowing
inhibited osteoclast formation in vitro. TSH also [117]. These skeletal changes result in dimin-
appears to promote osteoblast differentiation, ished bone formation and turnover, along with
presumably directed via TSH receptors also impaired osteoblastogenesis and osteoclastoge-
demonstrated on these cells. Thus, in the hyper- nesis. The reduction in osteoblast number and
thyroid state, where TSH is severely suppressed, function appears to be due to increased
failure to stimulate the pre-osteoclast TSH- osteoblast apoptosis.
Clinical Disorders Associated with Alterations in Bone Resorption 113

Recently OBrien et al. showed that glucocor- long bones, and hearing loss from compression
ticoid action must be present specically and of cranial nerves. Ludwig van Beethoven is
locally in the skeleton [83]. Mice transgenic for thought to have suffered from Pagets disease
an osteoblast-specic 11-beta-hydroxyster- that led to his unfortunate deafness. Most
oid dehydrogenase that locally inactivates the typical Pagets patients, however, complain of
actions of glucocorticosteroids were resistant to pain from degenerative joint disease, which is
osteoblast apoptosis caused by pharmacologic exacerbated by mechanical factors due to
doses of steroids. Interestingly, the bone loss abnormal loading and weight bearing in the
was similar to that in non-transgenic animals, affected bones. In the last century, the incidence
i.e., osteoclast recruitment due to steroids of Pagets in the US population has been about
exogenous to the skeleton was unchecked, but 23%. There appears to be a decreasing inci-
bone formation rates were signicantly higher. dence, which correlates with a decreasing preva-
These results show that glucocorticoids have lence of measles infection in this country over
differential effects on osteoclast and osteoblast the last half of last century, thought to be the eti-
type cells, both of which contribute to the ologic agent, leaving the paramyxoviral signa-
osteoporotic phenotype consistent with ture in the pagetic osteoclasts [30].
hypercortisolism. There is a genetic predisposition for Pagets
disease. Forty percent of index cases have a rst-
degree affected relative [65]. Two genetic phe-
Environmental/Genetic notypes have been documented. The rst shows
linkage to the RANK protein (TNFSF11A) on
Pagets Disease (OMIM 602080) chromosome 18. Patients with an 18-bp inser-
Pagets disease is a primary disease of osteo- tion in the RANK sequence present with early
clasts, with a still controversial etiology best Pagets disease as teenagers, and this may repre-
ascribed to subtle interactions of genetic and sent a variation of familial expansile osteolysis
environmental factors. Clinically Pagets disease [65] (see below). This RANK mutation appears
manifests as a uni- or multifocal skeletal disease to lead to hyperresponsiveness to RANKL, with
that begins with an intense local bone resorp- increased size and nucleation of osteoclasts
tion followed by formation of woven bone. [93]. The other genetic phenotype is a mutation
Pagetic osteoclasts are increased in number, are in the sequestosome 1 gene (SQSTM1). The
hypernucleated and large (Figure 7.4), and, in sequestosome (p62) is an ubiquitin-binding
contradistinction to osteoclasts in unaffected protein that scaffolds signaling molecules in
bone in the same patient, are found to contain the TNF inammatory cascade. It is unclear
paramyxoviral-like inclusions. The clinical syn- whether Pagetic mutations change availability
drome can be silent simply a radiologist nota- of ubiquitin for intracellular proteins, perhaps
tion of bone sclerosis on a pelvic x-ray or can enhancing the inammatory signal cascade, or
be severe with pathologic fractures, bowing of affect viral handling within the cell [45].

Gorham-Stout Disease
Gorham-Stout disorder (GSD), or disappearing
bone disease, is a monocentric osteolysis, rst
described in 1955 [37]. (This non-Mendelian
disease should be differentiated from another
disease, cystic angiomatosis of bone, which also
is called Gorham Stout. That syndrome is inher-
ited as an autosomal dominant trait, and is asso-
ciated with osteosclerosis [94].) The massive
osteolysis of GSD is recognized as an aggres-
sive local resorption of bone [25]. There
is an angiomatous proliferation of thin-walled
vessels in the region of lytic bone, as well as an
Figure 7.4 Pagetic lesion: The micrograph shows a hyper- intracortical brosis [67]. The increased osteo-
nucleated pagetic osteoclast at the lower left of the field. clastogenesis is thought to be stimulated by
114 Bone Resorption

release of local factors. The extreme rarity of bone shows increased numbers of active osteo-
GSD has prevented much investigation of its clasts. It has long been known that myeloma
etiology. cells release factors that induce osteoclast for-
mation and activity, but identication of a spe-
Myeloma cic contributory factor has deed a great deal
of effort. Several publications have shown that
The majority of patients with multiple myeloma stimulates a coordinated increase in
myeloma, a B-cell neoplasm, eventually experi- bone stromal cell expression of RANKL with
ence extensive bony destruction that results in a concomitant reduction in osteoprotegerin,
hypercalcemia. Myeloma bone disease can leading to osteoclastogenesis [88]. Recombinant
present either as osteoporosis, or as discrete osteoprotegerin protein can prevent the bone
bony lesions that lead to pathologic fracture destruction caused by injection of human
through loss of skeletal integrity (Figure 7.5). melanoma cells into mice [22], indicating at
Bisphosphonate therapy to repress bone resorp- least a secondary role for the RANKL/RANK/
tion has lately become a cornerstone of OPG axis in the rampant bone lesions of
myeloma chemotherapy. The lesion of myeloma myeloma.
In human disease, elevated serum levels of
RANKL along with decreased osteoprotegerin
levels have been noted, suggesting that myeloma
cells may even secrete this protein [35, 98].
There may be a role, as well, for the DKK1
protein, which is secreted by myeloma cells and
prevents Wnt signaling in osteoblasts the Wnt
pathway in osteoblasts. Elevated DKK1 levels in
bone marrow plasma and in peripheral blood
from myeloma patients are associated with focal
bone lesions, but this has yet to be linked to
osteoclast recruitment or activation [108].

Inherited Disorders of Osteoclasts


Juvenile Pagets Disease (OMIM 239000)
The skeletal lesions of juvenile Pagets disease,
or hyperostosis corticalis deformans juvenilis,
as described by Bakwin et al. [4], resemble adult
Pagets disease with increased skull size, deaf-
ness, and expanded and bowed long bones. This
rare autosomal recessive disease presents in
early childhood with musculoskeletal pain.
Affected bones have coarse trabeculations and
areas of sclerosis interspersed with osteoporo-
sis. This results in decreased mechanical
strength with deformity and increased sus-
ceptibility to fracture [16]. Examination of
osteoclasts from these patients does not
reveal paramyxoviral-like inclusions, and circu-
lating monocytes are clear of viral transcripts
[119].
The continuous and intense bone remodeling
Figure 7.5 Myeloma lytic bone lesions: Myeloma bone in juvenile Pagets disease, conrmed by ele-
disease can present as either diffuse osteoporosis, or as scat- vated markers of bone turnover, suggests abnor-
tered lytic lesions, as shown in the radiograph. mal activation of osteoclasts. Whyte et al.
Clinical Disorders Associated with Alterations in Bone Resorption 115

recently linked the disease to a mutation in pared to the 100 kb deletion in the Navajo family
osteoprotegerin [118]. Using a candidate-gene [118], resulted in the loss of an aspartate
approach, they considered genes involved in residue, but their osteoprotegerin levels were
osteoclast activation. They rst found the gene measurable. The mutant osteoprotegerin,
locus for RANK to be normal. Because the however, was ineffective in suppressing bone
osteoprotegerin-knockout mouse has acceler- resorption.
ated bone remodeling [14], the group focused The mouse osteoprotegerin knockout model
their search on osteoprotegerin. Both unrelated replicates the human disease [14]. The skeletal
Navajo patients were found to have a null muta- lesion is present early, with a decrease in total
tion in osteoprotegerin with an identical break- bone density and fracture. Interestingly the
point in chromosome 8; their osteoprotegerin transgenic mice also exhibit medial calcica-
levels were not measurable (Figure 7.6). tion of the medium and large arteries. This
A second osteoprotegerin mutation was raises the possibility that pathologic calcica-
described in an Iraqi family where three siblings tion of the vasculature involves interplay
were affected with Juvenile Pagets Disease [23]. between cells capable of both generating and
The in-frame deletion of three base pairs, com- resorbing mineral in this unusual location.

Figure 7.6 Juvenile Pagets disease: Radiographs of a 1-year-old patient from Whyte et al. [121] are shown: the left is the probands
right proximal femur with markedly widened, poorly modeled, and osteopenic bone, with cortical thinning and a coarse trabecular
pattern. The radiograph on the right was obtained 27 months after the initiation of calcitonin therapy, demonstrating improvement
of bone structure. Copyright 2002 Massachusetts Medical Society. All rights reserved.
116 Bone Resorption

Familial Expansile Osteolysis (FEO) Japan [81], but now has been reported globally.
(OMIM 174810) This condition, also called Nasu-Hakola disease,
presents in midlife with pain after muscle strain
Also known as polyostotic osteolytic dysplasia, has led to the diagnosis of cysts in the epiphy-
familial expansile osteolysis rst was described ses of long bones. The bone cysts are composed
in 1988 in a family from Northern Ireland [84]. of dysmorphic fatty and collagenous connec-
This rare autosomal dominant dysplasia shares tive tissue. Osteoporotic features are preval-
a focal nature with Pagets disease but appears ent. Patients in the fourth decade experience
in the teenage decade. In the focal lesions, os- neurological symptoms, which, unfortunately,
teoclast activity is associated with medullary progress to presenile dementia. The genetic eti-
expansion and painful deformity. The focal ology is a loss-of-function mutation in the gene
lesions, which frequently lead to fracture, are encoding the TYRO protein tyrosine kinase-
predominantly peripheral. Deafness due to skull binding protein called DAP12, or in some cases
dysplasia can be present before puberty. a mutation in the TREM2 adaptor [87, 89].
Linkage studies have shown that the respon- DAP12 is a transmembrane adaptor protein that
sible gene in the Irish FEO family was located associates with the cell surface receptor TREM
on chromosome 18 and was due to a mutation to form the DAP12-TREM2 complex on natural
in the signal peptide region of the receptor killer T cells and myeloid cells. The ligand for
activator of nuclear factor-B (RANK) [47]. TREM2 is unknown.
Expressing the recombinant mutant RANK in Paloneva et al. showed that peripheral blood
culture showed the mutant protein was resistant mononuclear cells from PLOSL patients that are
to cleavage of the signal peptide; the immature decient in DAP12 and TREM2 respond poorly
protein causes perturbations in RANK level and to osteoclastogenic stimuli. This results in a
results in a total increase in RANK signaling. reduced ability to resorb bone in vitro [86].Acti-
Thus, FEO is caused by an activating mutation vation of the DAP12/TREM2 complex appears
of RANK, leading to hyperactivation of osteo- to be a required co-stimulatory signal during
clasts. Why the disease is focal and peripheral RANKL-induced osteoclast differentiation [58].
remains unclear. This recent work has suggested that the intra-
Another expansile disease that was only cellular calcium signal generated by DAP12/
described in 2000, and is phenotypically distinct TREM2 is necessary for adequate NFAT1 sig-
from FEO, is expansile skeletal hyperphos- nalling (see Chapter 1); indeed, transgenic
phatasia (ESH) [120]. The cases were a mother knockout of the functional DAP12/TREM2 co-
and daughter who presented with expanding stimulatory receptor resulted in mice with
hyperostotic long bones, pain, and early onset severe osteopetrosis. Interestingly, the evolving
deafness. The skeletal lesions, which appear in understanding in the role of the DAP12/TREM2
the appendicular skeleton, are reminiscent in pathway in osteoclastogenesis came to light only
their distribution of Pagets disease, rather than through investigation of the skeletal lesion
of FEO. Measles virus gene transcripts are not associated with this rare genetic disorder.
present in cells of monocytic lineage. Whyte
et al. identied a mutation in these patients that
was identical to the 18-base pair tandem dupli- Rett Syndrome (OMIM 312750)
cation in the RANK signal peptide sequence Rett syndrome is an X-linked dominant disor-
described in the FEO families [118]. The phe- der recognized in the latter half of last century.
notypic variation between ESH and FEO sug- Its major problem is an unrelenting progressive
gests other genetic or epigenetic factors have a encephalopathy that causes severe dementia
role in remodeling dysplasias. in girls [42]. Growth retardation is associated
with a decreased metabolic rate [80]. There are
Polycystic lipomembranous osteodysplasia metatarsal and metacarpal abnormalities in
with sclerosing leukoencephalopathy some patients with Rett syndrome [69], as well
as a severe osteoporosis [15]. The cause of the
(PLOSL) (OMIM 221770) osteoporosis is, as yet, unknown, and may be
Polycystic lipomembranous osteodysplasia with due to problems with osteoclasts, osteoblasts, or
sclerosing leukoencephalopathy was recognized coupling. We include Rett syndrome in this
almost simultaneously in Finland [43] and in chapter because the bone lesion, associated with
Clinical Disorders Associated with Alterations in Bone Resorption 117

troublesome fracturing, may cause unregulated rescues insufcient bone resorption [28]. In
osteoclast activity. the last ten years, knowledge of spontaneous
Genetic nonsense and missense mutations and engineered genetic mutations has greatly
are found in the gene encoding methyl-CpG- increased understanding of osteopetrotic eti-
binding protein-2 (Mecp2) [115]. All of the ologies, although the cause of many human
nucleotide substitutions in Rett syndrome osteopetroses has not, as yet, been charted.
involve C to T transitions at CpG hot spots. The phenotype of osteopetrotic disease spans
Transgenic mice expressing the truncated that of completely marbled bones, with a lethal
Mecp2 Rett protein showed a similar pheno- defect of hematopoiesis due to the total absence
typic neurologic disease, as well as a severe of a marrow space, to diseases that can be con-
kyphosis, but the skeleton was not specically sidered advantageous, insofar as the increase
examined [99]. The authors of this study suggest in bone mineral promotes a fracture resistant
that histone H3 is hyperacetylated in this model, skeleton. The most severe forms of osteopetro-
possibly leading to abnormal gene expression; sis present early in life with skeletal sclerosis
however, the effect on bone resorption is not yet that obliterates the bone marrow compartment
understood. and impinges on cranial nerves. Extramedullary
hematopoiesis with hepatosplenomegaly rarely
rescues the marrow insufciency; pancytopenia
leads to infection, inanition and death. More
Decreased Osteoclast Activity mild forms of osteopetrosis, or osteosclerosis,
Resulting in Osteopetrosis can have cranial nerve pathology as well as
increased fracture risk. It is likely that the
mildest forms of osteoclast dysfunction evade
Osteopetrosis is a heterogeneous group of con- identication because pathologic ndings are
ditions where an impairment in bone resorp- necessary to initiate diagnosis. Sclerotic bone
tion leads to accrual of bone mass (Figure 7.7). disease is occasionally noted in other systemic
Classic osteopetrosis is considered a disease of diseases, which may result from changes in
osteoclasts. Walker showed that transplanting osteoclast function. Because information
the gray-lethal (malignant) osteopetrotic mouse regarding these silent disorders is lacking, we
with the bone marrow and spleen of normal lit- will cover only inherited forms of osteopetrosis
termates cured the osteopetrosis [114]. Indeed, that lead to clinical disease.
current medical therapy uses marrow trans-
plantation for the classic malignant osteope-
trosis: implantation of hematopoietic stem Inherited Diseases Resulting in
cells containing normal osteoclast precursors Dysfunctional Osteoclasts
Osteopetrosis, Malignant (OMIM 259700)
The malignant osteopetroses are autosomal
recessive diseases that present in early infancy
with macrocephaly, progressive deafness and
blindness, extramedullary hematopoiesis, and
pancytopenia. It is likely that genetic mutations
of many osteoclast functions will be identied
in human disease, but so far, mutations in only
three proteins that are necessary for osteoclast
function have been indexed. These include the
osteoclast vacuolar proton pump, the chloride
channel-7, and the gray-lethal protein, all nec-
essary for osteoclast function.
Figure 7.7 The weight of the osteopetrotic skull: A skull from Interestingly, the rst mutation identied as
a van Buchem patient is placed in the right pan of the scale. an etiology for the murine osteopetrotic pheno-
Figure is from [52], Janssens and Van Hul, Molecular genetics of type op/op, is not associated with human
too much bone. Hum Mol Genet 2002; 11:23852393, by per- osteopetrosis. The op/op mouse has a deletion
mission of Oxford University Press. in the gene for macrophage colony stimulating
118 Bone Resorption

factor, or MCSF, which results in an essentially is fully penetrant and lethal, but can be rescued
nonfunctioning protein [122]. In the absence of with bone marrow transplantation [113]. The
MCSF, hematopoietic stem cells cannot enter the murine gene was cloned and shown to be
macrophage lineage in the developing mouse, responsible for the osteopetrosis [19]. The GL
and monocytes, macrophages and osteoclasts protein function is required for osteoclast and
are not formed. This leads to lack of tooth erup- melanocyte function, but the protein itself
tion and the inability to wean. The op/op mouse (OMIM 607649) requires further study. One
dies because of its inability to wean if the human case has been identied as a GL
newborn is nursed beyond 2 weeks, osteoclasts mutation [19].
appear and the mouse lives. The rescue is
thought to be due to the ability of secreted vas- Osteopetrosis, Autosomal Dominant,
cular endothelial growth factor (VEGF) to
support macrophage/osteoclast differentiation
Types I and II
and survival [82]. A human analogue of this The autosomal dominant osteopetroses are
disease may thus be silent since infants do not not lethal disorders, but more mild forms due
eat solid food for many months. to impaired osteoclastic bone resorption. The
grouping is heterogeneous: the conditions in
many families are associated with an asymp-
Mutations in the Osteoclast tomatic osteosclerosis. The mild forms of
Vacuolar Proton Pump osteopetrosis can cause bone pain and hearing
(see also Chapter 3) loss, but affected individuals are less susceptible
to fracture than persons aficted with other
The osteoclast proton pump is a vacuolar forms of osteopetrosis. Most interestingly, some
ATPase [109]. This large molecule has 28 sub- forms of osteopetrosis are linked to protection
units encoded by 13 separate genes that vary from fracture, as is seen in lesions associated
in tissue expression. The osteoclast specic with activating mutations of the LRP5 signal
subunit, TCIRG1, is a 116-kd subunit [71]. In one cascade.
series, more than 50% of patients with infantile As knowledge of murine and human bone
malignant osteopetrosis had mutations in resorption and structure increases, it is likely
TCIRG1 [29]. The mutations are extremely vari- that more mutations will be assigned to this
able but uniformly lead to null expression of the disease category in the future. For instance, high
proton pump in osteoclasts [60, 105]. Without bone density traits have been described in
secretion of acid, osteoclasts cannot resorb several inbred mouse strains [6]. The high bone
bone. density in one of these models is linked to a
region on mouse chromosome 11, and is due to
a mutation in the 12/15-lipoxygenase enzyme
Mutations in the Osteoclast Chloride [57]. Pharmacologic inhibition of the 12/15-
Channel-7 lipoxygenase improved both bone density and
strength in ovariectomized mice. It is not
The process of proton secretion requires that unlikely that mutations that affect the activity of
the osteoclast also have a parallel conductance the analogous pathway in humans will be asso-
of a negative ion, i.e., a specic chloride channel. ciated with increased bone density either
In osteoclasts the CLCN7 gene encodes this through osteoblast controlled restriction of
channel. Disruption of the channel leads to a osteoclast function or through direct effects on
severe osteopetrosis in mice because osteoclasts osteoclast recruitment and activity.
were cannot secrete acid and bone is not It has been suggested that within this hetero-
resorbed. The same defect has also been identi- geneous grouping there are two distinct radi-
ed in a patient [59]. ographic types representing the generalized
osteosclerosis [3]. Type I differs mainly from
Mutation of the Gray-Lethal Gene type II in that the increased bone appears to
confer fracture protection. We have adhered to
The spontaneous GL, or gray-lethal, mutation in the designations of type currently suggested in
mice causes a gray coat color and a severe auto- the literature that also show some differences in
somal recessive osteopetrosis. The osteopetrosis serum phosphate (lower in type I) and serum
Clinical Disorders Associated with Alterations in Bone Resorption 119

acid phosphatase (markedly increased in type comitant increases in osteoclast activity. Thus,
II) [10]. even though the high bone density of this
disease may stem from hyperactive osteoblasts,
osteoclasts also are affected.
Osteopetrosis, Autosomal A Belgian family with previously docu-
Dominant, Type I (OMIM 607634) mented type I osteopetrosis was recently shown
to have a transversion in the same codon of the
Type I osteopetrosis is characterized by pro- LRP5 gene that caused the high-bone-density
nounced sclerosis of the cranial vault with little traits described in the United States [110]. A
increase in vertebral bone. Interestingly, type I Sardinian family assigned a diagnosis of van
confers protection from fracture risk. One form Buchem disease with osteosclerosis of the skull
of this disease can be caused by mutations in the was investigated for mutations in LRP5. Indeed,
low density lipoprotein receptor-related protein a novel mutation was found that was also
5 (LRP5) gene (OMIM 603506). The rst citation located in the amino-terminal region of the
was from a research group searching for high protein [110]. (The skull shown in Figure 7.7
bone mass traits. They investigated a family belonged to a patient affected with van Buchem
with high bone density that had a heterozygous disease (OMIM 607636)). Van Wesenbeck, in
G-to-T transversion in exon 3 of the LRP5 gene. reviewing six novel missense mutations in the
This mutation resulted in a gly171-to-val change LRP5 gene, suggested that conditions associated
in a predicted beta-propeller module of the with increased bone density affecting mainly
LRP5 protein that was observed in all affected the cortices of long bones and skull are caused
individuals [72]. The same mutation was found by mutations in this gene [110]. There is a phe-
in a second kindred with high bone density, notypic pleiomorphism, however, with some
wide mandibles, and torus palatinus (Figure families having mandible changes and torus
7.8) [12]. Interestingly this family, who did not palatinus, while others have none. One of the
have any symptoms, did note a difculty in areas that will need attention in the future is the
staying aoat while swimming! classication of those diseases resulting in
The discovery of the mutation in LRP5 and its endosteal hyperosteosis: are they the result of
previously unrecognized relationship to bone impaired osteoclast activity, or simply of exces-
remodeling set off a urry of investigations. sive bone formation?
Members of the low-density lipoprotein recep-
tor (LDLR) family are cell surface proteins that Osteopetrosis, Autosomal Dominant
bind and internalize ligands in the process of
receptor-mediated endocytosis. It would appear Type II: Albers-Schnberg Disease
that the LRP5 pathway through Wnt signaling is (OMIM 166600)
functional in osteoblasts, directing osteoblast
bone formation. It was quickly noted that Autosomal dominant osteopetrosis type II is
another mutation in this protein was associa- characterized by sclerosis, predominantly in-
ted with an osteopetrotic disease, osteopo- volving the vertebrae, the iliac wings, and the
rosis-pseudoglioma syndrome (OPPG, OMIM base of the skull. In contrast to type I patients,
259770). Mutations in LRP5 cause the autosomal type II patients do have increased fracture
recessive disorder, and indeed, heterozygous frequency [11]. Bone resorption is defective,
carriers of OPPG have reduced bone mass com- while bone formation is normal; this is reected
pared to age and gender-matched controls [36]. in elevated serum levels of tartrate-resistant
A secreted mutant form of LRP5 appears to acid phosphatase (TRAP) [112]. Why TRAP,
reduce bone apposition [36]. Mice with a tar- which represents osteoclast activity, rises, is
geted disruption of LRP5 have low bone density, unclear: perhaps it represents an attempt by
associated with decreased osteoblast prolifera- the dysfunctional osteoclast in this disease to
tion [54]. It now is accepted that LRP5 is increase activity. The elevations in TRAP are
required for optimal Wnt signaling in helpful in making the diagnosis in families
osteoblasts. The question of how LRP5 affects where osteopetrosis type II shows incomplete
osteoclasts is now open for discussion. The penetrance: gene carriers have TRAP values
increased osteoblast activity associated with the that are not different from those of controls
LRP5 mutations is not accompanied by con- [112].
120 Bone Resorption

A B C

D E F

Figure 7.8 Osteosclerosis due to LRP5: Photographs of an affected member at the ages of 12 years (A) and 45 years (B) show the
development of the wide, deep mandible that was characteristic of all affected members of the kindred. A large, lobulated torus
palatinus in an affected member (C, arrow) was also characteristic of all affected kindred members. Characteristic radiographic find-
ings included an abnormally thick mandibular ramus (arrow, D); a markedly thickened cortex and narrowed medullary cavity (arrow)
in the femur (E), which was otherwise normal; and dense but otherwise normal-appearing vertebrae (F). Figure used with permis-
sion from Boyden et al. [12]. Copyright 2002 Massachusetts Medical Society. All rights reserved.

Benichou et al., studying 42 patients with osteomyelitis after hip replacement. Increased
radiological evidence of type II disease, includ- surgical complications also were experienced by
ing the classic sandwich vertebra appearance those 50% of patients who required orthopaedic
(or rugger jersey spine) found that nearly 80% intervention.
of patients had fractures [8]. The fractures were The Benichou research team went on to
slow to heal. Hip osteoarthritis was common, perform a genome wide linkage analysis of an
and severe with an increased incidence of extended French family, localizing the disease to
Clinical Disorders Associated with Alterations in Bone Resorption 121

chromosome 16 [7]. The candidate region was missense, and stop codon mutations in the gene
then studied in 12 autosomal dominant type II encoding cathepsin K were subsequently found
families, and found to contain the CLCN7 gene in patients with pycnodysostosis [34]. Indeed,
with at least seven different mutations [21]. It in two siblings with the disease, cathepsin K
thus appears that this not-so-benign autosomal protein was entirely absent, while the parents,
dominant osteopetrosis is allelic with the who had no trace of disease, expressed half-
CLCN7 mutation that causes the autosomal normal levels of cathepsin K.
recessive malignant osteopetrosis. Although not Cathepsin K was rst described as cathepsin
yet proven, it is reasonable to infer that complete O [101] and is expressed at its highest levels in
absence of chloride transport would cause musculoskeletal tissues, with the strongest in
malignant osteopetrosis, and that the defect in situ signal in osteoclasts [92]. This protease has
chloride transport in type II benign osteopet- potent endoprotease activity against brinogen
rosis must be incomplete. at acid pH, and therefore is effective in de-
grading extracellular bone matrix under the
osteoclast resorption tent. Pycnodysostotic
Pycnodysostosis (OMIM 265800) osteoclasts are normal in numbers and in mor-
Pycnodysostosis is an autosomal recessive phology (normal rufed borders and clear
osteochondrodysplasia characterized by osteo- zones) and indeed resorb demineralized bone.
sclerosis and short stature, as well as deformity The problem occurs in degrading the organic
of the skull, maxilla and phalanges, osteo- matrix.
sclerosis, and bone fragility. The condition was Targeted mutation of the cathepsin K in the
rst described by Marotaux and Lamy who mouse results in a phenotype consistent with
also suggested that Toulouse-Lautre (1864 the human disease, showing increased bone
1901) had the disease [74]. The features of density and deformities [38, 64]. Both mice and
pycnodysostosis are shown in Figure 7.9, from human phenotypes become more pronounced
[44]. Gelb et al., on the basis of linkage analysis with age and tend to affect bones that have
in a large inbred Arab family, concluded that the higher rates of remodeling.
affected gene was located on chromosome 1q21
[33]. The gene cathepsin K, a cysteine protease Renal Tubular Acidosis and Carbonic
that is predominantly expressed in osteoclasts,
is located in this region. This discovery spurred
Anhydrase II Deficiency (OMIM 259730)
a search for mutations in this gene. Nonsense, The syndrome of autosomal recessive osteo-
porosis associated with renal tubular acidosis
was rst recognized in 1972 [103]. The disease
generally presents in the rst two years of life
with fracture, but allows for long survival since
there are few hematologic disturbances other
than a mild anemia. Other features include short
stature, mental retardation, dental malocclu-
sion, and visual impairment resulting from
optic nerve compression. Electrolyte changes
that indicate a hybrid of mild proximal and
prominent distal tubular disturbances are a sign
of a mild renal tubular acidosis.
The carbonic anhydrases (CAs) are a family
of zinc metalloenzymes that, when blocked by
sulfonamide inhibitors, produce renal tubular
acidosis and PTH-induced release of calcium
Figure 7.9 Pycnodysostosis: The profile shows a pycnodysos-
totic 9-year-old boy witha height ageof 5 years.Note the promi-
from bone. Sly et al. [104] postulated that the
nent forehead and micrognathia. The x-ray shows the generally renal tubular defects were due to an abnormal-
dense bone and partial loss of several distal phalanges. Figure ity in carbonic anhydrase II because the disease
reproduced from Ho et al. [44], J Bone Miner Res. 1999; 14: affected both kidney and brain: CAII is the only
16491653, with permission of the American Society for Bone CA present in both tissues (although the
and Mineral Research. etiology of cerebral calcications remains
122 Bone Resorption

unknown). CAII was shown to be absent in ery- deafness, and facial paralysis. Radiologically
throcytes of affected patients [102]. Study of 18 there is thickening of the diaphyseal cortex of
unrelated patients revealed a virtual absence the long bones and narrowing of the medullary
of CA II in erythrocyte hemolysates [104]. canal with sclerosis at the skull base. Kinoshita
Reduced CA II levels were found in obligate et al. assigned the disease locus to a region
heterozygotes. where human transforming growth factor-1
Carbonic anhydrase II is expressed at high gene (TGFB1) is located [56]. TGF-b1 is secreted
levels in active osteoclasts [61]. The disease has as a large precursor protein that requires pro-
been treated with marrow transplantation. cessing to cleave the signal peptide and dimer-
McMahon et al. [77] reported, in a fairly severely ization. The processed precursor is cleaved into
affected Irish kindred, that three years after a mature TGF-b1 form with a latency-associated
transplantation there was histologic and radio- protein that, while linked to the mature growth
logical resolution of osteopetrosis with stabi- factor, keeps it inactive by masking TGF-b1
lization of hearing and vision; the transplanted receptor-binding domains [51]. Activation of
children remained developmentally delayed latent TGF-b1 can be accomplished in vitro
with beginnings of cerebral calcication and through heat or chaotropic agents [90]. Three
continued to have renal tubular acidosis. Thus different missense mutations causing CED were
bone marrow stem cell replacement cures the expressed in vitro; all were shown to disrupt the
osteoclast component of CAII deciency, but association of latency-associated protein with
has little effect on the loss of CAII function in TGF-b1, affecting release of the mature TGF-b1
renal and cerebral tissues. [97].
A transgenic mouse model where a mutation Osteoclast formation from peripheral blood
in CA II was created with N-ethyl-N-nitrosourea mononuclear cells of CED patients was en-
produced homozygotes that are runted and have hanced in vitro [76]; the activation of osteo-
renal tubular acidosis, but interestingly do not clast activity is consistent with clinical reports
have osteopetrosis [70]. There is a great deal of that show biochemical evidence of increased
phenotypic heterogeneity in the human disease. bone resorption. The increase in osteoclastoge-
Most kindreds are found in the Mediterranean nesis was inhibited by a soluble TGF receptor.
and in the Middle East; the latter families have Total serum TGF-b1 levels were similar in
the greatest degree of mental retardation and affected and unaffected subjects, but concentra-
metabolic acidosis, but less conspicuous frac- tions of active TGF-b1 in conditioned medium
tures, while other kindred have only mild of osteoclast cultures were higher in the three
mental retardation with severe osteopetrotic CED patients than in the unaffected family
fracturing [104]. In one family the affected indi- member. Interestingly, when exposed to condi-
viduals are compound heterozygotes, each tioned media from CED broblasts, broblast
inheriting a different mutation from mother growth is suppressed, while osteoblastic cells
and father [96]. The maternal CA II mutation is are growth accelerated [97]. Indeed, the muscle
a missense mutation in exon 3 that substitutes a and skeletal pathophysiology in Camurati
tyrosine for a histidine. As a result mature Engelmann disease is likely due to differential
enzyme activity is reduced, but not absent. The effects of the TGF activation and response in
authors suggest that expression of the remnant muscle and skeleton. In the skeleton, the effects
enzyme activity has prevented mental retarda- of the CED TGF mutation lead to activation of
tion in this family. osteoclasts, with a concurrent increase in
osteoblast markers due to coupling; the increase
Camurati-Engelmann Disease in osteoblast activity is also likely a direct effect
of TGF-b1 on the osteoblast lineage.
(OMIM 131300)
CamuratiEngelmann disease (CED) is an auto-
somal dominant, progressive diaphyseal dyspla-
sia characterized by hyperosteosis and sclerosis
Conclusions
of the diaphyses of long bones. CED patients
have severe leg pains, muscular weakness, fati- During the last 15 years, understanding of the
gability, and decreased muscle mass. Other molecular signals involved in osteoclast recruit-
symptoms are exophthalmos, decreased vision, ment, the pathways necessary for adequate
Clinical Disorders Associated with Alterations in Bone Resorption 123

OC differentiation Osteoblast control of OC


Pagets Disease (p62, RANK) Juvenile Pagets Disease (OPG)
Nasu-Hakola Disease (DAP12/TREM) Osteopetrosis, AD Type I (LRP5)
Rett Syndrome (MECP2) Hyperparathyroidism (RANKL)
Camurati-Engelmann (TGFb)

OC function
Malignant osteopetrosis (H+ pump, ClC-7)
Osteopetrosis, AD Type II (ClC-7 )
Pycnodysostosis (CTSK)
Osteopetrosis with RTA (CAII)
Grey-lethal (?)

Figure 7.10 Clinical diseases of bone resorption. Specific diseases discussed in this chapter are also delineated in Tables I & II.

osteoclast function, and continued investigation mineral density responses to the same high-impact
of skeletal pathophysiology, have enlightened exercise. J Bone Miner Res 13:180513.
6. Beamer WG, Donahue LR, Rosen CJ, Baylink DJ (1996)
our understanding of both common and rare Genetic variability in adult bone density among inbred
bone diseases. Figure 7.10 presents the known strains of mice. Bone 18:397403.
diseases. Further basic and clinical research will 7. Benichou O, Cleiren E, Gram J, Bollerslev J, de
surely lead to new disease designations that Vernejoul MC, Van Hul W (2001) Mapping of autoso-
mal dominant osteopetrosis type II (Albers-Schonberg
involve altered remodeling. In turn, new nd- disease) to chromosome 16p13.3. Am J Hum Genet
ings are likely to revise understanding of the 69:64754.
diseases discussed here. 8. Benichou OD, Laredo JD, de Vernejoul MC (2000) Type
II autosomal dominant osteopetrosis (Albers-
Schonberg disease): clinical and radiological manifes-
tations in 42 patients. Bone 26:8793.
References 9. Bilezikian JP, Silverberg SJ (2004) Clinical practice.
Asymptomatic primary hyperparathyroidism. N Engl
J Med 350:174651.
1. Abe E, Marians RC, Yu W, Wu XB, Ando T, Li Y, et al. 10. Bollerslev J, Andersen PE, Jr. (1988) Radiological, bio-
(2003) TSH is a negative regulator of skeletal remodel- chemical and hereditary evidence of two types of auto-
ing. Cell 115:15162. somal dominant osteopetrosis. Bone 9:713.
2. Adams JS, Wahl TO, Lukert BP (1981) Effects of 11. Bollerslev J, Mosekilde L (1993) Autosomal dominant
hydrochlorothiazide and dietary sodium restriction on osteopetrosis. Clin Orthop. 4551.
calcium metabolism in corticosteroid treated patients. 12. Boyden LM, Mao J, Belsky J, Mitzner L, Farhi A, Mitnick
Metabolism 30:21721. MA, et al. (2002) High bone density due to a mutation
3. Andersen PE, Jr., Bollerslev J (1987) Heterogeneity of in LDL-receptor-related protein 5. N Engl J Med 346:
autosomal dominant osteopetrosis. Radiology 164: 151321.
2235. 13. Brown EM, Gamba G, Riccardi D, Lombardi M, Butters
4. Bakwin H, Golden A, Fox S (1964) Familial R, Kifor O, et al. (1993) Cloning and characterization
Osteoectasia with Macrocranium. Am J Roentgenol of an extracellular Ca(2+)-sensing receptor from
Radium Ther Nucl Med 91:60917. bovine parathyroid. Nature 366:57580.
5. Bassey EJ, Rothwell MC, Littlewood JJ, Pye DW (1998) 14. Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J,
Pre- and postmenopausal women have different bone Capparelli C, et al. (1998) Osteoprotegerin-decient
124 Bone Resorption

mice develop early onset osteoporosis and arterial cal- 31. Gagel RF (1997) Multiple endocrine neoplasia type II
cication. Genes Dev 12:12608. and familial medullary thyroid carcinoma. Impact of
15. Budden SS, Gunness ME (2003) Possible mechanisms genetic screening on management. Cancer Treat Res
of osteopenia in Rett syndrome: bone histomorpho- 89:42141.
metric studies. J Child Neurol 18:698702. 32. Garnero P, Sornay-Rendu E, Chapuy MC, Delmas PD
16. Caffey J (1972) Familial hyperphosphatasemia with (1996) Increased bone turnover in late post-
ateliosis and hypermetabolism of growing membra- menopausal women is a major determinant of osteo-
nous bone; review of the clinical, radiographic and porosis. J Bone Miner Res 11:33749.
chemical features. Bull Hosp Joint Dis 33:81110. 33. Gelb BD, Edelson JG, Desnick RJ (1995) Linkage of
17. Canalis E (1996) Clinical review 83: Mechanisms of pycnodysostosis to chromosome 1q21 by homozygos-
glucocorticoid action in bone: implications to gluco- ity mapping. Nat Genet 10:2357.
corticoid-induced osteoporosis. J Clin Endocrinol 34. Gelb BD, Shi GP, Chapman HA, Desnick RJ (1996)
Metab 81:34417. Pycnodysostosis, a lysosomal disease caused by
18. Cenci S, Weitzmann MN, Roggia C, Namba N, Novack cathepsin K deciency. Science 273:12368.
D, Woodring J, et al. (2000) Estrogen deciency induces 35. Glass DA, 2nd, Patel MS, Karsenty G (2003) A new
bone loss by enhancing T-cell production of TNF- insight into the formation of osteolytic lesions in mul-
alpha. J Clin Invest 106:122937. tiple myeloma. N Engl J Med 349:247980.
19. Chalhoub N, Benachenhou N, Rajapurohitam V, Pata 36. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S,
M, Ferron M, Frattini A, et al. (2003) Grey-lethal muta- Reginato AM, et al. (2001) LDL Receptor-Related
tion induces severe malignant autosomal recessive Protein 5 (LRP5) Affects Bone Accrual and Eye
osteopetrosis in mouse and human. Nat Med 9:399 Development. Cell 107:51323.
406. 37. Gorham LW, Stout AP (1955) Massive osteolysis (acute
20. Chandrasekharappa SC, Guru SC, Manickam P, spontaneous absorption of bone, phantom bone, dis-
Olufemi SE, Collins FS, Emmert-Buck MR, et al. (1997) appearing bone); its relation to hemangiomatosis.
Positional cloning of the gene for multiple endocrine J Bone Joint Surg Am 37-A:9851004.
neoplasia-type 1. Science 276:4047. 38. Gowen M, Lazner F, Dodds R, Kapadia R, Feild J,
21. Cleiren E, Benichou O, Van Hul E, Gram J, Bollerslev J, Tavaria M, et al. (1999) Cathepsin K knockout mice
Singer FR, et al. (2001) Albers-Schonberg disease develop osteopetrosis due to a decit in matrix degra-
(autosomal dominant osteopetrosis, type II) results dation but not demineralization. J Bone Miner Res
from mutations in the ClCN7 chloride channel gene. 14:165463.
Hum Mol Genet 10:28617. 39. Greeneld EM, Bi Y, Miyauchi A (1999) Regulation of
22. Croucher PI, Shipman CM, Lippitt J, Perry M, Asosingh osteoclast activity. Life Sci 65:1087102.
K, Hijzen A, et al. (2001) Osteoprotegerin inhibits the 40. Greenspan SL, Greenspan FS (1999) The effect of
development of osteolytic bone disease in multiple thyroid hormone on skeletal integrity. Ann Intern Med
myeloma. Blood 98:353440. 130:7508.
23. Cundy T, Hegde M, Naot D, Chong B, King A, Wallace 41. Gronowicz G, McCarthy MB, Raisz LG (1990)
R, et al. (2002) A mutation in the gene TNFRSF11B Glucocorticoids stimulate resorption in fetal rat pari-
encoding osteoprotegerin causes an idiopathic hyper- etal bones in vitro. J Bone Miner Res 5:122330.
phosphatasia phenotype. Hum Mol Genet 11:211927. 42. Hagberg B (1995) Rett syndrome: clinical peculiarities
24. Cushing H (1932) The basophil adenomas of the pitu- and biological mysteries. Acta Paediatr 84:9716.
itary body and their clinical manifestation. Johns 43. Hakola HP, Iivanainen M (1973) A new hereditary
Hopkins Bull 50:13760. disease with progressive dementia and polycystic
25. Devlin RD, Bone HG, 3rd, Roodman GD (1996) Inter- osteodysplasia: neuroradiological analysis of seven
leukin-6: a potential mediator of the massive osteoly- cases. Neuroradiology 6:1628.
sis in patients with Gorham-Stout disease. J Clin 44. Ho N, Punturieri A, Wilkin D, Szabo J, Johnson M,
Endocrinol Metab 81:18937. Whaley J, et al. (1999) Mutations of CTSK result in pyc-
26. Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, nodysostosis via a reduction in cathepsin K protein.
Lacey DL, Riggs BL (2003) Role of RANK ligand in J Bone Miner Res 14:164953.
mediating increased bone resorption in early post- 45. Hocking LJ, Lucas GJ, Daroszewska A, Mangion J,
menopausal women. J Clin Invest 111:122130. Olavesen M, Cundy T, et al. (2002) Domain-specic
27. Ehrlich PJ, Lanyon LE (2002) Mechanical strain and mutations in sequestosome 1 (SQSTM1) cause familial
bone cell function: a review. Osteoporos Int 13:688 and sporadic Pagets disease. Hum Mol Genet
700. 11:27359.
28. Fischer A, Griscelli C, Friedrich W, Kubanek B, 46. Hofbauer LC, Khosla S, Dunstan CR, Lacey DL,
Levinsky R, Morgan G, et al. (1986) Bone-marrow Spelsberg TC, Riggs BL (1999) Estrogen stimulates
transplantation for immunodeciencies and osteopet- gene expression and protein production of osteopro-
rosis: European survey, 19681985. Lancet 2:10804. tegerin in human osteoblastic cells. Endocrinology
29. Frattini A, Orchard PJ, Sobacchi C, Giliani S, Abinun M, 140:436770.
Mattsson JP, et al. (2000) Defects in TCIRG1 subunit of 47. Hughes AE, Ralston SH, Marken J, Bell C, MacPherson
the vacuolar proton pump are responsible for a subset H, Wallace RG, et al. (2000) Mutations in TNFRSF11A,
of human autosomal recessive osteopetrosis. Nat Genet affecting the signal peptide of RANK, cause familial
25:3436. expansile osteolysis. Nat Genet 24:458.
30. Friedrichs WE, Reddy SV, Bruder JM, Cundy T, Cornish 48. Hughes DE, Dai A, Tiffee JC, Li HH, Mundy GR, Boyce
J, Singer FR, et al. (2002) Sequence analysis of measles BF (1996) Estrogen promotes apoptosis of murine
virus nucleocapsid transcripts in patients with Pagets osteoclasts mediated by TGF-beta. Nat Med. 2:
disease. J Bone Miner Res 17:14551. 11326.
Clinical Disorders Associated with Alterations in Bone Resorption 125

49. Ikeda T, Utsuyama M, Hirokawa K (2001) Expression 65. Leach RJ, Singer FR, Roodman GD (2001) The genet-
proles of receptor activator of nuclear factor kappaB ics of Pagets disease of the bone. J Clin Endocrinol
ligand, receptor activator of nuclear factor kappaB, and Metab 86:248.
osteoprotegerin messenger RNA in aged and ovariec- 66. Lee K, Jessop H, Suswillo R, Zaman G, Lanyon L (2003)
tomized rat bones. J Bone Miner Res 16:141625. Endocrinology: bone adaptation requires oestrogen
50. Iqbal AA, Burgess EH, Gallina DL, Nanes MS, Cook CB receptor-alpha. Nature 424:389.
(2003) Hypercalcemia in hyperthyroidism: patterns of 67. Lee S, Finn L, Sze RW, Perkins JA, Sie KC (2003)
serum calcium, parathyroid hormone, and 1,25-dihy- Gorham Stout syndrome (disappearing bone disease):
droxyvitamin D3 levels during management of thyro- two additional case reports and a review of the litera-
toxicosis. Endocr Pract 9:51721. ture. Arch Otolaryngol Head Neck Surg 129:13403.
51. Janssens K, Gershoni-Baruch R, Guanabens N, Migone 68. Lee SK, Lorenzo JA (1999) Parathyroid hormone stim-
N, Ralston S, Bonduelle M, et al. (2000) Mutations in ulates TRANCE and inhibits osteoprotegerin messen-
the gene encoding the latency-associated peptide of ger ribonucleic acid expression in murine bone
TGF-beta 1 cause Camurati-Engelmann disease. Nat marrow cultures: correlation with osteoclast-like cell
Genet 26:2735. formation. Endocrinology 140:355261.
52. Janssens K,Van Hul W (2002) Molecular genetics of too 69. Leonard H, Thomson M, Glasson E, Fyfe S, Leonard S,
much bone. Hum Mol Genet 11:238593. Ellaway C, et al. (1999) Metacarpophalangeal pattern
53. Kanis JA, Delmas P, Burckhardt P, Cooper C, Torgerson prole and bone age in Rett syndrome: further radio-
D (1997) Guidelines for diagnosis and management logical clues to the diagnosis. Am J Med Genet 83:
of osteoporosis. The European Foundation for Osteo- 8895.
porosis and Bone Disease. Osteoporos Int 7:390406. 70. Lewis SE, Erickson RP, Barnett LB,Venta PJ, Tashian RE
54. Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, (1988) N-ethyl-N-nitrosourea-induced null mutation
Glass DA, 2nd, et al. (2002) Cbfa1-independent at the mouse Car-2 locus: an animal model for human
decrease in osteoblast proliferation, osteopenia, and carbonic anhydrase II deciency syndrome. Proc Natl
persistent embryonic eye vascularization in mice de- Acad Sci USA 85:19626.
cient in Lrp5, a Wnt coreceptor. J Cell Biol 157:30314. 71. Li YP, Chen W, Stashenko P (1996) Molecular cloning
55. Kimble RB, Srivastava S, Ross FP, Matayoshi A, Pacici and characterization of a putative novel human osteo-
R (1996) Estrogen deciency increases the ability of clast-specic 116-kDa vacuolar proton pump subunit.
stromal cells to support murine osteoclastogenesis via Biochem Biophys Res Commun 218:81321.
an interleukin-1and tumor necrosis factor-mediated 72. Little RD, Carulli JP, Del Mastro RG, Dupuis J, Osborne
stimulation of macrophage colony-stimulating factor M, Folz C, et al. (2002) A mutation in the LDL recep-
production. J Biol Chem 271:288907. tor-related protein 5 gene results in the autosomal
56. Kinoshita A, Saito T, Tomita H, Makita Y, Yoshida K, dominant high-bone-mass trait. Am J Hum Genet
Ghadami M, et al. (2000) Domain-specic mutations 70:119.
in TGFB1 result in Camurati-Engelmann disease. Nat 73. Manolagas SC, Kousteni S, Jilka RL (2002) Sex steroids
Genet 26:1920. and bone. Recent Prog Horm Res 57:385409.
57. Klein RF, Allard J, Avnur Z, Nikolcheva T, Rotstein D, 74. Maroteaux P, Lamy M (1965) The Malady of
Carlos AS, et al. (2004) Regulation of bone mass in mice Toulouse-Lautrec. Jama 191:7157.
by the lipoxygenase gene Alox15. Science 303:22932. 75. Marx SJ, Agarwal SK, Heppner C, Kim YS, Kester MB,
58. Koga T, Inui M, Inoue K, Kim S, Suematsu A, Kobayashi Goldsmith PK, et al. (1999) The gene for multiple
E, et al. (2004) Costimulatory signals mediated by the endocrine neoplasia type 1: recent ndings. Bone 25:
ITAM motif cooperate with RANKL for bone home- 11922.
ostasis. Nature 428:75863. 76. McGowan NW, MacPherson H, Janssens K, Van Hul W,
59. Kornak U, Kasper D, Bosl MR, Kaiser E, Schweizer M, Frith JC, Fraser WD, et al. (2003) A mutation affecting
Schulz A, et al. (2001) Loss of the ClC-7 chloride the latency-associated peptide of TGFbeta1 in
channel leads to osteopetrosis in mice and man. Cell Camurati-Engelmann disease enhances osteoclast for-
104:20515. mation in vitro. J Clin Endocrinol Metab 88:33216.
60. Kornak U, Schulz A, Friedrich W, Uhlhaas S, Kremens 77. McMahon C, Will A, Hu P, Shah GN, Sly WS, Smith OP
B,Voit T, et al. (2000) Mutations in the a3 subunit of the (2001) Bone marrow transplantation corrects
vacuolar H(+)-ATPase cause infantile malignant osteopetrosis in the carbonic anhydrase II deciency
osteopetrosis. Hum Mol Genet 9:205963. syndrome. Blood 97:194750.
61. Laitala T, Vaananen HK (1994) Inhibition of bone 78. Miura M, Tanaka K, Komatsu Y, Suda M, Yasoda A,
resorption in vitro by antisense RNA and DNA mole- Sakuma Y, et al. (2002) A novel interaction between
cules targeted against carbonic anhydrase II or two thyroid hormones and 1,25(OH)(2)D(3) in osteoclast
subunits of vacuolar H(+)-ATPase. J Clin Invest 93: formation. Biochem Biophys Res Commun 291:98794.
23118. 79. Montecucco C, Caporali R, Caprotti P, Caprotti M,
62. Lanyon L, Skerry T (2001) Postmenopausal osteoporo- Notario A (1992) Sex hormones and bone metabolism
sis as a failure of bones adaptation to functional in postmenopausal rheumatoid arthritis treated
loading: a hypothesis. J Bone Miner Res 16:193747. with two different glucocorticoids. J Rheumatol 19:
63. Laughlin MH, Welshons WV, Sturek M, Rush JW, Turk 1895900.
JR, Taylor JA, et al. (2003) Gender, exercise training, 80. Motil KJ, Schultz R, Brown B, Glaze DG, Percy AK
and eNOS expression in porcine skeletal muscle arter- (1994) Altered energy balance may account for growth
ies. J Appl Physiol 95:25064. failure in Rett syndrome. J Child Neurol 9:3159.
64. Lazner F, Gowen M, Pavasovic D, Kola I (1999) 81. Nasu T, Tsukahara Y, Terayama K (1973) A lipid
Osteopetrosis and osteoporosis: two sides of the same metabolic disease-membranous lipodystrophy-an
coin. Hum Mol Genet 8:183946. autopsy case demonstrating numerous peculiar mem-
126 Bone Resorption

brane-structures composed of compound lipid in bone because of the formation of a constitutively active
and bone marrow and various adipose tissues. Acta form of TGF-beta 1. J Biol Chem 276:1146972.
Pathol Jpn 23:53958. 98. Sezer O, Heider U, Zavrski I, Kuhne CA, Hofbauer LC
82. Niida S, Kaku M, Amano H, Yoshida H, Kataoka H, (2003) RANK ligand and osteoprotegerin in myeloma
Nishikawa S, et al. (1999) Vascular endothelial growth bone disease. Blood 101:20948.
factor can substitute for macrophage colony- stimulat- 99. Shahbazian M, Young J, Yuva-Paylor L, Spencer C,
ing factor in the support of osteoclastic bone resorp- Antalffy B, Noebels J, et al. (2002) Mice with truncated
tion. J Exp Med 190:2938. MeCP2 recapitulate many Rett syndrome features and
83. OBrien CA, Jia D, Plotkin LI, Bellido T, Powers CC, display hyperacetylation of histone H3. Neuron 35:
Stewart SA, et al. (2004) Glucocorticoids act directly on 24354.
osteoblasts and osteocytes to induce their apoptosis 100. Shevde NK, Bendixen AC, Dienger KM, Pike JW (2000)
and reduce bone formation and strength. Endocrinol- Estrogens suppress RANK ligand-induced osteoclast
ogy 145:183541. differentiation via a stromal cell independent mecha-
84. Osterberg PH, Wallace RG, Adams DA, Crone RS, nism involving c-Jun repression. Proc Natl Acad Sci
Dickson GR, Kanis JA, et al. (1988) Familial expansile USA 97:782934.
osteolysis. A new dysplasia. J Bone Joint Surg Br 70: 101. Shi GP, Chapman HA, Bhairi SM, DeLeeuw C, Reddy
25560. VY, Weiss SJ (1995) Molecular cloning of human
85. Pacici R (1996) Estrogen, cytokines, and pathogene- cathepsin O, a novel endoproteinase and homologue of
sis of postmenopausal osteoporosis. J Bone Miner Res rabbit OC2. FEBS Lett 357:12934.
11:104351. 102. Sly WS, Hewett-Emmett D, Whyte MP, Yu YS, Tashian
86. Paloneva J, Mandelin J, Kiialainen A, Bohling T, Prudlo RE (1983) Carbonic anhydrase II deciency identied
J, Hakola P, et al. (2003) DAP12/TREM2 deciency as the primary defect in the autosomal recessive syn-
results in impaired osteoclast differentiation and drome of osteopetrosis with renal tubular acidosis and
osteoporotic features. J Exp Med 198:66975. cerebral calcication. Proc Natl Acad Sci USA 80:
87. Paloneva J, Manninen T, Christman G, Hovanes K, 27526.
Mandelin J, Adolfsson R, et al. (2002) Mutations in two 103. Sly WS, Lang RA, Avioli LV, Haddad J, Lubowitz H,
genes encoding different subunits of a receptor signal- McAlister W (1972) Recessive osteopetrosis: new clin-
ing complex result in an identical disease phenotype. ical phenotype. Am J Hum Genet. 24 (Suppl)::34a.
Am J Hum Genet 71:65662. 104. Sly WS, Whyte MP, Sundaram V, Tashian RE, Hewett-
88. Pearse RN, Sordillo EM, Yaccoby S, Wong BR, Liau DF, Emmett D, Guibaud P, et al. (1985) Carbonic anhydrase
Colman N, et al. (2001) Multiple myeloma disrupts the II deciency in 12 families with the autosomal reces-
TRANCE/ osteoprotegerin cytokine axis to trigger sive syndrome of osteopetrosis with renal tubular
bone destruction and promote tumor progression. acidosis and cerebral calcication. N Engl J Med
Proc Natl Acad Sci USA 98:115816. 313:13945.
89. Pekkarinen P, Kestila M, Paloneva J, Terwillign J, Varilo 105. Sobacchi C, Frattini A, Orchard P, Porras O, Tezcan I,
T, Jarvi O, et al. (1998) Fine-scale mapping of a novel Andolina M, et al. (2001) The mutational spectrum of
dementia gene, PLOSL, by linkage disequilibrium. human malignant autosomal recessive osteopetrosis.
Genomics. 54:30715. Hum Mol Genet 10:176773.
90. Piek E, Heldin CH, Ten Dijke P (1999) Specicity, diver- 106. Srivastava S, Toraldo G, Weitzmann MN, Cenci S, Ross
sity, and regulation in TGF-beta superfamily signaling. FP, Pacici R (2001) Estrogen decreases osteoclast
Faseb J 13:210524. formation by down-regulating receptor activator of
91. Ralston SH (1997) The Michael Mason Prize NF-kappa B ligand (RANKL)-induced JNK activation.
Essay 1997. Nitric oxide and bone: what a gas! Br J J Biol Chem 276:883640.
Rheumatol 36:8318. 107. Szulc P, Hofbauer LC, Heufelder AE, Roth S, Delmas PD
92. Rantakokko J, Aro HT, Savontaus M, Vuorio E (1996) (2001) Osteoprotegerin serum levels in men: correla-
Mouse cathepsin K: cDNA cloning and predominant tion with age, estrogen, and testosterone status. J Clin
expression of the gene in osteoclasts, and in some Endocrinol Metab 86:31625.
hypertrophying chondrocytes during mouse develop- 108. Tian E, Zhan F, Walker R, Rasmussen E, Ma Y, Barlogie
ment. FEBS Lett 393:30713. B, et al. (2003) The role of the Wnt-signaling antago-
93. Reddy SV, Kurihara N, Menaa C, Roodman GD (2001) nist DKK1 in the development of osteolytic lesions in
Pagets disease of bone: a disease of the osteoclast. Rev multiple myeloma. N Engl J Med 349:248394.
Endocr Metab Disord 2:195201. 109. Vaananen HK, Zhao H, Mulari M, Halleen JM (2000)
94. Reid AB, Reid IL, Johnson G, Hamonic M, Major P The cell biology of osteoclast function. J Cell Sci 113
(1989) Familial diffuse cystic angiomatosis of bone. (Pt 3):37781.
Clin Orthop 2118. 110. Van Wesenbeeck L, Cleiren E, Gram J, Beals RK,
95. Riggs BL, Khosla S, Melton LJ, 3rd (2002) Sex steroids Benichou O, Scopelliti D, et al. (2003) Six novel mis-
and the construction and conservation of the adult sense mutations in the LDL receptor-related protein 5
skeleton. Endocr Rev 23:279302. (LRP5) gene in different conditions with an increased
96. Roth DE, Venta PJ, Tashian RE, Sly WS (1992) bone density. Am J Hum Genet 72:76371.
Molecular basis of human carbonic anhydrase II de- 111. Vestergaard P, Mosekilde L (2003) Hyperthyroidism,
ciency. Proc Natl Acad Sci USA 89:18048. bone mineral, and fracture riska meta-analysis.
97. Saito T, Kinoshita A, Yoshiura K, Makita Y, Wakui K, Thyroid 13:58593.
Honke K, et al. (2001) Domain-specic mutations of 112. Waguespack SG, Hui SL, White KE, Buckwalter KA,
a transforming growth factor (TGF)-beta 1 latency- Econs MJ (2002) Measurement of tartrate-resistant
associated peptide cause Camurati-Engelmann disease acid phosphatase and the brain isoenzyme of creatine
Clinical Disorders Associated with Alterations in Bone Resorption 127

kinase accurately diagnoses type II autosomal domi- 120. Whyte MP, Mills BG, Reinus WR, Podgornik MN,
nant osteopetrosis but does not identify gene carriers. Roodman GD, Gannon FH, et al. (2000) Expansile
J Clin Endocrinol Metab 87:22127. skeletal hyperphosphatasia: a new familial metabolic
113. Walker DG (1975) Bone resorption restored in bone disease. J Bone Miner Res 15:233044.
osteopetrotic mice by transplants of normal bone 121. Whyte MP, Obrecht SE, Finnegan PM, Jones JL,
marrow and spleen cells. Science 190:7845. Podgornik MN, McAlister WH, et al. (2002)
114. Walker DG (1993) Bone resorption restored in Osteoprotegerin deciency and juvenile Pagets
osteopetrotic mice by transplants of normal bone disease. N Engl J Med 347:17584.
marrow and spleen cells. 1975. Clin Orthop 294:46. 122. Wiktor-Jedrzejczak W, Bartocci A, Ferrante AJ, Ahmed-
115. Wan M, Lee SS, Zhang X, Houwink-Manville I, Song Ansari A, Sell K, Pollard J, et al. (1990) Total absence of
HR, Amir RE, et al. (1999) Rett syndrome and beyond: colony-stimulating factor 1 in the macrophage-de-
recurrent spontaneous and familial MECP2 mutations cient osteopetrotic (op/op) mouse. Proc Natl Acad Sci
at CpG hotspots. Am J Hum Genet 65:15209. USA 87:482832.
116. Weinstein RS (2001) Glucocorticoid-induced osteo- 123. Wu J, Wang XX, Takasaki M, Ohta A, Higuchi M, Ishimi
porosis. Rev Endocr Metab Disord 2:6573. Y (2001) Cooperative effects of exercise training and
117. Weinstein RS, Jilka RL, Partt AM, Manolagas SC genistein administration on bone mass in ovariec-
(1998) Inhibition of osteoblastogenesis and promotion tomized mice. J Bone Miner Res 16:182936.
of apoptosis of osteoblasts and osteocytes by gluco- 124. Yasuda H, Shima N, Nakagawa N, Yamaguchi K,
corticoids. Potential mechanisms of their deleterious Kinosaki M, Mochizuki S, et al. (1998) Osteoclast dif-
effects on bone. J Clin Invest 102:27482. ferentiation factor is a ligand for osteoprotegerin/
118. Whyte MP, Hughes AE (2002) Expansile skeletal hyper- osteoclastogenesis-inhibitory factor and is identical to
phosphatasia is caused by a 15-base pair tandem dupli- TRANCE/RANKL. Proc Natl Acad Sci USA 95:3597
cation in TNFRSF11A encoding RANK and is allelic to 602.
familial expansile osteolysis. J Bone Miner Res 17:269.
119. Whyte MP, Leelawattana R, Reddy SV, Roodman GD
(1996) Absence of paramyxo virus transcripts in juve-
nile Paget bone disease. J Bone Miner Res 11:1041.
8.
Pharmaceuticals for Bone Disease
Targeting the Osteoclast
Lorraine A. Fitzpatrick

that results in different efcacy and toxicity


Introduction for each one. Bisphosphonates bind to the
surface of bone at sites of active remodeling
Many pharmaceutical agents target the osteo- where two distinct molecular mechanisms are
clast, as this is the cell that resorbs bone leading thought to be responsible for the ability of
to bone loss. Recently, important information bisphosphonates to modify osteoclast func-
regarding osteoclast recruitment and function tion. Non-nitrogen-containing bisphosphonates
has been discovered, leading to a better under- (etidronate, clodronate, and tiludronate) alter
standing of osteoclast biology. Many available cellular function by being metabolized to
agents targeting the osteoclast decrease bone cytotoxic ATP-bisphosphonate analogues [44].
turnover markers, increase bone mineral den- The nitrogen-containing bisphosphonates
sity (BMD) and prevent fractures. (alendronate, risedronate, ibandronate, and
zolendronic acid) inhibit farnesyl pyrophos-
phatase and other steps in the intracellular
mevalonate pathway [5, 48, 75]. As a result, the
Bisphosphonate Overview post-translational modication by prenylation
of proteins such as Ras and Rho is impaired,
Pyrophosphates have been used extensively leading to diminished recruitment or differenti-
in industry because they inhibit precipitation ation of osteoclast precursors and to osteoclast
of calcium carbonates. Pyrophosphates bind death. The loss of osteoclast activity results in a
avidly to calcium phosphate and impair both decrease in the rate at which new bone remod-
the formation of calcium phosphate crystals in eling units are activated and a decrease in the
vitro and in solution. Bisphosphonates display amount of bone resorbing activity in each bone
similar physical chemical activity, but resist remodeling unit. The resulting reduction in
enzymatic hydrolysis. Bisphosphonates are bone turnover indirectly decreases osteoblast
characterized by two carbon-phosphate (CP) function and bone formation.
bonds, and specicity is determined by variance Bisphosphonates are poorly absorbed and
in side chains (Figure 8.1). Many bisphospho- can cause esophageal irritation or even ulcera-
nates have been tested in humans with respect tion if passage through the esophagus is delayed.
to their ability to inhibit bone resorption. As result, instructions for taking medications are
Etidronate, alendronate, clodronate, iban- complex. Although bisphosphonates are poorly
dronate, pamidronate, tiludronate, and zolen- absorbed, approximately 50% of the absorbed
dronic acid are available across the world. amount is taken up by the skeleton. The remain-
However, these bisphosphonates possesses der undergoes renal excretion. In skeletal tissue,
physical, chemical, and biological properties bisphosphonates have a long residual half-life.
128
Pharmaceuticals for Bone Disease Targeting the Osteoclast 129

Figure 8.1 Chemical structures of various bisphosphonates.

Differences in structure determine potency, high doses has resulted in toxicity in children
duration of action, side effects, and efcacy. [79]. Regarding the efcacy of bisphosphonates
The pharmacologically active bisphospho- to inhibit bone resorption, no clear-cut struc-
nates inhibit bone resorption in organ culture. ture and associated effect relationship has
In the intact animal, bisphosphonates block the emerged regarding the pyro-carbon-phosphate
degradation of both primary and secondary tra- (PCP) structure. Because of their strong af-
beculae; this results in arrest of the modeling nity for bone mineral, bisphosphonates are
and remodeling of the metaphysis. In growing deposited in bone and osteoclasts are inhibited
children, this would be predicted to create a when they engulf bisphosphonate-containing
club-shaped bone with a dense metaphysis, a bone. Bisphosphonates inhibit the osteoclast
picture similar to that seen in osteopetrotic function by altering the cytoskeleton and the
animals. This effect can be used to estimate the rufed border, decreasing acid extrusion and
potency of new compounds, and, in rare enzyme activity and increasing osteoclast
instances, bisphosphonate administration in apoptosis. A second target cell is the osteoblast,
130 Bone Resorption

which secretes an osteoclast recruitment inhi- produced increases in BMD that were 2.2%
bitor that is stimulated by the action of greater than in the placebo group (P < 0.001).
bisphosphonates. During the study, 22 (6.2%) of the 355 women
in the placebo group had at least 12 vertebral
Alendronate fractures compared with 17 (3.2%) of the 526
women in the combined alendronate groups
Alendronate is a nitrogen-containing bisphos- (P = 0.03). The relative risk of a new fracture
phonate that was approved by the Food and among the women treated with alendronate (all
Drug Administration in 1995 for treatment of doses pooled) as compared to the placebo group
osteoporosis in the United States. As a result, it was 0.52 (95% CI, 0.280.95). The increased risk
is the most extensively studied bisphosphonate. among women receiving placebo was found in
Alendronate is approved for treatment and pre- both multicenter studies. The estimated risk of
vention of postmenopausal osteoporosis, treat- non-vertebral fractures in women treated with
ment of osteoporosis in men, treatment of alendronate was 0.79 (95% CI, 0.521.22). In this
glucocorticoid induced osteoporosis and Pagets clinical trial setting, alendronate was generally
disease in men and women. well tolerated with no clinical laboratory evi-
In two multicenter, dose-ranging studies, the dence of adverse effects that were greater than
efcacy of continuous oral alendronate therapy those found in the placebo group. Similar rates
in postmenopausal women with osteoporosis of adverse upper gastrointestinal (GI) effects
was studied [45]. One multicenter study was were noted, and it was only later that it was real-
conducted in the United States, and the other ized that GI side effects could be an issue, result-
multicenter study recruited women worldwide. ing in specialized dosing instructions. The
These two trials were randomized, double- results of several trials were consistent with the
blind, placebo-controlled studies with identical prospective epidemiologic studies of the rela-
designs. Women who were 45 to 80 years of age tionship between BMD of the lumbar spine and
and at least ve years post-menopause were risk of vertebral fracture. Daily treatment with
enrolled in the study. Enrollment required that oral alendronate progressively increased bone
the lumbar spine BMD be reduced, with T- mass of the spine, hip, and total body and
scores more than 2.5 standard deviations below reduced the risk of vertebral fracture and the
peak mass. Women were randomly assigned to progression of vertebral deformities in post-
receive placebo or 5, 10, or 20 mg of alendronate menopausal women with osteoporosis. In a sep-
per day for two years, to be followed by open arate study, therapy for one year signicantly
label therapy during the third year. The proto- reduced the incidence of non-vertebral frac-
col was modied to include the third year of tures in women with low bone density (T-score
double-blind therapy and those subjects receiv- <-2.0) [60]. In a prospective study of 2,027
ing 20 mg of alendronate were switched to doses women with pre-existing vertebral fractures,
of 5 mg/day for the third year. All women alendronate therapy reduced the incidence of
received a daily supplement of calcium carbon- vertebral, hip and wrist fractures by approxi-
ate that provided 500 mg of elemental calcium. mately 50% over three years [8]. In this study,
There were signicant increases in BMD of the women aged 5081 years with low femoral neck
spine, femoral neck, trochanter, and total body BMD were enrolled in study groups based on
at six months in the alendronate groups and sig- the presence or absence of an existing vertebral
nicant losses at all sites in the placebo group. fracture. Postmenopausal women were ran-
The mean differences in BMD between those domly assigned to placebo or alendronate and
women receiving 10 mg of alendronate and followed for 36 months. The dose of alendronate
those receiving placebo were 8.8% in the spine, was initially 5 mg daily and was increased to
5.9% in the femoral neck, 7.8% in the trochanter 10 mg daily at 24 months. Compared with the
and 2.5% in the total body (P < 0.01 for all com- placebo group, treatment with alendronate sig-
parisons). The 10-mg dose of alendronate nicantly increased the average bone mass at
resulted in BMD increases at all sites during all the femoral neck (4.1% difference, P < 0.001), at
three years, although the increase in total body the total hip (4.7%, P < 0.001), and in the poste-
BMD during year three was not signicant. In rior-anterior lumbar spine (6.2%, P < 0.001).
the midforearm, a site consisting almost entirely Follow-up radiographs were obtained in 98% of
of cortical bone, the 10-mg dose of alendronate those in the study at closeout. The risk of new
Pharmaceuticals for Bone Disease Targeting the Osteoclast 131

radiographic vertebral fracture was 40% lower group completed the three years of the clinical
in the alendronate groups than in the placebo trial. There were no differences in reasons
group and signicantly fewer women in the for patient withdrawal among treatment and
alendronate groups had clinical vertebral frac- placebo groups. A signicant reduction of 65%
tures (hazard reduction 55%, P < 0.001). There in vertebral fracture risk (P < 0.001) was seen as
was a signicant reduction in hip fractures in early as in the rst year of treatment. Over the
the alendronate-treated group compared to the three-year period, there was a 41% reduction in
placebo treated women (P = 0.047). the risk of new vertebral fractures in patients
In a younger a group of postmenopausal treated with risedronate, compared to women
women, alendronate 5 mg daily for four years receiving placebo (P = 0.003). The cumulative
prevented the bone loss, but no effect on frac- incidence of nonvertebral fractures over three
ture rate was observed [62]. A trial was con- years of treatment was lowered by 39% com-
ducted to compare daily alendronate (5 to pared to women receiving placebo. Over the
10 mg/day) to once-weekly dosing of either 35 three years, there was a signicant increase in
or 70 mg. In this trial, BMD endpoints were BMD from baseline at the lumbar spine (5.4%),
equivalent, although no fracture data are avail- femoral neck (1.6%), and femoral trochanter
able for the groups receiving weekly doses. (3.9%) in the risedronate-treated patients.
In a two-year double-blind trial, alendronate Signicant differences in BMD were notable as
was tested for its efcacy in men. Some 241 men early as six months, compared to the placebo
aged 3187 years with osteoporosis, one-third group. At the mid-shaft radius, the risedronate
with low testosterone levels, were recruited for group had no change in BMD (0.2%) compared
the trial [57]. The men who received alen- to a signicant bone loss of -1.4% in the placebo
dronate had a mean increase in BMD of 7.1% group (P < 0.05). Markers of bone turnover,
the lumbar spine, 2.5% at the femoral neck, and which were measured in a subset of subjects,
2.0% for the total body (P < 0.001 for all com- were reective of these changes and the mecha-
parisons to baseline). There were no signicant nism of action of risedronate. Bone-specic
changes in the femoral neck or total body BMD. alkaline phosphatase, a marker of bone forma-
The incidence of vertebral fractures was lower tion, declined with risedronate treatment, with
in the alendronate group than in the placebo a nadir of -35% compared to baseline (median
group (P = 0.02). Overall, the benets of alen- % change) at six months, compared to -12%
dronate therapy in men with osteoporosis were with the placebo group. At the end of three
very similar to those seen for postmenopausal years, the changes were -33% and -7%, respec-
women. tively, for risedronate and placebo. When
markers of bone resorption were evaluated, the
Risedronate changes were similar. The deoxypyridinoline-
creatinine ratio reached a nadir at six months of
Risedronate is a pyridinyl bisphosphonate -38% in the risedronate group compared with -
approved for the prevention and treatment of 8% in the placebo group and rose by the end of
osteoporosis in postmenopausal women gluco- the study to -26% and -1%, respectively.
corticoid induced osteoporosis and Pagets Overall, adverse events were similar across
disease. In a randomized, placebo-controlled treatment groups. Digestive symptoms com-
clinical trial, risedronate demonstrated efcacy plaints were the most common adverse events
for vertebral and non-vertebral fracture pre- that led to discontinuing the study, with 42%
vention [37]. In this trial, 2,458 postmenopausal of placebo group and 36% of the risedronate
women younger than 85 years with at least one group withdrawing. The most common GI
vertebral fracture at baseline were enrolled. events were dyspepsia, abdominal pain, and gas-
Subjects received oral risedronate (2.5 or tritis, and these events were similar in both
5.0 mg/day) or placebo. All participants received treatment and placebo groups. Indeed the rise-
calcium (1,000 mg/day) and vitamin D to nor- dronate group showed no difference from
malize 25-hydroxyvitamin D levels. The 2.5 mg/ placebo in the incidence of gastrointestinal tract
day of risedronate arm was discontinued after endoscopy or the rate of abnormal ndings on
one year, and all results refer to the 5-mg/day endoscopy.
dosing schedule. Fifty-ve per cent of patients Bone biopsy samples taken at baseline and
in the placebo group and 60% in the treatment at study end showed that bone was normal,
132 Bone Resorption

without evidence of mineralization problems or placebo. The incidence of hip fracture in the
marrow abnormalities in risedronate-treated group of women 70 to 79 years old was 1.9%
subjects. Histomorphometric analysis demon- among those assigned to risedronate and 3.2%
strated a 50% reduction in bone turnover and a among those assigned to placebo (relative risk,
slight increase in cortical thickness in rise- 0.6; 95% CI, 0.40.9; P = 0.009). In the group of
dronate treated patients compared to placebo. women 80 years of age or older, risedronate had
In both groups, there was an increase in median no effect on the incidence of hip fracture. The
cortical porosity that may have resulted from majority (58%) of the women in this group were
the prolongation of the remodeling period. recruited solely on the basis of clinical risk
Overall, this trial demonstrated the efcacy of factors and only 16% were recruited on the basis
risedronate in the treatment of women with of low BMD at the femoral neck. In an analysis
established postmenopausal osteoporosis. The of all the women, the incidence of nonvertebral
incidence of both vertebral and nonvertebral fractures was 9.4% among those assigned to
fractures was decreased by daily oral rise- risedronate, as compared to 11.2% among those
dronate therapy. One of the differences in this assigned to placebo (relative risk, 0.8; 95% CI,
trial compared to other anti-resorptives is that 0.71.0; P = 0.03). Among the women selected
the initial criterion for vertebral fracture was primarily on the basis of nonskeletal risk
based on a vertebral height loss of 15% instead factors, the assigned treatment had no effect on
of the more traditional 20% [8, 45]. A re-analy- the incidence of nonvertebral fracture. Thus,
sis of the study using the 20% reduction in ver- risedronate prevented hip fractures in elderly
tebral fracture criterion was completed and women with osteoporosis as determined by low
showed similar results. BMD at the femoral neck; interestingly in a very
Risedronate reduced the risk of new vertebral elderly group without criteria for osteoporosis
fractures by 41% and reduced the cumulative as measured by BMD, risedronate therapy did
incidence of non-vertebral fractures by 39% not prevent hip fractures.
over a three-year period [37, 66]. In women with
osteoporosis as dened by low femoral neck Ibandronate
BMD who also had a prevalent vertebral frac-
ture, risedronate reduced hip fracture by 60% Oral ibandronate has recently been approved
[55]. for use in the United States at a dose of 2.5 mg/
The only prospective trial in which hip frac- day, but is not available commercially. Iban-
ture was the primary endpoint enrolled 9,331 dronate has been tested in animal models and
women [55]. Two groups of women were found to inhibit bone resorption and improve
enrolled, one of which was composed of subjects bone mechanical properties. Ibandronate is an
70 to 79 years old who had osteoporosis as indi- amino bisphosphonate and is more potent than
cated by either a BMD with a femoral neck <T- risedronate, alendronate, pamidronate, and clo-
score -4.0 or femoral neck T-score lower than dronate. In a small, phase-one study involving
-3.0, along with at least one risk factor for hip 12 healthy men and 5 postmenopausal women,
fracture. The other group consisted of women a single intravenous injection of ibandronate
80 years of age or older who had at least one was associated with an increase in lumbar spine
non-skeletal risk factor for hip fracture, BMD of 2% (1 mg) and of 3% (2 mg). The 1-mg
a femoral neck T-score lower than -4.0, or a dose of ibandronate decreased urinary excre-
femoral neck T-score lower than -3.0, plus a tion of the C-terminal portion of type 1 colla-
hip-axis length of 11.1 cm or greater. Women gen cross-links (CTX) by 200% in men, and the
were randomized to placebo or 2.5 or 5.0 mg/ 2-mg dose by 300% after 14 days. CTX excretion
day of risedronate. Complete follow-up data continued to decrease up to 30 days after the
were available for 64% of women (69% of those single ibandronate injection. In a phase II dose-
with conrmed osteoporosis and 58% of those nding study that extended over 12 months, 180
with mainly clinical risk factors). The duration postmenopausal women who had distal forearm
of follow-up was 2.3 years and the mean dura- T-scores less than 1.5 were randomized to oral
tion of therapy was 2.0 years. In the analysis of ibandronate (0.5, 1.0, 2.5, 5.0 mg/day) or to
all women, the incidence of hip fracture was placebo [63]. A total of 141 patients completed
2.8% among the women assigned to risedronate the study and women who received 2.5 or
as compared to 3.9% among those assigned to 5.0 mg/day had signicant increases in spine
Pharmaceuticals for Bone Disease Targeting the Osteoclast 133

and femoral neck BMD, as well as dose-related treatment (n = 982). In this study, both groups
decrements in biochemical markers of bone received the same cumulative dose of iban-
turnover. In another study, 125 postmenopausal dronate. The primary endpoint was the inci-
women with osteoporosis as dened by a T- dence of new vertebral fractures after three
score < -2.5 received either placebo or intra- years of treatment. Ibandronate, when admin-
venous ibandronate (0.25, 0.5, 1.0, or 2.0 mg IV istered orally on a daily basis, signicantly
every three months for one year) [73]. Lumbar reduced new vertebral fractures by 62%, and by
spine BMD did not change signicantly in the 50% when using intermittent oral administra-
placebo group, whereas BMD increased in a tion. Urinary CTX and serum osteocalcin levels
dose dependent manner up to 5.2% compared were signicantly decreased. In a post hoc sub-
to baseline after 12 months; only the lowest dose group analysis, non-vertebral fractures were
of ibandronate (0.25 mg) did not produce sig- reduced by ibandronate in patients with the
nicant changes. In addition, total hip BMD lowest baseline femoral neck BMD (T-score <
increased in the 1- (1.8%) and 2-mg (2.9%) -3.0).
groups. Ibandronate administered intra- The most common adverse reaction observed
venously has been tested in a clinical trial with with intravenous ibandronate is a transient
fracture as the primary outcome variable [64]. fever that is a common response to intravenous
A total of 2,860 postmenopausal women with a administration of nitrogen containing bisphos-
spine BMD T-score less than or equal to -2.0 phonates. Ibandronate can also cause hypocal-
and at least one vertebral fracture were ran- cemia as well as decreased serum phosphate.
domized to receive either placebo or iban- Flu-like symptoms including chills, bone and
dronate (0.5 or 1 mg) intravenously every three muscle pain may occur and gastrointestinal dis-
months for three years. After three years the comfort has been reported. In seven patients
BMD of spine increased approximately 5% and receiving ibandronate 3 mg intravenously, pro-
that of femoral trochanter BMD by 3.5% from teinuria of uncertain clinical signicance has
baseline. The incidence of vertebral fracture was been noted [58].
too low in both groups to show a signicant
change due to treatment. Pamidronate
Oral ibandronate has also been studied. In
a randomized, placebo-controlled trial, 240 Pamidronate was the rst nitrogen containing
postmenopausal women with osteoporosis re- bisphosphonate to be studied. Pamidronate is
ceived either placebo, daily oral ibandronate available for the treatment of hypercalcemia of
(2.5 mg/day) or intermittent oral ibandronate malignancy, for osteolytic lesions in multiple
(20 mg every other day for the rst 24 days only) myeloma, to treat metastatic cancer growing in
[69]. After 12 months, women taking placebo bone, and for Pagets disease. Currently it is not
were switched to ibandronate for another approved for use in osteoporosis. When admin-
12 months. All patients received calcium istered intravenously, pamidronate increased
500 mg/day and vitamin D 400 IU/day. BMD was BMD or prevented bone loss in postmenopausal
increased by treatment with ibandronate at one women with osteoporosis when given as an
year. After two years, women receiving continu- initial dose of 90 mg followed by 30 mg IV every
ous ibandronate had an increase in lumbar three months [59].
spine BMD of 5.6%, while women taking inter-
mittent ibandronate gained 5.5% in lumbar Etidronate
spine BMD. Similarly, women on both forms of
ibandronate treatment showed identical BMD Etidronate was the rst bisphosphonate to be
gains of 3.4% in the total hip. In a second studied for treatment of osteoporosis. Continu-
double-blind randomized trial, 2,946 post- ous administration of etidronate was found to
menopausal women with a lumbar spine T- impair mineralization of new bone [38]. For this
scores <-2.0 and at least one prevalent vertebral reason, etidronate was given intermittently fol-
fractures received either placebo (n = 982) or lowing a cyclical regimen (400 mg/d for 14 days
oral ibandronate [21]. Women on oral iban- every three months). Under these conditions,
dronate received either continuous 2.5 mg/day etidronate appeared to be safe and effective
or intermittent 20 mg every other day for the when tested in small clinical trials. In prospec-
rst 24 days, followed by nine weeks without tive, randomized, placebo-controlled trials,
134 Bone Resorption

intermittent etidronate therapy produced sig- and/or had one prevalent fracture at the time of
nicant increases in lumbar spine BMD. The entry into the study. Subjects received two doses
trials were not powered to show benet for frac- of 400-mg oral clodronate per day or placebo. A
tures, but the results can be interpreted as total of 593 women with osteoporosis were
indicative of a decrease in the risk of vertebral recruited. In all patients, clodronate administra-
fractures among high-risk patients [72, 77]. At tion was associated with a signicant increase in
high doses, etidronate impairs mineralization of mean spine BMD over the 3 year period (4.35%
newly formed bone and may cause bone pain change from baseline with clodronate compared
and fractures. For this reason, this older bis- to 0.64% in the placebo group, P < 0.0001). At
phosphonate is not recommended for use in the hip, clodronate maintained BMD (0.70%)
osteoporosis. compared to a loss of BMD in the placebo group
(-3.03%, P < 0.0001). There were no differences
Tiludronate in the treatment effect in the postmenopausal
vs. the secondary osteoporosis groups. Distal
Tiludronate has been administered to post- forearm BMD did not change in either clo-
menopausal women to determine the efcacy dronate group. Serum CTX was reduced by a
and safety of this compound in the treatment of median of 46% in the clodronate-treated group
postmenopausal osteoporosis [65]. Two placebo- and was signicantly lower at all times com-
controlled, randomized, double masked, pared to patients receiving placebo. At the end
multicenter, cyclical, intermittent, dose-ranging of the three-year study, the incidence of new
studies were performed in 1,805 women with fractures in the clodronate-treated patients was
low vertebral BMD and prevalent vertebral 12.7% (RR 0.54; 95% CI, 0.370.80), compared to
fractures and in 488 women with low BMD an incidence of 23.3% in the placebo-treated
and no prevalent vertebral fracture. Subjects group. The relative risk was more robust in the
were randomized to tiludronate 50 mg/day, women with a secondary cause of osteoporosis
200 mg/day, or placebo, given orally for the rst (RR 0.35; 95% CI, 0.160.76 for the latter; RR
seven days of each month. A supplement of 0.63; 95% CI 0.400.98 in the postmenopausal
500 mg elemental calcium was provided daily osteoporosis group). Treatment caused a reduc-
from day 8 to the end of the month. Neither study tion in the incidence of single and multiple ver-
demonstrated a statistical or clinically relevant tebral fractures, with the reduction in vertebral
trend in the incidence of adverse events in the fracture risk noted as early as after one year
three treatment groups. Tiludronate adminis- [54]. The ability of clodronate to reduce fracture
tered at these two doses in a cyclical, intermittent risk was independent of traditional risk factors
fashion was not effective in reducing the inci- for fracture such as the presence of baseline
dence of vertebral fractures or in increasing vertebral fractures, age, corticosteroid use,
spinal BMD. smoking habits, weight, and BMD determined
by ultrasound. Most importantly, the authors
Clodronate suggest that the reduction of fracture risk was
independent of baseline BMD.
Several double blind, placebo-controlled studies
have shown that clodronate signicantly re- Zolendronic Acid
duces the incidence of both vertebral and non-
vertebral fractures in patients with osteolysis Zolendronic acid is the most potent bisphos-
caused by breast cancer and myeloma. Con- phonate that has been studied to date. Small
trolled studies have also demonstrated the doses are required for the inhibition of bone
benet of clodronate in either an oral or intra- resorption, and long dosing intervals may be
venous form to increase BMD at the spine used. A phase II study examined the effect of
and hip in osteoporotic patients [29, 32, 74]. A intravenous zolendronic acid on bone density
randomized, placebo-controlled trial with clo- and bone turnover in postmenopausal women
dronate was conducted that had as its endpoint with low bone density [67]. A total of 351 post-
the reduction of the risk of vertebral fracture menopausal women, aged 45 to 80 years, were
[54]. Women with postmenopausal or sec- studied at 24 centers. Women were at least ve
ondary osteoporosis were recruited if they had years postmenopausal and had a BMD at the
BMD in the osteoporotic range (T-score -2.5) lumbar spine with a T-score of at least -2.0 and
Pharmaceuticals for Bone Disease Targeting the Osteoclast 135

no more than one vertebral fracture at screen- venously on an intermittent basis, the resultant
ing. All women received 1 g calcium supplement suppression of biochemical markers of bone
per day. Women were randomly assigned to turnover and the increases in BMD were similar
receive 16 treatment regimens in a double-blind to those encountered when bisphosphonate is
fashion. Three groups received zolendronic acid given orally on a daily basis. The lowering of
by intravenous infusion every three months, at bone turnover due to a single infusion of zolen-
doses of 0.25, 0.5, and 1 mg. Two other groups dronic acid is long lasting: it is believed that
received the total dose of 4 mg of zolendronic zolendronic acid promptly becomes bound
acid either as a single infusion at the beginning to bone and is slowly released back into the
of the trial or in divided doses at six-month circulation.
intervals. A placebo group was included.
At baseline, the mean BMD values in the
lumbar spine corresponded to a T-score of Long-Term Efficacy of the
-2.9. All groups that received zolendronic acid
showed a progressive, statistically signicant
Bisphosphonates
increase in BMD at the lumbar spine through-
out the 12-month study. There were no signi- Because bisphosphonates are relatively new
cant differences among the treatment groups. At antiresorptive agents with a unique mechanism
12 months, the mean lumbar spine BMD in the of action, long-term safety concerns have been
treatment groups was 4.35.1% higher than in raised. In an extension of a three-year (VERT)
the placebo group. The BMD at the femoral neck multinational study [71], subjects were followed
increased progressively throughout treatment, for a total of ve years. Women who completed
with the femoral neck BMD having increased the study in the original study centers were
3.1 to 3.5% over that of the controls at the end entered in an extension study where they con-
of 12 months (P < 0.001). At the distal radius, tinued to receive either risedronate or placebo.
BMD of the treated group had increased by Over the two-year extension, risedronate
0.81.6% at the end of 12 months, whereas in the reduced the risk of new vertebral fracture by
control group the BMD of the distal radius had 59% (95% CI, 1979%; P = 0.01). Over the ve
decreased by 0.8%. Markers of bone resorption years, risedronate treatment produced signi-
reached a nadir at one month; the median cant increases in the lumbar spine BMD (9.3%),
decrease was more than half for serum C- with the differences between treated and control
telopeptide and for the urinary N-telopeptide. groups increasing with time. The proportion of
These decreases were maintained for the next 11 women reporting serious adverse events or
months, while no signicant changes were noted withdrawing from the study was similar in both
in the placebo group. Biopsies of transiliac the treatment and control groups. These obser-
bone specimens from treated women showed vations are reassuring regarding the safety and
decreases in mineralizing surfaces, in rates of efcacy of ve years of treatment.
bone formation, in adjusted mineral apposition Recently, the results of 10 years of alendronate
rates, and in the activation frequency. No treatment of postmenopausal women with
changes were noted in cortical bone thickness, osteoporosis have been published [9]. Treat-
porosity, or cancellous bone volume. Likewise, ment with alendronate, 10 mg/day, for 10 years
there was no evidence of osteomalacia. Mean produced increases in BMD of 13.7% of the
serum calcium concentrations were similar to lumbar spine, 10.3% of the trochanter, 5.4% of
those in the placebo group after three-months. the femoral neck, and 6.7% of the total proximal
Intact parathyroid hormone (PTH) was mea- femur as compared to baseline values. Impor-
sured at baseline and 12 months. The mean tantly, during years 6 through 10, the alen-
value was about 30% higher than at baseline in dronate treated groups had no signicant
the group receiving four doses of 1 mg of zolen- decline in BMD at any skeletal site. Markers of
dronic acid. Adverse events were similar in all bone remodeling were reduced to the normal
active treatment groups, but were signicantly premenopausal range and this effect was sus-
more common than in controls (4567% versus tained through 10 years of treatment. In the
27% in the placebo group). Overall, it may be group in which medication was discontinued,
concluded that when the potent bisphospho- BMD at the lumbar spine and trochanter did
nate zolendronic acid was administered intra- not change signicantly after year ve, but BMD
136 Bone Resorption

decreased signicantly at the femoral neck and fracture efcacy is not known, it is difcult to
forearm. During the initial three-year study, make recommendations on the length of treat-
6.2% of women in the placebo group had new ment. As more potent bisphosphonates become
morphometric vertebral fractures, as compared available, this issue will become more difcult to
to 3.2% in the pooled alendronate groups resolve because it is likely that continued sup-
(P = 0.03). The rates of fracture could not be pression of bone turnover may occur long after
accurately assessed because the date of oc- the drug has been discontinued.
currence within the observational interval was
not known. Other difculties in interpretation
of fracture rates are due to the lack of a long-
term placebo group. After discontinuation of
Bisphosphonate Use in
alendronate, levels of markers of bone remodel- Children with Osteoporosis or
ing increased within a year, but remained below
baseline values. Safety proles were similar in Genetic Bone Disorders
all three groups, including the incidence of
gastrointestinal adverse events. The observed Children with juvenile osteoporosis have been
increases in BMD at the lumbar spine during treated with bisphosphonates. Although the
long-term alendronate therapy are consistent trials are small in size, they point to a small clin-
with a positive bone balance. The increase in ical benet [6, 10]. Several studies have evalu-
secondary mineralization is a contributing ated the effect of bisphosphonates in children
factor to the increase in BMD. The measured with osteogenesis imperfecta [3, 33]. These
levels of bone remodeling markers remained studies showed that bisphosphonate treatment
stable and within the premenopausal range caused an increase in bone density and reduct-
during treatment. The long retention time of ion of fracture incidence without apparent
alendronate in bone may result in recycling of impairment of skeletal growth. However, a
the compound and residual suppression of bone recent case of a child who was overtreated
resorption after discontinuation. with bisphosphonate and developed profound
osteopetrosis was reported [79]. Bisphospho-
nate treatment should, therefore, be used with
How Long Should caution in children.
Bisphosphonate Therapy
Be Continued? Summary of Bisphosphonates
Because bisphosphonate is incorporated into The bisphosphonates are excellent antiresorp-
bone, a single administration can retain its tive therapies that have been in clinical practice
activity for a long period of time even after drug for a number of years. Two orally administered
discontinuation. Controversy exists regarding bisphosphonates, alendronate and risedronate,
the appropriate duration of therapy with bis- are approved for use in United States. Both of
phosphonates. With continuous bisphosphonate these agents are available for weekly dosing.
use, there has been no waning of efcacy up to Intravenously administered bisphosphonates
10 years with alendronate [9]. In early post- are currently under investigation. The conve-
menopausal women, after two years of treat- nience of intravenous administration, which
ment with alendronate or risedronate, indices of may be as infrequently as yearly, may provide
bone turnover returned towards baseline and signicant advantage over weekly oral dosing. It
bone loss continued at a rate similar to the is anticipated that compliance would be greatly
control group. When alendronate therapy was enhanced by this infrequent administration,
discontinued after two to ve years in older however reimbursement issues are unknown. In
postmenopausal women, BMD remained stable addition, avascular necrosis of the jaw [53] and
and bone turnover remained suppressed for at renal toxicity [39, 52] have been reported as
least two years [9]. The relationship to fracture occurring when certain doses of bisphospho-
efcacy, however, remains less well dened due nates have been administered intravenously. For
to the lack of a placebo group in this trial. If this reason, until data are nalized and regula-
Pharmaceuticals for Bone Disease Targeting the Osteoclast 137

tory approval is completed, the weekly oral bis- ders to therapy. Estimates of the number of non-
phosphonates are recommended. In patients responders vary widely and range from less
who are intolerant to oral bisphosphonates than 1% to as high as 30%. Difculties occur
administration, other antiresorptives may be because within-person measurement of BMD
tried. requires longitudinal measurements. In an
analysis of the postmenopausal estrogen/prog-
estin interventions (PEPI) trial, 875 healthy
Estrogen as an Anti- postmenopausal women aged 4564 years of
age were randomized to one of ve treatment
resorptive Agent on Bone groups: placebo, conjugated equine estrogens
(CEE) 0.625 mg/day; CEE, 0.625 mg/day plus
Early cross-sectional studies revealed that BMD medroxyprogesterone acetate (MPA), 10 mg/
decreased with the onset of the menopause. In day for 12 day/month; CEE 0.625 mg/day plus
comparison with age-matched controls, women MPA, 2.5 mg/day; or CEE 0.625 mg/day plus
who undergo premature menopause exhibit a micronized progesterone, 200 mg/day for 12
lower bone density in the spine and femur com- days/month. Replicate BMD measurements
pared to age-matched controls [68]. Bone loss is were used to calculate the within-person mea-
associated with lower serum levels of estradiol surement errors. The investigators also devel-
and is most rapid in the years following natural oped several criteria to classify subjects who
or surgical menopause. This rapid phase of bone were actually losing bone. Only subjects who
loss results in bone loss at rate of 3% per year in adhered to the prescribed protocol were
the spine and lasts for approximately ve years included in the results analysis. In the rst 12
[31]. Thereafter, bone loss slows to a rate of months 2.3% of the subjects lost bone at the
about 0.51.0% per year. Estrogen improves total hip. This percentage fell to 0.4% in the next
activation frequency and preserves bone remo- 1236 months of the study. This indicates that
deling, whereas estrogen deciency decreases bone loss in women taking hormone therapy is
the activation of bone remodeling units. The rare [34].
major action of estrogen is to inhibit osteo-
clasts, which occurs through several different Estrogen and Bone Markers
mechanisms. Evidence obtained in avian, rabbit,
and human species suggests that estrogen acts There are few randomized, placebo-controlled
directly on the osteoclast by regulating specic trials that have evaluated the effect of estrogen
genes. RANKL (receptor activator of NFkB on biochemical markers of bone turnover. The
ligand) binds to the receptor RANK on the PEPI trial [34] (see above) tested eight different
osteoclast surface; this results in the differenti- bone turnover markers and correlated bone
ation, activation and increased survival of markers with BMD of the spine and hip. In the
osteoclasts and leads to an increase in bone active treatment groups, the mean levels of all
resorption [42]. The decoy receptor to RANKL, bone turnover markers decreased and remained
osteoprotegerin (OPG) is secreted by osteo- below baseline values throughout the three
blasts and reduces RANKRANKL interactions years of study. In the 54 women assigned to the
to inhibit the osteoclast. The fall in estrogen placebo group, no changes from baseline in the
at the time of the menopause decreases the mean levels of any bone turnover marker
production of OPG; this allows increased occurred.With further analysis, the authors sug-
RANKRANKL interactions and increased gested that that at baseline, age and bone mass
bone resorption. index accounted for 16% and 25% of the vari-
ance in the hip and spine BMD, respectively.
Randomized, Placebo-Controlled One of the most sensitive markers was the N-
telopeptide of type I collagen (NTX), which
Trials Using Estrogen: Adherence accounted for 12% and 8% of the variance at the
and Response spine and hip, while the carboxy-terminal
telopeptide of type I collagen accounted for 8%
One of the important issues regarding preven- and 7% of the variance, respectively. Neverthe-
tion or treatment of osteoporosis with estrogen less, biochemical markers of bone turnover are
is the number of responders versus nonrespon- not considered useful surrogates for BMD to
138 Bone Resorption

identify individuals with low BMD who might overall increase was 12% above baseline in the
benet from treatment [51]. lumbar spine BMD in the estrogen-treated
group. Measurements of biochemical markers
BMD in Postmenopausal Estrogen such as serum osteocalcin, serum bone alka-
line phosphatase, and urinary hydroxyproline
Progestin Intervention (PEPI) remained depressed [49]. Other studies indicate
Trial: BMD that transdermal estradiol increased BMD of
the spine from 0.8% to 3.7% [28]. Transcuta-
In the PEPI trial, BMD measured at baseline, 12 neous estradiol gel increased BMD approxi-
and 36 months were outcome measures of great mately 3% in the lumbar spine over the BMD of
interest. In the placebo-treated group, there was the placebo group [70].
an average loss of 1.8% and 1.7%, respectively,
of lumbar spine and hip BMD at the end of three Ultra-low-dose Estrogen
years. In the active-treatment groups, BMD
gains ranged from 3.5% to 5.0% in the lumbar Several studies have indicated that even in post-
spine and averaged 1.7% in the hip. Among the menopausal women who are not treated with
adherent participants, no signicant differences estrogen, higher serum levels of estradiol are
in BMD were noted among the active treatment associated with higher BMD and reduced frac-
groups. Women who were older, had lower BMD ture risk, compared with those women within
at baseline, or no history of hormone therapy the same cohort with lower serum estradiol
had larger, more signicant gains in BMD com- levels. Ultra low doses of parental 17-b estradiol
pared to younger women, those with higher administered in the form of a vaginal ring were
baseline BMD or those with prior hormone studied in older women (60 years or older) for
therapy [2]. a six month period. The ring provided 7.5 mg of
estradiol/24 h. Forearm bone density increased
Other Formulations of Estrogen by 2.1% in the estradiol users, whereas nonusers
lost 2.7%. Bone formation markers decreased in
Other types of estrogen have been evaluated for the treatment group, suggesting reduced bone
their effects on BMD. In one study, 75 women, turnover. In this short-term study, there was no
median age 65 years, received transdermal evidence of endometrial proliferation as deter-
patches that delivered 0.1 mg/day of estradiol mined by ultrasonography [56].
for days 1 to 21 and oral medroxyprogesterone Recently, a much lower dose of transdermal
acetate (10 mg/day) for days 11 to 21 of the 28 estradiol was studied than used traditionally. In
day cycle [50]. Women had a lumbar spine and a randomized, placebo-controlled, blinded trial,
femoral neck BMD below the 10th percentile of 417 postmenopausal women with a BMD z-
premenopausal women and greater than one score < -2.0 were given a transdermal patch that
vertebral fracture dened as a decrease in ver- delivered 0.014 mg/day of either estradiol (to
tebral height of greater than 15%. Eight new increase serum estradiol to 1015 pg/ml) or
fractures occurred in seven women in the estro- placebo. All subjects were treated with 800 mg
gen group, compared with 20 fractures in 12 calcium and 400 IU vitamin D. In this two-year
women in the placebo group (relative risk in the trial, there was a signicant increase in lumbar
estrogen group for a new vertebral fracture, spine BMD of 2.3% in the estradiol-treated
0.39; 95% CI, 0.160.95; P = 0.04). The median group at one year compared with an increase of
percentage change in BMD in the estrogen only 0.5% in the placebo-treated group (P <
group was compared to the placebo group. At 0.001). After two years, an increase in the estra-
the lumbar spine, estrogen-treated women diol treated group of 3.0% occurred (P < 0.001
experienced a 5.3% change compared to 0.2% in compared to placebo) [27]. At the total hip, the
the placebo group. At the femoral trochanter, placebo group lost 0.7% of bone at 24 months
hormone treated women had a 7.6% increase compared to a gain of 0.8% in the estradiol
compared with 2.1% in the placebo group. treated group (P < 0.001). Median fall in CTX
Histomorphometric evaluation of iliac biopsy was -20% in the estradiol treatment group, with
samples conrmed the effect of estrogen on about a 15% fall in markers of bone formation.
bone formation rate expressed on a per bone In this small trial, progestogen was not used,
volume basis. Over a three-year period, the and there were no differences in adverse events
Pharmaceuticals for Bone Disease Targeting the Osteoclast 139

in the treatment and placebo groups. Of note, number of women with osteoporosis, hip frac-
there were no changes in the endometrium, ture was reduced by 33% (HR, 0.67; 95%
serum lipid levels, or common side effects such nominal CI, 0.470.96). In a subgroup analysis,
as breast tenderness. Other trials have evaluated hormone therapy decreased the risk of hip frac-
low-dose estradiol as an alternative to the stan- ture by 60% among women who reported a
dard doses of hormone therapy provided in the baseline calcium intake of more than 1,200 mg/
past [61]. day. Hazards ratios for total fractures were also
lower for all groups who received hormone
therapy (HR 0.67; nominal 95% CI, 0.690.83).
Results from the Womens Total hip BMD increased on the average by 1.7%
during the rst year of hormone therapy and by
Health Initiative (WHI) 3.7% by the end of year 3. After three years of
treatment, the difference in favor of hormone
In clinical trials with estrogen, many of the therapy reached 4.5% in the lumbar spine and
earlier studies were uncontrolled or contained a 3.6% in total hip. The WHI hormone therapy
awed control group. The recent publication of trial was the rst randomized clinical trial
the results from the WHI has demonstrated to demonstrate that combination hormone
unequivocally that estrogen reduces fracture therapy reduced the risk of fractures at the hip,
risk. The use of estrogen alone is currently rec- vertebrae, and wrist, even in women who on the
ommended for women who have undergone basis of BMD did not meet diagnostic criteria
hysterectomy. Proven benets include relief of for osteoporosis.
vasomotor symptoms, of vaginal atrophy, and
prevention of osteoporosis. In the WHI, women
who had had a hysterectomy received either
0.625 mg/day of conjugated equine estrogen or
Selective Estrogen Receptor
placebo. The cohort consisted of 10,739 post- Modulators (SERMs) and
menopausal women, aged 5079 years, and the
study was conducted as a randomized, double-
Postmenopausal Osteoporosis
blind, placebo-controlled disease prevention
trial. The average follow-up was 6.8 years. In a Although estrogen therapy is approved for pre-
manner similar to that found with combined vention of osteoporosis, compliance is low due
hormone therapy, there was a reduction in frac- to adverse effects, especially continued men-
ture risk with estrogen alone. The hazard ratio strual bleeding. In a recent telephone survey of
of hip fracture on estrogen therapy was 0.61 women who are members of the Kaiser founda-
(nominal 95% CI, 0.410.91), for vertebral frac- tion health plan, 62% of those aged 5055 years
ture was 0.65 (nominal 95% CI, 0.420.93), and and 48% of women 65 years or older discontin-
for total fractures was 0.70 (nominal 95% CI, ued hormone therapy within one year of initi-
0.630.79). This trial provides strong evidence ating treatment [25]. Thus, agents that mimic
that conjugated equine estrogen alone reduces the effect of estrogen on bone, but not on the
the risk of hip, clinical vertebral, and other frac- uterus, were studied.
tures. The magnitude of reduction was similar
to that observed in the WHI combination Tamoxifen
hormone trial and is consistent with prior
observational studies and recent meta-analyses Triphenylethylene compounds were initially
[80]. evaluated as antitumor agents. Tamoxifen is a
Interestingly, in the WHI, BMD was only mea- partial estrogen agonist with properties of
sured in a small subset of women (N = 1,024), estrogen antagonism. Tamoxifen binds to estro-
who were thought to represent the overall pop- gen receptors and in certain tissues blocks the
ulation enrolled in the WHI. The number of effect of endogenous estrogen. Tamoxifen
women who were osteoporotic was relatively antagonizes the growth of estrogen-dependent
small: 4 % had osteoporosis at the total hip in breast tumor cells and is commonly used in
the hormone therapy group and 6% of women breast cancer patients as an adjuvant therapy.
had osteoporosis in the placebo group (dened Several studies suggested that tamoxifen may
as a T-score of -2.5). Notwithstanding the small act as a bone antagonist in estrogen-replete pre-
140 Bone Resorption

menopausal women. The National Cancer placebo-controlled trial with 7,705 participants
Institute sponsored a trial to test the effect of aged 3180 years [24].At enrollment, the women
tamoxifen to prevent breast cancer in high risk had to be at least two years postmenopausal
subjects. A subset of the trial participants had with a BMD T-score of less than or equal to -2.5
BMD measurements at baseline, and at one, two, at the lumbar spine or femoral neck. Partici-
and ve years. The primary endpoint of this trial pants were randomly assigned to take placebo,
was to prevent breast cancer, and the trial was raloxifene 60 mg/day, or raloxifene 120 mg/day.
stopped early because of success in achieving In terms of bone turnover markers, osteocalcin
the primary endpoint. Although the number decreased by an average of 8.6%, 26.3%, and
of subjects with BMD measurements at the 31.1% in the placebo, 60-mg/day-raloxifene,
ve-year time point thus was reduced, pre- and 120-mg/day-raloxifene groups, respectively.
menopausal women experienced a signicant Urinary CTX decreased in a similar manner.
loss of BMD at the hip and spine. Based on Women receiving raloxifene had fewer new frac-
limited data, monitoring of BMD in pre- tures, whether or not they had a vertebral frac-
menopausal women using tamoxifen is prudent. ture at the time of study entry. The relative risk
In contrast, tamoxifen has a partial protective of vertebral fracture was 0.7 (95% CI, 0.50.8)
effect on the skeleton in postmenopausal for the 60 mg/day raloxifene-treated group and
women [47]. 0.5 (95% CI, 0.40.7) for the 120 mg/day treated
group. After 4 years the reduction in vertebral
Raloxifene fracture risk was maintained (RR 0.64 in
the 60 mg/day raloxifene-treated group) [22].
Raloxifene was developed as an antitumor As compared to the placebo treated women,
agent, but its ability to maintain bone density in women treated with 60 mg/day increased BMD
animal studies led to the implementation of at the femoral neck and spine (by 2.1% and 2.6%
clinical testing. In postmenopausal women, respectively).
administration of raloxifene decreased bone One of the major side effects in this trial was
turnover markers, although to a lesser extent that women receiving raloxifene had an
than when they received conjugated equine increased risk of venous thromboembolism (RR
estrogens. A double-blind, randomized con- 3.1; 95% CI, 1.56.2). There was a slight increase
trolled study compared the effects of raloxifene in the number of hot ashes and of leg cramps
(60 mg per day) to those of a conjugated equine in the raloxifene-treated subjects (hot ashes:
estrogen (0.625 mg/day) [61]. There was a 6.4% in the placebo-treated group vs. 9.7% in
marked decrease in CTX and NTX (59% and the 60-mg/day-raloxifene group; leg cramps:
43%, respectively) in the estrogen-treated 3.7% in the placebo-treated group vs. 7.0% in
women, compared to decreases of 23% and the 60-mg/day-raloxifene group) [24].
22%, respectively, in raloxifene-treated women. Two identical studies enrolled 1,145 healthy,
Greater reductions in markers of bone forma- postmenopausal women worldwide in a parallel,
tion such as osteocalcin, bone specic alkaline double-blind, placebo-controlled trial. Subjects
phosphatase, and C-terminal type I procollagen received placebo, or one of three doses (30, 60, or
peptide occurred in the estrogen-treated group. 150 mg/day) of raloxifene for three years. In the
BMD increased in both raloxifene and estrogen placebo group, there was a -1.32% loss of BMD
treated groups, but the women treated with con- at the lumbar spine. In the raloxifene-treated
jugated equine had a greater increase in BMD groups, there was a respective increase of 0.71%,
after six months. Bone biopsies revealed no dis- 1.28%, and 1.20% (30, 60, or 150 mg/d) in BMD
tinct histomorphometric differences in static at the lumbar spine. Similar changes in BMD
indices. Dynamic indices of bone remodeling were noted at the hip. As with the larger MORE
were decreased from baseline in the conjugated trial, the only signicant adverse event was an
equine estrogen group, but not in the raloxifene increase in the number of hot ashes [40].
group. Mineralizing surface did not change in
either group, suggesting that mineralization was New Generation SERMs
not adversely affected. Bone architecture was
also similar in both groups. The Multiple Levormeloxifene has been evaluated in a phase
Outcomes of Raloxifene Evaluation (MORE) II clinical trial in which there was a signicant
study was a multicenter, randomized, blinded, increase in lumbar spine BMD. Unfortunately,
Pharmaceuticals for Bone Disease Targeting the Osteoclast 141

endometrial thickness increased in the lev- the relative risk of developing a new vertebral
ormeloxifene group and 8 women experienced fracture took place in the salmon calcitonin
uterovaginal prolapse. As a result, this SERM is nasal spray 200 IU group compared with the
no longer in clinical development [1]. placebo group (RR = 0.67, 95% CI, 0.470.97; P
Lasofoxifene is a potent SERM under = 0.03). In women who had one to ve fractures
evaluation for prevention and treatment of at enrollment, the relative risk was reduced by
postmenopausal osteoporosis. A phase two 35% (P = 0.13) [12]. There were no signicant
randomized, double-blind, placebo-controlled differences in any of the outcomes when the 100
study examined the efcacy in bone loss pre- and the 400 IU treatment groups were compared
vention in 190 postmenopausal women. The to the placebo group. Compared with placebo,
trial duration was for one year in subjects aged the percentage of participants with nonverte-
5068 years. Subjects received lasofoxifene (0.4, bral fractures was signicantly lower in the
2.5, and 10 mg/day), or conjugated equine estro- group who had received 100 IU of salmon calci-
gen/medroxyprogesterone (0.6 25/2 .5 mg/day) tonin nasal spray (P < 0.05). This was not true
or placebo. All received calcium and vitamin D for the groups that had received either 200 or
daily. All lasofoxifene doses increased lumbar 400 IU. The small number of hip and femoral
spine and BMD compared with placebo; fractures precluded a meaningful statistical
however, there were no signicant differences analysis. There was a nonsignicant reduction
among groups for hip BMD. Lasofoxifene de- in the risk of hip fracture in the group that had
creased biochemical markers of bone turnover received 200 IU (RR = 0.5, 95% CI, 0.21.6). A
(P < 0.01) and lowered LDL-cholesterol (P < signicant reduction in hip fractures was
0.001). The most commonly reported adverse recorded in the group that received 100 IU (RR
events associated with lasofoxifene were hot = 0.1, 95% CI, 0.01 to 0.9; P = 0.04), but not in
ashes, leg cramps, and leucorrhea [26]. the group that received 400 IU. The risk of frac-
tures of the arm (humerus, radius, ulna, wrist)
Calcitonin was signicantly reduced in the group that had
received the 100 IU dose, but this was not true
Calcitonin is a physiologic, endogenous inhi- for the 200 IU or 400 IU groups. Lumbar spine
bitor of bone resorption. Its inhibition of osteo- BMD did not change substantially in the
clast formation and attachment results in placebo group during the course of the study. At
bone resorption in vivo and in vitro. Several years 1 and 2, the BMD of the lumbar spine
studies have evaluated calcitonin as an agent to increased signicantly (P < 0.05) in all-calci-
decrease bone resorption in postmenopausal tonin-treated groups. At the end of year 3, the
osteoporosis. Salmon calcitonin has been increase in BMD of the lumbar spine was statis-
administered by injection or by nasal spray. A tically greater in the 400 IU treatment group
ve-year, multicenter clinical trial (the Prevent only. In the course of ve years, lumbar spine
Recurrence of Osteoporotic Fractures [PROOF] BMD in each treatment group increased signif-
study) determined the long-term safety and icantly from baseline every time point measured
efcacy of salmon calcitonin nasal spray in (P < 0.01). No clinically signicant treatment
the prevention of vertebral fractures in post- effect on BMD was apparent at the femoral neck
menopausal women with osteoporosis [12]. or trochanter. There was a statistically signi-
This double-blind, placebo-controlled trial was cant increase in years one and two in BMD at
conducted in 42 centers in the United States and Wards triangle in the group that received 200
in ve centers in the United Kingdom. Patients IU.
were eligible to be included in the study if they Serum C-telopeptide levels decreased signi-
were postmenopausal and a lumbar spine BMD cantly from baseline in the groups that had
T-score of less than or equal to -2. A total of received 200 IU and 400 IU salmon calcitonin
1,255 women received salmon calcitonin nasal by nasal spray. Compared with placebo,
spray at a dose of 100, 200, or 400 IU. All partic- serum bone-specic alkaline phosphatase levels
ipants received 1,000 mg oral calcium and a decreased signicantly in the 200 IU group at
multivitamin containing 400 IU vitamin D. each time point (P < 0.05). Serum osteocalcin
Fifty-nine percent of the participants withdrew levels decreased signicantly from baseline
from the study prematurely, with dropout values, but did not differ from the placebo
similar in all dosage groups. A 33% reduction in values. Twenty-six percemt of participants in
142 Bone Resorption

the 100 IU group, 29% in the 200 IU group, and postmenopausal women with comparable
34% in the 400 IU group developed antibodies lumbar spine BMD. Some women received
that bind salmon calcitonin with titers >1,000. 0.625 mg/day conjugated equine estrogens plus
However, the high titers of antibodies did not 5 mg/day risedronate, whereas others received
alter the effect of salmon calcitonin on the risk 0.625 mg/day conjugated equine estrogens
reduction of new vertebral fractures. (plus placebo) daily for 12 months. Both the
Adverse effects were similar in the treated hormone-only and combination risedronate
and control groups, except for an increase in hormone groups produced signicant increases
rhinitis in the calcitonin treatment groups. from baseline in mean BMD at the lumbar spine,
Rhinitis was dened as nasal congestion, nasal femoral neck, trochanter, midshaft radius, and
discharge, or sneezing and occurred in 22% of distal radius. Markers of bone formation and
treated participants compared to 15% of the resorption decreased signicantly at all time
controls (P < 0.01). Headache was reported less points in both groups. The decrease in bone
frequently in the treated groups compared to turnover markers was signicantly greater at
the control group. most time points in the combination therapy
This ve-year clinical trial indicates that 200 group compared to the group receiving hor-
IU of salmon calcitonin administered daily by mone alone. Differences between treatment
nasal spray signicantly reduces the risk of groups were relatively small and of uncertain
new vertebral fractures by 3336%. In post- clinical signicance. The major limitation of the
menopausal women with low bone mass or study was the short duration and the lack of a
prevalent vertebral fractures, it was surprising group that received risedronate only.
that there was no dose response to treatment. In another study of combination therapy, 573
Nevertheless, this ve-year study demonstrated postmenopausal women were given 0.625 mg/
a sustained effect on reduction of fracture risk day of estrogen combined with medroxypro-
at the spine, maintenance of improved BMD, gesterone (2.5 mg/day), alendronate (10 mg/
and suppression of bone turnover. This suggests day), or a combination of hormone therapy plus
that resistance to the drug did not develop. alendronate. Bone mass of the entire cohort was
Overall, salmon calcitonin nasal spray categorized as osteopenic by the World Health
appears to have been well tolerated. The high Organization criteria; and among this cohort,
rate of dropout (12% per year over the ve 34% had osteoporosis. At the end of three years,
years) during this trial was disappointing, and it total hip BMD showed a mean increase of 5.9%
is difcult to anticipate what the outcome might with combination alendronate and hormone
have been had a larger number of subjects com- therapy, an increase of 4.2% with alendronate
pleted the trial. One explanation for the dropout alone and an increase of 3.0% with hormone
rate may be that it was initially not considered alone (all P < 0.001). BMD remained level in the
ethical to withhold BMD results from the inves- placebo group that was given calcium and
tigator and the participant in a long trial. Two vitamin D. At 36 months, women treated with
new treatments for osteoporosis were approved combination therapy exhibited signicantly
in the United States while the trial was under greater increase in total hip and lumbar spine
way. Conceivably, the investigators or partici- BMD than those on either alendronate or
pants may have perceived the calcitonin spray hormone therapy alone. At the posterior ante-
to be ineffective, which might have caused rior lumbar spine, BMD increased 10.4% in the
some participants to drop out of the study combination group, 7.7% in the alendronate
prematurely. group, 7.1% in the hormone therapy group, and
1.1% in the placebo group. It thus seems that
combination therapy yielded greater improve-
The Use of Combination ments in BMD at the spine and total hip than
treatment with either hormone therapy or alen-
Antiresorptive Therapies dronate alone. The study is important because
the cohort of women were approximately a
Several studies tested the effects of combination decade older than the subjects in the WHI that
antiresorptive therapies to enhance efcacy excluded women older than 79 years of age.
further. Harris et al. [37] combined hormone Interestingly, therapy with alendronate alone
therapy with risedronate in 524 ambulatory was superior to hormone therapy, and combi-
Pharmaceuticals for Bone Disease Targeting the Osteoclast 143

nation therapy with both was superior to either dronate alone or the two in combination were
one alone [35]. studied [7]. Postmenopausal women, aged
In a three-year, double-blind, placebo-con- 5585 years with a BMD T-score of -2.5, or a T-
trolled trial, women were randomized to receive score of -2.0 with an additional risk factor (age
alendronate (10 mg/day, n = 92), conjugated >65 years, vertebral or nonvertebral fracture, or
equine estrogen (0.625 mg/day, n = 143), alen- maternal history of fracture) were enrolled.
dronate and conjugated equine estrogen (n = Women who had previously been treated with
140), or placebo (n = 50) [34]. Women who bisphosphonate for more than one year were not
received estrogen during the rst two years excluded. All subjects were given 500 mg ele-
and then were switched to placebo showed mental calcium and 400 IU of vitamin D. A total
signicant losses in BMD at the spine (-4.5%), of 238 women were randomly assigned to
total hip (-1.8%), femoral neck (-2.4%), and PTH(184) (n = 119), alendronate 10 mg/day
trochanter (-2.7%). Patients in the alendronate (n = 60), or a combination of these agents
and combination therapy groups that were (N = 59). Matching placebos were provided to
switched to placebo in year 3 showed no signif- the patients on monotherapy. Changes in the
icant changes in BMD at any skeletal site. BMD of the lumbar spine in the PTH alone and
Women who continued taking combination the combination of PTH and alendronate
therapy with alendronate and conjugated groups were similar (6.3 and 6.1%, respectively)
equine estrogens for the third year had a spine at 12 months. There was no statistical difference
BMD that was 11.3% higher and a hip BMD that between these data and the 4.6% change in BMD
was 6.6% higher than placebo recipients. The of the lumbar spine in the group that received
accelerated rate of bone loss after discontinua- only alendronate. At the total hip and femoral
tion of hormone therapy has been conrmed neck sites, BMD remained unchanged in the
in other studies. It is interesting to note that group given only PTH. However, an increase in
women who took placebo with calcium supple- BMD at these sites occurred in the subjects on
mentation for three years maintained bone combination therapy or on alendronate alone.
mass at the hip and spine. The increase in the total hip BMD on the com-
bination therapy was statistically signicant. At
the distal radius, there was a statistically signif-
The Use of Combination icant decrease in the PTH treated group and this
effect was attenuated by the addition of
Anabolic and Antiresorptive alendronate. To evaluate volumetric density in
these subjects, quantitative computer tomogra-
Therapies phy was performed. The combined trabecular
and cortical volumetric density of the lumbar
The question of efcacy and safety of combina- spine revealed a nding similar to that found in
tion therapy with antiresorptive and anabolic the areal results. In the trabecular component of
agents is frequently raised. An initial study that the lumbar spine, there was a signicant
looked only at bone markers indicated that increase in volumetric density of the lumbar
the combination of a bisphosphonate such as spine in all groups, but the increase in the PTH
alendronate and of PTH, an anabolic agent, treated group was approximately twice that
would have an additive effect [14]. Recently, found in the combination group. The amount of
several studies have been published that increase in volumetric density of the trabecular
combine PTH with a bisphosphonate, estrogen, lumber spine was similar in the two groups that
or SERM. Both PTH (134) and PTH (184) received either alendronate alone or a combina-
stimulate bone formation, in contrast to the tion of the two agents. In all groups, the patterns
antiresorptive agents that target the osteoclast. of volumetric density were similar at the
The actual ndings in these studies were sur- femoral neck. In the patients treated with PTH
prising, as using a bisphosphonate in combina- alone, cortical bone volumetric density in-
tion with an anabolic agent was predicted to creased signicantly at the total hip (3.5%)
produce a larger increase in BMD than in either and femoral neck (3.4%); this effect was miti-
agent alone. gated by alendronate, suggesting that the anti-
In a randomized, multicenter, double-blind resorptive may prevent bone formation due to
clinical trial, therapy with rhPTH(184), alen- PTH.
144 Bone Resorption

Finkelstein et al. [30] studied 83 men between These two studies [30, 7] indicate that with
46 and 85 years of age with low BMD (T-score regard to trabecular bone at the spine, treatment
less than 2). The men were stratied into blocks by PTH alone is better than combination
on the basis of age (<65 years of age or >65 years therapy of PTH and alendronate, either choice
of age) and according to BMD of the spine. Sub- of which is better than alendronate alone. This
jects received alendronate (10 mg/day), PTH contradicts the hypothesis that alendronate and
(40 mg/day injected subcutaneously), or both. PTH would have additive or synergistic effects.
Alendronate was given for six months prior to In men who were treated for a longer period of
the initiation of PTH therapy. The study was not time [30], PTH therapy alone was the most
double blind, because the IRB considered it effective treatment. Khosla [41] proposed that
unethical to administer placebo injections for the overall inhibition of bone turnover by alen-
two years. Calcium intake was ~1,000 mg/day dronate impairs the anabolic activity of PTH.
through diet or supplementation and all sub- There is little evidence that PTH can cause the
jects received 400 IU/day of vitamin D. The pool of pre-osteoblastic cells to expand. If bone
baseline characteristics of the 73 men included turnover and, therefore, bone formation is
in the intention-to-treat analysis were similar reduced, PTH may be less effective. Alterna-
among all treatment groups. BMD at the poste- tively, alendronate may inhibit the osteoblast,
rior anterior spine increased more in men preventing PTH from exerting its anabolic
treated with PTH alone than in those treated effect.
with alendronate alone or with a combination of Combining PTH treatment with gonadal
the two. This was true even though the period hormone replacement therapy (HRT) showed
of active PTH treatment was six months shorter markedly different results. In one study, women
when given alone. The BMD at the posterior who had been on HRT for at least two years were
anterior spine increased more with combination divided randomly into two groups. The rst
therapy than with alendronate alone (P < 0.001). group remained on HRT and received daily PTH
At the femoral neck, BMD increased more in the (134) (25 mg/day, subcutaneously) for three
PTH (9.7%; 95% CI 6.213.4) than in the alen- years, while the second group remained on HRT
dronate group (3.2%; 95% CI 1.54.8) (P < alone. Subjects in the combination therapy
0.001) or the combined therapy group (6.2%; group increased their bone mass by 13.4% in the
95% CI 4.08.4) (P = 0.01). The BMD at the total lumbar spine, 4.4% in the total hip, and 3.7% in
hip increased more in the PTH (6.4%; 95% CI the total body. After discontinuation of PTH,
3.69.1) than in the alendronate group (4.8%; bone density measurements remained stable for
95% CI 3.36.3). Density at the total hip changed one year.Vertebral fractures were reduced in the
to the same degree in the alendronate and the women on the combination PTH plus HRT
combined therapy groups. The BMD at the therapy compared to women who did not get
radial shaft increased slightly in the alendronate PTH (P < 0.02) [13]. In a smaller study (n = 17
(1.0%; 95% CI 0.21.8) and in the combined per group), women received estrogens for one
therapy groups (1.0%; 95% CI -0.12.1), but year and then were randomly assigned to
decreased slightly in the PTH treated group receive either human PTH (134) plus estrogen
(-0.8%; 95% CI -2.30.6). Total body BMD did or remain on estrogen alone. During the early
not differ in the three treatment groups. months of PTH treatment, osteocalcin levels
Black et al. [7] reported similar data show- increased signicantly; this had not been true in
ing a negative effect of alendronate on PTH- patients treated with a combination of alen-
induced bone formation in postmenopausal dronate and PTH. Bone mass in the lumbar
women. Alendronate administration dimin- spine increased continuously throughout the
ished the PTH-associated increase in serum study in the PTH-treated group. At the end of
total alkaline phosphatase levels normally asso- three years of treatment, vertebral bone mass
ciated with PTH induction of osteoblastic activ- had increased by 13% in the PTH treated
ity. The nding that alendronate reduced the group and had not signicantly declined in the
ability of PTH to increase the activity of serum group treated with hormone therapy alone.
alkaline phosphatase and reduced the effect on There was only a modest increase of 2.7% in the
BMD at the spine and femoral neck suggests total hip for the PTH-treated patient and no sig-
that the effect of PTH depends, at least in part, nicant decline in the hormone-treated group
on its ability to reduce bone resorption. [46].
Pharmaceuticals for Bone Disease Targeting the Osteoclast 145

In a head-to-head study, Ettinger et al. [23] Meta-analysis of Antiresorptive


compared alendronate, raloxifene, and their
combination with PTH on BMD in post- Therapies
menopausal women aged 60 to 87 years with
a prior lumbar spine or total hip BMD T-score A series of meta-analyses were recently pub-
of -2.0 on entry. Before the study, subjects had lished that utilized the Cochrane Collection
been regularly receiving alendronate (10 mg/ system to evaluate the changes in BMD and frac-
day) or raloxifene (60 mg/day) for 1836 ture risk associated with antiresorptive thera-
months. All women received daily self-adminis- pies [15, 20]. The methodology is described in
tered subcutaneous injections of teriparatide detail in the rst paper of the series [15] and the
(rhPTH134), 20 mg/day, 1,000 mg calcium, and summary [20] cautions the reader that these
400 IU of vitamin D. After one month of teri- data should not be used as head-to-head com-
paratide treatment, both groups of women had parisons among antiresorptive agents.
statistically signicant increases in median Eleven trials evaluated the effect of alen-
osteocalcin, PINP, and bone alkaline phos- dronate for at least one year [16]. BMD changes
phatase levels. At 3 and 6 months, lumbar spine varied positively with both dose and time of
BMD increased 2.1% and 5.2%, respectively, rel- alendronate administration. After three years of
ative to baseline in the group that had been on treatment with 10 mg/day of alendronate or
raloxifene. This was not true for the group that more, the pooled estimate of the difference in
had been receiving alendronate. After 6 months, percentage change between alendronate and
increases in BMD were similar in all groups. At placebo was 7.48% (95% CI, 6.128.85) for the
18 months, the lumbar spine BMD had increased lumbar spine (23 years), 5.60% (95% CI,
10.2% in the group that had been on raloxifene. 4.806.39) for the hip (34 years), and 2.08%
It had increased only 4.1% in the group that had (95% CI, 1.532.63) for the forearm (24 years).
been on alendronate. Early changes in total hip The effects on BMD were similar in the preven-
BMD also differed: at 6 months, the raloxifene- tion and treatment trials. The pooled relative
treated group showed little change, whereas hip risk for vertebral fractures in subjects given
BMD decreased signicantly in the alendronate 5 mg/day or more of alendronate was 0.52 (95%
group (-1.8%; P = 0.0002). CI, 0.430.65). The relative risk of nonvertebral
This study highlights differences in the mech- fractures in patients given 10 mg/day or more of
anisms of action by raloxifene and alendronate, alendronate was 0.51 (95% CI, 0.380.69). The
which may be related to the low availability relative risk of adverse events was pooled from
of target cells for the anabolic actions of data reported from nine trials: the relative risk
teriparatide. After two to three years of alen- leading to discontinuation of alendronate was
dronate treatment, histomorphometric analyses 1.15 (95% CI 0.931.42) largely due to gastro-
of bone biopsy specimens indicated that bone intestinal side effects. Although these relative
activation frequency and mineralizing surfaces risks for adverse events are low, they may not
decreased by 90% compared to baseline [11]. apply to the general population, which on
Consequently, few osteoblasts or pre-osteoblasts average may be less healthy than participants in
are available for teriparatide to act on. An alter- a clinical trial setting.
native explanation is that alendronate reduced For risedronate, a total of nine randomized,
the PTH-induced increase in bone resorption placebo-controlled clinical trials in post-
and thus prevented the osteoclast-dependent menopausal women were reviewed [17]. The
release of cytokines that stimulate bone forma- increase in BMD at the lumbar spine, femoral
tion. Raloxifene may cause less suppression of neck, and combined forearm was generally
bone turnover, and thus allow osteoblast, pre- larger for the 5-mg/day dose than seen for cyclic
osteoblast and stromal target cells to remain administration or for lower doses. The pooled
available as target cells for the anabolic actions estimate of the difference in percentage change
of PTH. The nding that lumbar spine BMD and in BMD between 5 mg/day of risedronate and
bone turnover markers were not correlated in placebo after the nal year of treatment (1.53
the subjects treated previously with alendronate years) was 4.54% (95% CI, 4.124.97) for the
supports the concept that changes in the remod- lumbar spine, and 2.75% (95% CI, 2.323.17) at
eling rates and in mineralization are responsible the femoral neck. The pooled relative risk for
for obscuring the anabolic effects of PTH. vertebral fractures in postmenopausal women
146 Bone Resorption

given 2.5 mg or more of risedronate was than 20% loss to follow up and the largest
decreased to 0.64 (95% CI, 0.540.77). The trial, PROOF, had an almost 60% loss to follow
pooled relative risk of non-vertebral fractures up. For bone density of the lumbar spine, the
in patients given risedronate was also de- pooled weekly dose of 250 to 2,800 IU per week
creased to 0.73 (95% CI, 0.610.87). In the eight resulted in signicant increase in the weighted
trials that provided information about adverse mean difference of 3.74% (95% CI, 2.045.43,
events and discontinuation, no treatment effect P < 0.01, n = 2,260, 24 trials). The combined
was noted [77]. forearm showed a similar effect, with a
For HRT, 57 randomized placebo trials lasting difference of 3.02% (95% CI, 0.985.07, P < 0.01,
more than one year (calcium/vitamin D as n = 468, 9 trials). At the femoral neck, the
placebo) were reviewed [78]. Seven trials pooled weighted mean difference showed a
included fracture risk. HRT had a consistent statistically nonsignicant trend toward benet,
effect on BMD at all sites, with the largest effect 3.80% (95% CI, 0.327.91, P = 0.07, 9 trials,
noted at the lumbar spine. The difference n = 513).
between HRT and control in the percent change These meta-analyses provide precise pooled
in bone density at two years was 6.76% (95% CI, estimates of the magnitude of treatment effects
5.837.89; 21 trials) at the lumbar spine and for many antiresorptive therapies. Rigorous cri-
4.53% (95% CI, 3.685.36; 14 trials) and 4.12% teria were used to assess inclusion of the clini-
(95% CI, 3.454.80; 9 trials) at the forearm and cal trials in the analysis. Data are provided in
femoral neck, respectively. The effects of com- Tables 8.1 and 8.2, but inferences and compar-
bined estrogen and progestogen therapy were isons among the various therapies should not be
similar to unopposed estrogen, but consistently drawn, as the condence intervals around the
larger, from 1% to 3%, after two years of therapy treatment effects vary considerably and differ-
[78]. No differences were noted when the data ent patient populations and methodologies
were analyzed by the type of estrogen prepara- were employed in the various trials. For an indi-
tion. Although the relative risk for vertebral and vidual patient, other factors weigh in on treat-
non-vertebral fractures was estimated, this esti- ment selection such as side effects, cost, and
mate has been supplanted by the data obtained convenience. These values and preferences may
from the WHI (see above). drive treatment decisions.
Seven trials with raloxifene were included for
meta-analysis. Raloxifene resulted in positive
effects on the percentage change in BMD, which
increased over time and was independent of
Conclusion
dose. There was an increased impact on BMD
with longer duration of use of raloxifene, but no Currently, approximately 10 new compounds in
impact of dose. Data from one large dominating phase II clinical trials are under investigation
trial suggested a reduction in vertebral fractures for the treatment of bone loss. In addition to the
with a relative risk of 0.60 (95% CI, 0.500.70, P newer bisphosphonates and SERMs discussed
< 0.01). Raloxifene slightly increased rates of above, other therapies that target the osteoclast
withdrawal from therapy as a result of adverse have been developed. Several compounds that
effects in three trials that were evaluated (RR are now in preclinical or early clinical trials may
1.15, 95% CI, 1.001.33, P = 0.05). For other be found useful to reduce osteoclast-mediated
adverse events, four trials were evaluated. There bone resorption. One such agent is an estrogen-
was a signicant increase in deep venous throm- related synthetic ligand that has non-genotropic
bosis in the raloxifene arm, as noted in the effects on bone, thereby by-passing the
results from the MORE trial (3.51; 95% reproductive organs. In animal studies, estren
CI,1.448.56, P = <0.01) [18]. (4-estren-3a,17-diol) did not induce transcrip-
A number of randomized trials suggested tional activity in reproductive tissues of female
that calcitonin could prevent loss of BMD of the mice, but inhibited etoposide-induced apoptosis
spine in late postmenopausal women. Due to in isolated osteoblasts and increased apoptosis
the wide variability in the daily doses used, the of osteoclasts. In the estren-treated mice, the
authors used total weekly doses as the basis ovariectomy-induced bone loss was reduced
for comparison. Of 30 trials, all but two utilized and bone strength was restored, at least in part.
salmon calcitonin [19]. Nine trials had a greater These mechanism-specic ligands may provide
Pharmaceuticals for Bone Disease Targeting the Osteoclast 147

Table 8.1. Magnitude of Effect on Vertebral Fractures


Intervention No. of Trials/Patients Relative Risk (95% CI) Relative Risk, Heterogeneity,
P-value P-value
Calcium 5 (576) 0.77 (0.541.09) 0.14 0.40
Vitamin D 8 (1130) 0.63 (0.450.88) <0.01 0.16
Alendronate (540 mg) 8 (9360) 0.52 (0.430.65) <0.01 0.99
Etidronate (400 mg) 9 (1076) 0.63 (0.440.92) 0.02 0.87
Risedronate 5 (2604) 0.64 (0.540.77) 0.01 0.89
Calcitonina 1 (1108) 0.79 (0.621.00) 0.05 n/a
Raloxifene 1 (6828) 0.60 (0.500.70) 0.01 n/a
HRT 5 (3117) 0.66 (0.41, 1.07) 0.12 0.86
Fluoride (4 yr) 5 (646) 0.67 (0.38, 1.19) 0.17 0.01
a
Due to the potential for publication bias, we are presenting the estimate for the larger RCT estimate from the PROOF trial. The pooled esti-
mate for calcitonin from the PROOF trial and the three smaller studies combined is 0.46 (95% CI 0.250.87, P = 0.02, n = 1,404).
Reproduced with permision from Cranney A, Guyatt G, Griffith L, Wells G, et al. (2002) Summary of meta-analyses of therapies for post-
menopausal osteoporosis. Endocrine Reviews 23(4):570578. Copyright 2002, The Endocrine Society.

Table 8.2. Magnitude of Effect on Nonvertebral Fractures


Intervention No. of Trials/Patients Relative Risk (95% CI) Relative Risk, Heterogeneity,
P-value P-value
Calcium 2 (222) 0.86 (0.43, 1.72) 0.66 0.54
Vitamin D 6 (6187) 0.77 (0.57, 1.04) 0.09 0.09
Etidronate 7 (867) 0.99 (0.69, 1.42) 0.97 0.94
Alendronate (5 mg) 8 (8603) 0.87 (0.73, 1.02) 0.09 0.31
Alendronate (1040 mg) 6 (3723) 0.51 (0.38, 0.69) <0.01 0.88
Raloxifene 2 (6961) 0.91 (0.79, 1.06) 0.24 0.43
Calcitonina 1 (1245) 0.80 (0.59, 1.09) 0.16 n/a
Risedronate 7 (12958) 0.73 (0.61, 0.87) <0.01 0.81
HRT 6 (3986) 0.87 (0.71, 1.08) 0.10 0.57
Fluoride 5 (950) 1.46 (0.92, 2.32) 0.11 0.06
a
Due to the potential for publication bias, we are presenting the estimate for the larger RCT estimate from the PROOF trial. The pooled esti-
mate for calcitonin from the PROOF trial and the three smaller studies combined is 0.52 (95% CI 0.221.23, P = 0.14, n = 1,481).
Reproduced with permision from Cranney A, Guyatt G, Griffith L, Wells G, et al. (2002) Summary of meta-analyses of therapies for post-
menopausal osteoporosis. Endocrine Reviews 23(4):570578. Copyright 2002, The Endocrine Society.

advantages over the more traditional estrogen- tion and survival of the osteoclast. The receptor
like compounds [43]. activator of NFkB (RANK) is found on the
Cathepsin K is a cysteine protease that is osteoclast and is a specic mediator of osteo-
highly expressed in osteoclasts and degrades clast differentiation, function and survival. The
bone matrix proteins, including collagen. Muta- ligand (RANKL) is secreted by the osteoblast
tions of the gene that regulates cathepsin K and the binding of RANK to RANK ligand
result in mouse models consistent with pheno- results in an increase in the number and activ-
typic features of pcynodysostosis with ity of osteoclasts. Osteoprotegerin (OPG) is
increased bone density. Several areas are tar- a decoy receptor that interferes with the bind-
geted on this pathway, since loss of cathepsin K ing of RANK ligand to its receptor RANK.
function results in a decrease in osteoclast- Interference with any steps of this pathway
mediated bone resorption. In addition, estrogen results in decreased activity and differentiation
down-regulates cathepsin K expression, result- of osteoclasts. Data from a recent phase 1
ing in bone loss after the menopause [76]. trial that tested an antibody to RANK ligand
Other agents interfere with physiologic path- in postmenopausal women have shown a
ways responsible for the differentiation, activa- rapid, dose-dependent decrease in urinary
148 Bone Resorption

NTX/creatinine and serum NTX in healthy 12. Chesnut CH, III, Silverman S, Andriano K, et al. (2000)
women [4]. A randomized trial of nasal spray salmon calcitonin in
postmenopausal women with established osteoporosis:
Other targets directed toward the osteoclast The prevent recurrence of osteoporotic fractures study.
include the alpha v beta 3 integrins that are American Journal of Medicine 109:26776.
responsible for the attachment of osteoclasts to 13. Cosman F, Nieves J, Woelfert L, et al. (2001) PTH added
bone, or interference with the GM-CSF differ- to established hormone therapy: effects on vertebral
fracture and maintenance of bone mass after PTH with-
entiation pathway. Several compounds block drawal. JBMR 16(5):92532.
osteoclast function or activity by interfering 14. Cosman F, Nieves J, Woelfert L, et al. (1998) Alendronate
with the actin ring at the site of osteoclast does not block the anabolic effect of PTH in post-
attachment to bone. menopausal osteoporotic women. JBMR 13(6):1051
5.
15. Cranney A, Tugwell P, Wells G, Guyatt G, et al. (2002)
Systematic reviews of randomized trials in osteoporo-
sis: introduction and methodology. Endocrine Reviews
References 23(4):497507.
16. Cranney A, Wells G, Willan A, Grifth L, et al. (2002)
Meta-analysis of alendronate for the treatment of
1. Alexandersen P, Riis BJ, Stakkestad JA, Delmas PD, postmenopausal women. Endocrine Reviews 23(4):
Christiansen C (2001) Efcacy of levormeloxifene in the 50816.
prevention of postmenopausal bone loss and on the 17. Cranney A, Tugwell P, Adachi J, Weaver B, et al. (2002)
lipid prole compared to low dose hormone replace- Meta-analysis of risedronate for the treatment of post-
ment therapy. JCEM 86:75560. menopausal osteoporosis. Endocrine Reviews 23(4):
2. Anonymous (1996) Effects of hormone therapy on 51723.
BMD: results from the postmenopausal estrogen/prog- 18. Cranney A, Tugwell P, Zytaruk N, Robinson V, et al.
estin interventions (PEPI) trial. The writing group for (2002) Meta-analysis of raloxifene for the prevention
the PEPI. Journal of the American Medical Association and treatment of osteoporosis. Endocrine Reviews
276:138996. 23(4):5248.
3. Astrom E, Soderhall S (2002) Benecial effect of long- 19. Cranney A, Tugwell P, Zytaruk N, Robinson V, et al.
term intravenous bisphosphonates treatment of osteo- (2002) Meta-analysis of calcitonin for the treatment
genesis imperfecta. Arch Dis Child 86:35664. of postmenopausal osteoporosis. Endocrine Reviews
4. Bekker PJ, Holloway DL, Rasmussen AS, Murphy R, 23(4):54051.
et al. (2004) A single-dose placebo-controlled study of 20. Cranney A, Guyatt G, Grifth L, Wells G, et al. (2002)
AMG 162, a fully human monoclonal antibody to Summary of meta-analyses of therapies for post-
RANKL, in postmenopausal women. JBMR 19:1059 menopausal osteoporosis. Endocrine Reviews 23(4):
66. 5708.
5. Bergstrom JD, Bostedor RG, Masarachia PJ et al (2000) 21. Delmas PD, Recker RR, Chestnut CH, et al. (2004) Daily
Alendronate is a specic, nanomolar inhibitior of far- and intermittent oral ibandronate normalize bone
nesyl diphosphate synthase. Arch Biochem Biophys turnover and provide signicant reduction in vertebral
373:23141. fracture risk: results from the BONE study. Osteoporo-
6. Bianchi ML, Cimaz R, Bardare M, et al. (2000) Efcacy sis International 15:7928.
and safety of alendronate for the treatment of osteo- 22. Eastell R, Adachi J, Harper K, Sarkar S, Delmas PD,
porosis in diffuse connective tissue diseases in children: Ensrud K, et al. (2000) The effects of raloxifene on inci-
a prospective multicenter study. Arthritis Rheum 43: dent vertebral fractures in postmenopausal women with
19606. osteoporosis: 4-year results from the MORE trial. JBMR
7. Black DM, Greenspan SL, Ensrud KE, Palermo L, et al. 15:S229.
(2003) The effects of PTH and alendronate alone or in 23. Ettinger B, San Martin J, Crans G, and Pavo I (2004)
combination in postmenopausal osteoporosis. NEJM Differential effects of teriparatide on BMD after treat-
349(13):120715. ment with raloxifene or alendronate. JBMR 19(5):745
8. Black DM, Cummings SR, Karpf DB, Cauley JA, et al. 51.
(1996) Randomised trial of effect of alendronate on risk 24. Ettinger B, Black DM, Mitlak BH, Knickerbocker RK,
of fracture in women with existing vertebral fractures. Nickelsen T, Genant HK, et al. (1999) Reduction of ver-
The Lancet 348:153541. tebral fractures in postmenopausal women with osteo-
9. Bone HG, Hosking D, Devogelaer JP, Tucci JR, et al. porosis treated with raloxifene: results from a 3-year
(2004) Ten years experience with alendronate for osteo- randomized trial. JAMA 282:63745.
porosis in postmenopausal women. NEJM 350(12): 25. Ettinger B, Pressman A, Silver P (1999) Effect of age on
118999. reasons for initiation and discontinuation of hormone
10. Brumsen C, Hamdy NA, Papapoulos SE (1997) replacement therapy. Menopause 6:289.
Long-term effects of bisphosphonates on the growing 26. Ettinger M, Schwartz E, Emkey R, Moffett AF, et al.
skeleton. studies of young patients with severe osteo- Lasofoxifene, a next generation selective estrogen recep-
porosis. Medicine (Baltimore) 76:216683. tor modulator (SERM), in the prevention of bone loss in
11. Chavassieux PM, Arlot ME, Reda C, Wei L, et al. (1997) postmenopausal women. The Endocrine Societys 86th
Histomorphometric assessment of the long-term effects Annual Meeting, New Orleans, LA, June 1419, 2004:
of alendronate on bone quality and remodeling in pa- Abstract S352.
tients with osteoporosis. J Clin Invest 100:147580. 27. www.fda.gov
Pharmaceuticals for Bone Disease Targeting the Osteoclast 149

28. Field CS, Ory SJ,Wahner HW, Herrmann RR, et al. (1993) dronate: inhibition of mitochondrial ADP/ATP translo-
Preventive effects of transdermal 17 beta-estradiol on case by a nonhydrolyzable, adenine-containing metabo-
osteoporotic changes after surgical menopause: a two- lite. Mol Pharmacol 61:125562.
year placebo-controlled trial. Am J Obstet Gynecol 45. Lieberman UA, Weiss SR, Brll J, Minne HW, et al. (1995)
168:11421. Effect of oral alendronate on BMD and the incidence
29. Filipponi P, Cristallini S, Rizzello E, Policani G, et al. of fractures in postmenopausal osteoporosis. NEJM
(1996) Cyclical intravenous clodronate in post- 333(22):143743.
menopausal osteoporosis: results of a long-term clinical 46. Lindsay R, Nieves J, Formica C, et al. (1997) Randomised
trial. Bone 18:17984. controlled study of effect of PTH on vertebral-bone
30. Finkelstein J, Hayes A, Hunzelman J, et al. (2003) The mass and fracture incidence among postmenopausal
effects of PTH, alendronate, or both in men with osteo- women on oestrogen with osteoporosis. The Lancet
porosis. NEJM 349(13):121626. 350:5505.
31. Gallagher JC (2001). Role of estrogens in the manage- 47. Love RR, Mazess RB, Barden HS, Epstein S, Newcomb
ment of postmenopausal bone loss. Rheum Dis Clin PA, Jordan VC, et al. (1992) Effects of tamoxifen on BMD
North Am 27:14362. in postmenopausal women with breast cancer. NEJM
32. Giannini S, DAngelo A, Sartori L. Passeri G, et al. (1996) 326:8526.
Continuous and cyclical clondronate therapies and 48. Luckman SP, Hughes DE, Coxon FP, et al. (1998)
bone density in postmenopausal bone loss. Obstet Nitrogen-containing bisphosphonates inhibit the
Gynecol 88:4316. mevalonate pathway and prevent post-translational
33. Glorieux FH, Bishop NJ, Plotkin H, et al. (1998) Cyclic prenylation of GTP-binding proteins, including Ras.
administration of pamidronate in children with severe JBMR 13:5819.
osteogenesis imperfecta. NEJM 339:94752. 49. Lufkin EG, Riggs LB (1996) Three-year follow up on
34. Greendale GA, Wells B, Marcus R, Barrett-Connor E effects of transdermal estrogen. Ann Intern Med
(2000) How many women lose BMD while taking 125(1):77.
hormone replacement therapy? Results from the post- 50. Lufkin EG, Wahner HW, OFallon WM, Hodgson SF,
menopausal estrogen/progestin interventions trial. et al. (1992) Treatment of postmenopausal osteoporosis
Arch Intern Med 160:306571. with transdermal estrogen. Ann Intern Med 117(1):19.
35. Greenspan SL, Resnick NM, and Parker RA (2003) 51. Marcus R, Holloway L, Wells B, Greendale G, James MK,
Combination therapy with hormone replacement and Wasilauskas C, et al. (1999) The relationship of bio-
alendronate for prevention of bone loss in elderly chemical markers of bone turnover to bone density
women. JAMA 289(19):252533. changes in postmenopausal women: results from the
36. Greenspan SL, Emkey RD, Bone HG, Weiss SR, et al. postmenopausal estrogen/progestin interventions
(2002) Signicant differential effects of alendronate, (PEPI) trial. JBMR 14:158395.
estrogen, or combination therapy on the rate of bone 52. Markowitz GS, Appel GB, Fine PL, Fenves AZ, et al.
loss after discontinuation of treatment of post- (2001) Collapsing focal segmental glomerulosclerosis
menopausal osteoporosis. Ann Intern Med 137:87583. following treatment with high-dose pamidronate.
37. Harris ST, Watts NB, Genant HK, McKeever CD, et al. Journal of the American Society of Nephrology 12(6):
(1999) Effects of risedronate treatment on vertebral and 116472.
nonvertebral fractures in women with postmenopausal 53. Marx RE (2003) Pamidronate (Aredia) and zoledronate
osteoporosis. JAMA 282(14):134452. (Zometa) induced avascular necrosis of the jaws: a
38. Heaney RP, Saville PD (1976). Etidronate disodium in growing epidemic. Journal of Oral & Maxillofacial
postmenopausal osteoporosis. Clin Pharm Therap 20: Surgery 61(9):111517.
593604. 54. McCloskey E, Selby P, Davies M, Robinson J, et al. (2004)
39. Ibrahim A, Scher N, Williams G, Sridhara R, et al. (2003) Clodronate reduces vertebral fracture in women with
Approval summary for zoledronic acid for treatment of postmenopausal or secondary osteoporosis: results of a
multiple myeloma and cancer bone metastases. Clinical double-blind, placebo-controlled 3-year study. JBMR
Cancer Research. 9(7):23949. 19(5):72836.
40. Johnston CCJr, Bjarnason NH, Cohen FJ, Shah A, 55. McClung MR, Geusens P, Miller PD, Zippel H, et al.
Lindsay R, Mitlak BH, et al. (2000) Long-term effects of (2001) Effect of risedronate on the risk of hip fracture
raloxifene on BMD, bone turnover, and serum lipid in elderly women. NEJM 344(5):33340.
levels in early postmenopausal women: three-year data 56. Naessen T, Berglund L, Ulmsten U (1997) Bone loss
from 2 double-blind, randomized, placebo-controlled in elderly women prevented by ultralow doses of
trials. Arch Intern Med 160:344450. parenteral 17 beta-estradiol. Am J Obstet Gynecol
41. Khosla S (2003) PTH plus alendronate a combination 177:11519.
that does not add up. NEJM 349(13):12779. 57. Orwoll E, Ettinger M, Weiss S, Miller P, et al. (2000)
42. Kostenuik PJ and Shalhoub V (2001) Osteoprotegerin: a Alendronate for the treatment of osteoporosis in men.
physiological and pharmacological inhibitor of bone NEJM 343:60410.
resorption. Current Pharmaceutical Design 7(8):613 58. Pecherstorfer M, Ludwig H, Schlosser K, et al. (1996)
35. Administration of the bisphosphonate ibandronate
43. Kousteni S, Han L, Chen J, Almeida M, Plotkin LI, et al. (BM 21.0955) by intravenous bolus injection. JBMR
(2003) Kinase-mediated regulation of common tran- 11(5):58793.
scription factors accounts for the bone-protective 59. Peretz A, Body JJ, Dumon JC, et al. (1996) Cyclical
effects of sex steroids. Journal of Clinical Investigation pamidronate infusions in postmenopausal osteoporo-
111(11):165164. sis. Maturitas 25:6975.
44. Lehenkari PP, Kellinsalmi M, Napankangas JP, et al. 60. Pols HA, Felsenberg D, Hanley DA, et al. (1999)
(2002) Further insight into mechanism of action of clo- Multinational, placebo-controlled, randomized trial of
150 Bone Resorption

the effects of alendronate on bone density and fracture postmenopausal bone loss. Am J Obstet Gynecol 156:
risk in postmenopausal women with low bone mass: 615.
results of the FOSIT study. Osteoporosis International 9: 71. Sorensen OH, Crawford GM, Mulder H, Hosking DJ,
4618. et al. (2003) Long-term efcacy of risedronate: a 5-
61. Prestwood KM, Gunness M, Muchmore DB, Lu Y, Wong year placebo-controlled clinical experience. Bone 32:
M, Raisz LG (2000) A comparison of the effects of ralox- 1206.
ifene and estrogen on bone in postmenopausal women. 72. Storm T, Thamsborg G, Steiniche T, et al. (1990) Effect
JCEM 85:2197202. of intermittent cyclical etidronate therapy on bone mass
62. Ravn P, Weiss SR, Rodriquez-Portales JA, McClung MR, and fracture rate in women with postmenopausal osteo-
et al. (2000) Alendronate in early postmenopausal porosis. NEJM 322:126571.
women: effects on bone mass during long-term treat- 73. Thibaud D, Burckhardt P, Kriegbaum H, et al. (1997)
ment and after withdrawal. JCEM 85(4):14927. Three monthly intravenous injections of ibandronate in
63. Ravn P, Clemmesen B, Riis BJ, Christiansen C (1996) The the treatment of postmenopausal osteoporosis. The
effect on bone mass and bone markers of different doses American Journal of Medicine 103:298307.
of ibandronate: a new bisphosphonate for prevention 74. Valimaki M, Laitinen K, Patronen A, Puolijoki H, et al.
and treatment of postmenopausal osteoporosis: a 1- (2002) Prevention of bone loss by clondronate in early
year, randomized, double-blind, placebo-controlled postmenopausal women with vertebral osteopenia: a
dose-nding study. Bone 19(5):52733. dose-nding study. Osteoporosis International 13:
64. Recker RR, Stakkestad JA, Felsenberg D, et al. (2000) A 93747.
new treatment paradigm: quarterly injections of iban- 75. VanBeek E, Pieterman E, Cohen L, et al. (1999)
dronate reduce the risk of fractures in women with post- Nitrogen-containing bisphosphonates inhibit isopen-
menopausal osteoporosis: results of 3-year trial. tenyl pyrophosphate isomerase/farnesyl pyrophosphate
Osteoporosis International 11(Suppl. 2):S209. synthase activity with relative potencies corresponding
65. Reginster JY, Christiansen C, Roux C, et al. (2001) Inter- to their antiresorptive potencies in vitro and in vivo.
mittent cyclic tiludronate in the treatment of osteo- Biochem Biophys Res Commun 255:4914.
porosis. Osteoporosis International 12(3):16977. 76. Votta BJ, Levy MA, Badger A, Bradbeer J, et al. (1997)
66. Reginster JY, Minne HW, Sorensen O, Hooper M, Roux Peptide aldehyde inhibitors of cathepsin K inhibit bone
C, Brandi ML, et al. (2000) Randomized trial of the resorption both in vitro and in vivo. JBMR 12:1396
effects of risedronate on vertebral fractures in women 406.
with established postmenopausal osteoporosis. 77. Watts NB, Harris ST, Genant HK, et al. (1990)
Osteoporosis International 11:8391. Intermittent cyclical etidronate treatment of post-
67. Reid IR, Brown JP, Burckhardt P, Horowitz Z, et al. (2002) menopausal osteoporosis. NEJM 323(2):739.
Intravenous zoledronic acid in postmenopausal women 78. Wells G, Tugwell P, Shea B, Guyatt G, et al. (2002)
with low BMD. NEJM 346(9):65361. Meta-analysis of the efcacy of hormone replacement
68. Richelson LS, Wahner HW, Melton LJ, III, Riggs BL therapy in treating and preventing osteoporosis in
(1984) Relative contributions of aging and estrogen postmenopausal women. Endocrine Reviews 23(4):529
deciency to postmenopausal bone loss. NEJM 311: 39.
12735. 79. Whyte MP, Wenkert D, Clements KL, McAllister WH,
69. Riis BJ, Ise J, Von Stein T, et al. (2001) Ibandronate: a Mumm S (2003) Bisphosphonate-induced osteoporosis.
comparison of oral daily dosing versus intermittent NEJM 349(5):45763.
dosing in postmenopausal osteoporosis. JBMR 16(10): 80. The Womens Health Initiative Steering Committee
18718. (2004) Effects of conjugated equine estrogen in post-
70. Riis BJ, Thomsen K, Strom V, Christiansen C (1987) The menopausal women with hysterectomy. JAMA 291(14):
effect of percutaneous estradiol and progesterone on 170112.
9.
Skeletal Complications of Malignancy:
Central Role for the Osteoclast
Gregory A. Clines, John M. Chirgwin, and Theresa A. Guise

Introduction Hypercalcemia of Malignancy


Skeletal complications of malignancy are Malignancy is the most common cause of
diverse and include hypercalcemia of malig- hypercalcemia in the hospitalized patient, and
nancy, bone metastases, and oncogenic osteo- malignancy-associated hypercalcemia is one of
malacia [78]. Of these syndromes, bone the more common paraneoplastic syndromes.
metastases are most common, followed by Up to 30% of patients with cancer may develop
hypercalcemia of malignancy. Although the hypercalcemia during the course of their
pathophysiology of these skeletal complications disease [75, 78]. Hypercalcemia of malignancy
of cancer is diverse, it is clear that the osteoclast may be due to (1) factors, secreted by tumors,
is a major player in the development and pro- that act systemically on target organs of bone,
gression of both malignant hypercalcemia and kidney, and intestine to disrupt normal calcium
bone metastases. As a result of the widespread homeostasis; (2) local factors secreted by
treatment with bisphosphonates, hypercalcemia tumors in bone, either metastatic or hemato-
of malignancy is much less prevalent than it was logical, which directly stimulate osteoclastic
10 years ago and new therapies for bone metas- bone resorption; and (3) coexisting primary
tases have become the focus of current research. hyperparathyroidism.
Furthermore, cancer survival has increased as a The rst two of these situations should be
result of advances in treatment. Because of the viewed as constituting the opposite ends of a
longer lifespan of cancer patients, a new skele- continuous spectrum, rather than as completely
tal complication of malignancy, referred to as unrelated pathologies. Clearly the pathophysiol-
cancer-treatment-induced bone loss, is likely to ogy of hypercalcemia in patients with solid
be the most common skeletal complication of tumors and no bone metastases at one end of
malignancy in the coming years. Similar to the spectrum, and myeloma associated with
hypercalcemia and bone metastases, the patho- extensive local bone destruction adjacent to the
physiology of this complication of cancer also tumor cells at the other, is very different.
intimately involves the osteoclast. Oncogenic However, in between these extremes are
osteomalacia remains relatively rare and will hypercalcemic patients with squamous cell car-
not be addressed in this chapter. cinomas in whom hypercalcemia may occur,
accompanied by not very extensive, osteolytic
bone metastases, and hypercalcemic patients
with advanced breast carcinoma in whom
hypercalcemia almost never occurs unless oste-
151
152 Bone Resorption

olytic bone destruction is extensive. Separating regulation of cartilage differentiation and bone
hypercalcemia into categories on the assump- formation [129]; (2) the growth and differenti-
tion that the underlying mechanisms are dis- ation of skin [219], mammary gland [221], and
tinct is not entirely satisfactory because the teeth [149]; (3) cardiovascular function [175];
same mediator molecule may be of local or sys- (4) transepithelial calcium transport in
temic origin. However, if tumor burden is exten- mammary epithelia and placenta [108, 220]; (5)
sive, enough mediators may be generated by the relaxation of smooth muscle in uterus, bladder,
tumors to have both local and systemic effects. arteries, and ileum [15, 150, 198, 223]; and (6)
This review will categorize the effects of cancer host immune function [61, 62]. The normal
on bone, but in a signicant proportion of functions of PTHrP have been extensively
cancer patients, i.e., those in the middle of this reviewed elsewhere [191]. Normal subjects do
spectrum, the effects on bone are due to both not have detectable circulating levels of PTHrP
humoral and local mediators. this suggests that, in the tissues where it is
produced, PTHrP acts locally as a regulator or
Humoral Mediators of cytokine.
Approximately 80% of hypercalcemic pa-
Hypercalcemia of Malignancy tients with solid tumors have detectable or
increased plasma concentrations PTHrP [23]. In
Parathyroid Hormone-related Protein cancer, PTHrP has these additional functions:
Hypercalcemia has been associated with malig- (1) mediating hypercalcemia; (2) aiding in the
nancy since the development of serum calcium development and progression of osteolytic bone
assays in the 1920s. Only in 1941, however, did metastasis (see below); (3) regulating cancer cell
Fuller Albright advance the hypothesis that PTH growth [112, 119]; and (4) acting as a cell sur-
is ectopically produced by certain tumors. Over vival factor [28].
the subsequent ve decades investigators deter-
mined that PTH was not, in fact, the cause for Modulation of PTHrP Effects by Other
most cases of humoral hypercalcemia of malig-
nancy (HHM), but that a PTH-like factor was
Tumor-associated Factors
responsible. In 1987 this PTH related-protein Cytokines and comparable tumor-associated
(PTHrP) was isolated from human lung cancer factors modulate the actions of PTH and PTHrP
[131], breast cancer [24], and renal cell carci- in similar fashions. Both PTH and PTHrP
noma [190], and was cloned shortly thereafter induce a comparable degree of hypercalcemia,
[194]. as well as increases in osteoclastic bone resorp-
PTHrP shares 70% sequence homology with tion, more committed marrow mononuclear
PTH over the rst 13 amino acids at the N- osteoclast precursors, and mature osteoclasts
terminus. Beyond the initial amino-terminal [206]. However, the multipotent osteoclast pre-
sequence the protein is unique and shows no cursors, the granulocyte/macrophage colony-
further sequence homology with PTH. The forming units, were not stimulated. IL-6
PTHrP protein is larger than PTH and three dis- potentiated the hypercalcemia and bone resorp-
tinct isoforms of 139, 141, and 173 amino acids tion mediated by PTHrP in vivo by stimulating
have been identied. Similarly, the human production of early osteoclast precursors [41].
PTHrP gene is much larger and more complex Likewise, TGFa has been shown to enhance the
than the PTH gene, spanning 15 kb of genomic hypercalcemic effects of PTHrP in an animal
DNA with nine exons and three promoters. model of malignancy-associated hypercalcemia
These differences not withstanding, both PTH [81] and to modulate the renal and bone effects
and PTHrP bind to a common PTH/PTHrP of PTHrP. Sato et al. [172] demonstrated that IL-
receptor [1, 66] and share similar biological 1a and PTHrP may have synergistic effects in
activities [86]. vivo, and others have shown that IL-1 may mod-
PTHrP has been detected in a variety of ulate the renal effects of PTHrP [202]. Finally,
tumor types as well as in normal tissue. The Uy et al. [207] have demonstrated that tumor
widespread expression of PTHrP in normal necrosis factor-a (TNFa) enhanced the hyper-
tissue was the rst evidence that the hormone calcemic effect of PTHrP by increasing the pool
had a role in normal physiology. In addition to of committed osteoclast progenitors with a sub-
its PTH-like effects, PTHrP plays a role in the (1) sequent increase in osteoclastic bone resorp-
Skeletal Complications of Malignancy: Central Role for the Osteoclast 153

tion. Bone formation parameters indicate that appears to have both paracrine and endocrine
TNFa did not inhibit the new bone formation functions.
stimulated by PTHrP in nude mice. PTHrP was detected by immunohistochemi-
Such tumor-associated factors also appear to cal staining in 60% of 102 invasive breast
be important regulators of PTHrP expression tumors that had been removed from normocal-
and secretion by tumors [78]. EGF has been cemic women. It is not found in normal breast
shown to stimulate PTHrP expression in a ker- tissue [186].At least four other studies have con-
atinocyte and a mammary epithelial cell line, rmed these percentages [21, 22, 104, 114]. One
whereas TGFa enhanced PTHrP expression in a of these reported the presence of immunoreac-
human squamous cell carcinoma of the lung. tive PTHrP within the cytoplasm of lobular and
Interleukin-6, TNF, IGF-I, and IGF-II increased ductal epithelial cells in normal and brocystic
the production of PTHrP in vitro by a human breast tissues [114]. Furthermore, 6592% of
squamous cell carcinoma. TGFb, which is hypercalcemic breast cancer patients (with or
abundant in bone, is released in active form without bone metastases) had plasma PTHrP
by resorbing bone, and is expressed by some concentrations at a level similar to that in
breast cancers and cancer-associated stromal patients whose hypercalcemia was the result of
cells, has been shown to enhance secretion nonbreast tumors [21, 74]. Thus, PTHrP is an
and stabilization of PTHrP message in renal important mediator of hypercalcemia in breast
and squamous cell carcinomas. The role of cancer. It is also a major mediator of bone
TGFb in the regulation of PTHrP will be dis- destruction in breast cancer that has metasta-
cussed below. sized to bone (see also below).

PTHrP in Hypercalcemia Associated with PTHrP in Hypercalcemia Associated with


Breast Cancer Hematologic Malignancies
The role of PTHrP in breast cancer-associated Hypercalcemia associated with hematologic
hypercalcemia deserves special consideration. malignancies results both from systemically
Breast cancer affects bone predominantly produced tumor factors such as 1,25-(OH)2D3
through metastatic mechanisms, typically (discussed below), and from locally produced
leading to lytic deposits in the skeleton. Approx- cytokines, such as IL-6, IL-1, and lymphotoxin
imately 10% of women with breast cancer will or TNFb, that have their origin in bone tumors.
at some point in their disease have hypercal- PTHrP has recently been shown to be an impor-
cemia. The association of hypercalcemia with tant pathogenetic factor in the development of
extensive osteolytic lesions in breast cancer is so hypercalcemia in some patients with hemato-
strong that the presentation of hypercalcemia logic malignancies. In a clinical study of 76
without bone metastases suggests coexistence of patients with various hematological malignan-
primary hyperparathyroidism [53]. It was there- cies, 50% of the fourteen hypercalcemic patients
fore long held that breast cancer-associated had signicant increases in plasma PTHrP
hypercalcemia is the result of excessive resorp- concentrations [109]. Of these, ve had non-
tion of bone around tumor deposits. However, Hodgkins lymphoma, one had Hodgkins
clinical studies [154] failed to demonstrate any disease, and one had multiple myeloma. The
relationship between extent of bone metastasis serum 1,25-(OH)2D3 concentrations were low in
and serum calcium levels. Conversely, studies the patients with non-Hodgkins lymphoma
exploring the potential role for a humoral medi- patients whose plasma PTHrP concentrations
ator of hypercalcemia in breast cancer have were increased. Also of interest is that plasma
clearly demonstrated altered renal handling of PTHrP concentrations were increased in several
calcium and phosphate, as well as an increase in normocalcemic patients with non-Hodgkins
nephrogenous camp. This suggests that at least lymphoma, Hodgkins lymphoma, multiple
some of hypercalcemic breast cancer patients myeloma, and Waldenstroms macroglobuline-
have a circulating factor with PTH-like proper- mia [177]. Using a sensitive two-site immuno-
ties. The identication of PTHrP as this factor is radiometric assay, other investigators have
hardly surprising when it is recalled that PTHrP noted increased plasma PTHrP concentrations
may play an important role in normal breast in patients with adult T-cell leukemia and B-cell
physiology. In the case of breast cancer, PTHrP lymphoma [92]. Finally, in separate studies,
154 Bone Resorption

plasma concentrations of PTHrP similar to tions tend to be low and indomethacin therapy
those in HHM were encountered in two of four did not seem to lower plasma calcium [54].
hypercalcemic patients with non-Hodgkins Thus, the mechanisms responsible for hyper-
lymphoma, in three of nine with myeloma, and calcemia in lymphoma are multifactorial,
in a patient with myeloid blast crisis of chronic including an increase in intestinal calcium
myeloid leukemia [54, 178]. Thus, the humoral absorption and in osteoclastic bone resorption.
mediators in the hypercalcemia associated with Many of the patients also had altered renal
hematological malignancies include both 1,25- function; this suggests that impaired renal
(OH)2D3 and PTHrP. calcium clearance may contribute to the hyper-
calcemia. It is likely that in lymphoma, as in
granuloma, the diseased tissue hydroxylates 25-
1,25-dihydroxyvitamin D hydroxyvitamin D to the active 1,25-(OH)2D3.
Under normal physiologic conditions, serum 1a-Hydroxylase activity has been demonstrated
calcium and 1,25-(OH)2D3 concentrations bear in human T-cell lymphotrophic virus type-I-
an inverse relationship to one another. In hyper- transformed lymphocytes, as well as in other
calcemia, serum 1,25-(OH)2D3 concentrations extrarenal tissues [51, 230]. None of the patients
are suppressed unless there is persistent sti- with 1,25-(OH)2D3-mediated hypercalcemia had
mulation of 1a-hydroxylase activity from an granulomatous disease. Their hypercalcemia
autonomous source of PTH, as in primary was often reduced as a result of medical or sur-
hyperparathyroidism. Other than that the gical therapy that led to a decrease in their
failure of 1,25-(OH)2D3 to go down as calcium serum 1,25-(OH)2D3 concentration. Recurrence
goes up indicates abnormal regulation of 1,25- of disease is associated with recurrent hyper-
(OH)2D3 synthesis and suggests that 1,25- calcemia and increased plasma 1,25-(OH)2D3
(OH)2D3 may be produced extrarenally, as is concentrations [127].
the case in granulomatous disease. In that
condition, activated macrophages within the
granuloma synthesize 1,25-(OH)2D3 [2, 71, 125].
Parathyroid Hormone (PTH)
Similarly, 1,25-(OH)2D3 seems to be the major For many years after Fuller Albrights observa-
factor causing hypercalcemia in Hodgkins tions in 1941 malignancy-associated hypercal-
disease, non-Hodgkins lymphoma, and other cemia was attributed to PTH produced by an
hematological malignancies [177, 178]. The ectopic tumor. It is now clear that in the major-
mechanism is similar to that observed in ity of cases of HHM, PTHrP is the ectopically
granulomatous disease. In this scenario, hyper- produced factor. There have been rare cases,
calcemic patients have increased plasma 1,25- however, when the hypercalcemia was due to
(OH)2D3 concentrations, even though their authentic, tumor-produced PTH. Ectopic PTH
plasma PTH levels and urinary cAMP output production has been documented in small cell
are low to normal [164]. There is no bone carcinoma of the lung [229], in squamous cell
involvement. Lymphoma patients exhibit an carcinoma of the lung [142], in ovarian cancer
increase in urinary calcium excretion [164], as [143], in widely metastatic primitive neuroecto-
well as in intestinal calcium absorption, as dermal tumor [189], in papillary adenocarci-
measured with the aid of radiocalcium [19]. noma of the thyroid [90], and in thymoma
Increased 1,25-(OH)2D3 concentrations were [160]. Molecular analysis of the ovarian carci-
also noted in 12 of 22 hypercalcemic patients noma revealed both DNA amplication and
with non-Hodgkins lymphoma. In addition, rearrangement in the upstream regulatory
71% of 22 normocalcemic patients with non- region of the PTH gene. Interestingly, the prim-
Hodgkins lymphoma were hypercalciuric, and itive neuroectodermal tumor produced both
18% had increased serum 1,25-(OH)2D3 concen- PTH and PTHrP that resulted in severe hyper-
trations. These ndings led the investigators to calcemia. These patients did not have coexisting
conclude the dysregulated 1,25-(OH)2D3 pro- primary hyperparathyroidism, because their
duction is common in patients with diffuse parathyroid glands were normal on neck explo-
large cell lymphoma [178]. The low serum PTH ration or autopsy. Nevertheless, ectopic produc-
and urinary cAMP concentrations indicate that tion of PTH is rare. Indeed most patients with
neither PTH nor PTHrP mediates the hypercal- malignancy-associated hypercalcemia have low
cemia in this setting. Prostaglandin concentra- plasma PTH concentrations [188]. Therefore
Skeletal Complications of Malignancy: Central Role for the Osteoclast 155

hypercalcemia of malignancy where the serum indicates that the prostaglandin EP2 receptor
PTH concentration is normal or increased is mediates PGE2-mediated hypercalcemia in
most often the result of hyperparathyroidism. mice [113].
Recently RANK ligand has been directly
Other Tumor-associated Factors implicated in the pathogenesis of hypercalcemia
of malignancy. A novel cDNA that encodes a
There is accumulating evidence that solid secreted form of an osteoclast differentiation
tumors may produce other humoral factors, factor/tumor necrosis factor that induces a
alone or in combination with PTHrP, that can cytokine was sequenced from 5 RACE cDNA
stimulate osteoclastic bone resorption and clones of squamous cell carcinoma cell lines,
cause hypercalcemia [162]. These factors SCC-4 and T3M-1 Cl.2, parental malignant
include IL-1, IL-6, TGFa, tumor necrosis factor tissues of which had caused severe humoral
(TNF) and granulocyte colony-stimulating hypercalcemia [137].
factor (G-CSF). Administration of IL-1 to mice
causes a mild hypercalcemia [17, 168], which
can be effectively blocked by the IL-1 receptor Treatment of Hypercalcemia
antagonist [77]. Mice bearing Chinese hamster of Malignancy
ovarian tumors transfected with the cDNA for
IL-6 develop mild hypercalcemia [11], as do Eradication of the underlying cancer constitutes
mice that bear renal carcinoma that co-secreted the denitive treatment of malignancy-
IL-6 and PTHrP [216]. Patients with humoral associated hypercalcemia. However, because
hypercalcemia of malignancy have high serum cure may not be possible, therapy of patients
concentrations of IL-6 [5, 205]. with symptomatic or life-threatening hyper-
Human TGFa and TNFa stimulate osteoclas- calcemia must be aimed against the mediating
tic bone resorption in vitro and cause hypercal- mechanisms. Increased osteoclastic bone
cemia in vivo [8, 89, 224]. In mice, TNFa causes resorption is present in virtually all patients
hypercalciuria, without an increase in with hypercalcemia of malignancy. Bone
nephrogenous cAMP, and increases osteoclastic resorption becomes, therefore, a key target for
bone resorption [96]. As noted in the previous treatment of hypercalcemia. Two mechanisms
section, some of these factors modulate the end- in particular, the increase in osteoclastic bone
organ effects of PTHrP on bone and kidney. resorption and renal tubular calcium reabsorp-
In some instances, factors such as TGFa, IL-1, tion, are encountered in most patients with
IL-6, and TNF enhance the hypercalcemic hypercalcemia of malignancy, even in cases not
effects of PTHrP [41, 81, 207]. The ability of associated with PTHrP production [173, 203,
IL-6 to enhance PTHrP-mediated hyper- 204]. Medical therapy should therefore be aimed
calcemia appears to be due to increased at inhibiting bone resorption and promoting
production of the early osteoclast precursors, renal calcium excretion.
granulocyte macrophage colony forming units. Because many hypercalcemic patients are
IL-6, along with the production of more com- dehydrated, this rst step of therapy is to rehy-
mitted osteoclast precursors, stimulates PTHrP, drate the patient. This will enhance calciuresis
thereby enhancing hypercalcemia [41]. by increasing the glomerular ltration rate and
Prostaglandins of the E series are powerful reduce the fractional reabsorption of calcium
stimulators of bone resorption [103], although and sodium. The use of loop diuretics, such as
their role in bone destruction associated with furosemide, to increase calcium excretion may
malignancy remains unclear [135]. Some of the exacerbate uid loss and worsen dehydration;
effects of cytokines on bone may be mediated therefore the use of loop diuretics should be
by prostaglandins. Indomethacin, a pro- limited to the volume-replete patient and only
staglandin synthesis inhibitor, has been shown then with close monitoring of volume status
to block part of the osteoclast-stimulatory [193]. Because hydration alone rarely results in
effects of IL-1 in vivo [17, 168]. Although full resolution of hypercalcemia [88], more
prostaglandins are produced by cultured tumor aggressive therapies are usually needed.
cells in vitro, indomethacin treatment of The bisphosphonates are currently the main-
malignancy-associated hypercalcemia is only stay for long-term treatment of hypercalcemia
effective on occasion [134]. Recent evidence and osteolytic bone disease. They bind to bone
156 Bone Resorption

surfaces that undergo active resorption and are When used at the recommended dose of 30
released in the bone microenvironment during 90 mg IV over 424 hours, pamidronate is highly
remodeling. Bisphosphonates decrease osteo- effective in normalizing serum calcium concen-
clastic bone resorption by two mechanisms (cf. trations and does not cause mineralization
Chapter 8). The nitrogen-substituted bisphos- defects [87].
phonates, such as alendronate, risedronate and The clinical response to bisphosphonates is
zoledronic acid, are potent inhibitors of the somewhat delayed. In studies that used a 90-mg
enzyme farnesyldiphosphate synthase, thereby infusion of pamidronate over four hours the
blocking protein isoprenylation. The prenyla- mean time to achieve normocalcemia was
tion of small GTP-binding proteins is important approximately four days, with the resulting
for maintaining structural integrity of the normocalcemia lasting 28 days [153]. By using
osteoclast: without it, the osteoclasts undergo pamidronate in combination with calcitonin,
apoptosis. The non-nitrogen-containing bis- one can achieve a rapid and sustained reduction
phosphonates, clodronate and etidronate, are in serum calcium [197].
less potent and induce osteoclastic apoptosis, Zoledronic acid, the most potent bisphospho-
but by a mechanism that differs from that of the nate to date, has been approved for the treat-
nitrogen-substituted bisphosphates. The non- ment of hypercalcemia of malignancy. This
nitrogen-containing bisphosphonates are incor- bisphosphonate, at a dose of 4 mg and 8 mg,
porated into nonhydrolyzable ATP analogs that when compared to a single infusion of 90 mg
inhibit ATP-dependent intracellular enzymes. pamidronate, was superior with respect to the
The FDA has approved the use of bisphospho- time needed to normalize plasma calcium and
nates to treat myeloma osteolytic bone disease the duration of response [121]. The FDA-
in 1995 and osteolytic bone disease due to breast approved dose is 4 mg, as 8 mg induced some
cancer in 1996. renal dysfunction.
Several lines of evidence indicate that bis- Calcitonin inhibits osteoclastic bone resorp-
phosphonates affect osteoclastic bone resorp- tion and renal tubular calcium reabsorption
tion through their actions on osteoblasts. within minutes of administration; this makes it
Bisphosphonates have multiple actions on an excellent agent for the acute treatment of
osteoblasts such as (1) modulation of prolifera- hypercalcemia. Unfortunately, tachyphylaxis
tion and differentiation [156]; (2) prevention of frequently develops within 48 hours as a result
apoptosis [151]; (3) modulation of extracellular of down regulation of the calcitonin receptor.
matrix protein production [69, 102]; and (4) Concomitant use of glucocorticoids, however,
regulation of the expression and excretion of IL- can prolong treatment duration [9]. This
6 [70, 201]. Recently, Viereck et al. [212] pre- appears to result from a glucocorticoid-
sented in vitro evidence that bisphosphonates mediated up-regulation of cell-surface calci-
act directly on human osteoblasts causing them tonin receptors and an increase in the de novo
to increase the production of both osteoprote- production of calcitonin receptors in the osteo-
gerin (OPG) mRNA and protein. It seems clast [213]. Calcitonin can be administered
reasonable to infer that by increasing OPG every 612 hours in doses of 48 U/kg. Human
expression, the RANKL mediated stimulation of calcitonin is available, but salmon calcitonin is
osteoclasts can be neutralized. more generally used. If salmon calcitonin is to
Available bisphosphonates vary in potency, be used, then a test dose of 1 unit should be
but all are poorly absorbed and are most effec- administered rst, to check for a possible ana-
tive for treatment of hypercalcemia when used phylactic reaction.
intravenously. Intravenous etidronate, the least Two antineoplastic agents have been used to
potent of the class, normalized calcium concen- treat malignancy-associated hypercalcemia.
tration in only 30%40% of patients when given Plicamycin, or mithramycin, inhibits DNA-
in doses of 7.5 mg/kg on three consecutive days dependent RNA synthesis and is a potent
[76, 101, 182]. Oral etidronate, at dosages of inhibitor of bone resorption. The dosage used to
25 mg/kg per day for more than 6 months treat hypercalcemia (25 mg/kg) is one-tenth the
can cause bone mineralization defects [55]. usual chemotherapeutic dose and the drug
Pamidronate combines high potency with low should be infused over four hours. Its con-
toxicity and has become the agent of choice for siderable side effects include nephrotoxicity,
the treatment of hypercalcemia of malignancy. hepatotoxicity, thrombocytopenia, nausea, and
Skeletal Complications of Malignancy: Central Role for the Osteoclast 157

vomiting. Gallium nitrate is another antineo- explained by any theory of embolism alone.
plastic agent with calcium lowering effects. It is The mechanisms responsible for osteotropism
also nephrotoxic and somewhat inconvenient to are complex and involve both the breast cancer
use, as it must be administered continuously cells and the bone to which the tumors metas-
over a ve-day period. Use of these antineoplas- tasize. Paget [148] proposed the seed and soil
tic agents should therefore be restricted to cases hypothesis to explain this: When a plant goes
of hypercalcemia that are unresponsive to to seed, its seeds are carried in all directions; but
bisphosphonates. they can only grow if they fall on congenial soil.
About 30% of patients treated with glucocor- The bone microenvironment provides the
ticoids for malignancy-associated hypercal- fertile soil in which the cancer cells grow [52,
cemia respond with a drop in serum calcium 136]. The cancer cells seem to activate bone
concentration. Glucocorticoids are likely to be cells, thereby stimulating local bone destruction
most effective in hypercalcemia associated with and formation.
multiple myeloma or in hematologic malignan- Pagets century-old concept continues to
cies associated with 1,25-(OH)2D3. Glucocorti- guide the study of breast cancer metastasis and
coids inhibit osteoclastic bone resorption by our understanding of how the vicious cycle is
decreasing tumor production of locally active driven by molecules produced by bone and
cytokines, in addition to having direct tumoro- tumor cells [155]. The bone cell highlighted in
lytic effects [133]. Glucocorticoids should be this volume of Topics in Bone Biology, the osteo-
given in doses equivalent to 4060 mg of pred- clast, plays an integral role in this cycle. As indi-
nisone, daily. If no response is observed in cated below, osteoclast activity is stimulated by
10 days, then glucocorticoid therapy should be a variety of mechanisms in patients with bone
discontinued. metastases: (1) by tumor cell products; (2) by
Hypercalcemia in experimental animals is increased osteoblast activity which leads to
inhibited by antibodies to PTHrP [110]. In a increased expression of receptor activator of
search for small-molecule inhibitors of PTHrP nuclear factor kappa B ligand (RANKL); and (3)
production or effects, Gallwitz et al. [45] found by sex-steroid deciency, a frequent result of
that guanine-nucleotide analogs inhibit PTHrP cancer therapy.
expression by human tumor cells. In nude
athymic mice these compounds reduce hyper- Classification of Bone Metastases
calcemia caused by excessive PTHrP production
by a lung squamous-cell carcinoma. The PTHrP Bone metastases are typically classied by their
gene promoter may therefore be a suitable radiographic appearance as either osteolytic or
target for treating the skeletal effects of malig- osteoblastic. In reality, these classications rep-
nancy [65]. resent two ends of a spectrum as patients with
solid tumor metastases to the skeleton often
have components of both bone destruction and
Solid Tumor Metastasis new bone formation. In addition to cancers of
the breast, other tumors such as those of
to Bone prostate, lung, kidney, and thyroid, also metas-
tasize to the skeleton, and all but prostate
When cancers metastasize to bone, they often cancers cause predominantly osteolytic lesions.
cause bone destruction, or osteolysis, which Approximately 15% of patients with breast
leads to bone pain, fracture, hypercalcemia, and cancer bone metastases have osteoblastic
nerve compression. Cancer patients with bone lesions that are characterized by disorganized
metastases often now survive several years new bone formation [78]. A substantial fraction
during which they suffer bone pain and related of skeletal metastases exhibit a mixture of oste-
sequelae. Breast cancer is the solid tumor that olytic and osteoblastic responses. Although
most commonly metastasizes to bone, the cells patient material containing skeletal metastases
showing a marked afnity to grow in bone. The is seldom available for detailed analyses, recent
association of breast cancer with skeletal mor- autopsy studies demonstrate that cancer metas-
bidity was already noted in 1889, when Stephen tases to the skeleton display marked hetero-
Paget stated in a cancer of the breast the bones geneity with respect to the osteolytic and
suffer in a special way, which cannot be osteoblastic phenotypes [165]. It is also likely
158 Bone Resorption

that individual metastases change phenotypi- Frequency of Bone Metastases


cally with time. As tumor-bone interactions
alter gene expression patterns of both, the local Seventy percent of patients dying of breast
balance between bone formation and destruc- cancer have bone metastases [32]. Bone is the
tion may change. Although tumor cells can most common site of breast cancer metastases,
directly destroy or synthesize bone [48, 111, and one of the earliest organs to be affected by
115], it seems likely their stimulation of bone metastatic disease [183]. In about 20% of cases
cells is the physiologically important factor in bone metastatic lesions are isolated [16]. Skele-
the etiology of bone metastases. In advanced tal metastases are virtually incurable in breast
disease direct tumor actions may contribute to cancer patients. However, the disease, once
existing pathological bone lesions. housed in bone, is relatively indolent, with a
Osteolytic and osteoblastic lesions are most median survival of two years and ve-year sur-
often diagnosed by bone scans and radiographs. vival in nearly 40% of patients 180, 183]. Since
Biochemical markers of bone resorption and metastases to organs such as liver and brain
formation are often increased in both [56, 105]. have much poorer prognoses, it appears that
In cases of pure osteolytic disease, as in bone metastases are not a major source of sec-
myeloma, bone scans may be negative. ondary metastases to these sites.
The major organic component of bone matrix
is type I collagen. The biosynthesis of collagen
by the osteoblast releases soluble pro-peptide Skeletal Involvement in
fragments, which can serve as markers of new Malignant Disease
bone formation and osteoblastic activity. Simi-
larly, osteoclastic bone resorption releases frag- The sites within the skeleton preferentially col-
ments from cross-linked collagen, which can be onized by tumor cells appear to be those which
utilized as markers of bone destruction. Bio- receive the most abundant vascular supply.
chemical markers are used to monitor active These include regions with extensive red bone
bone remodeling in patients and their responses marrow within the axial skeleton, ribs, the prox-
to treatment [67]. Markers of bone formation imal metaphyses of the long bones, and the
include bone-specic alkaline phosphatase and spine. Solid tumors, such as in breast cancer, and
serum procollagen I amino-terminal propep- hematological malignancies show the same dis-
tide (PINP). Markers of resorption are urinary tribution of sites within bone [64]. Prostate
collagen cross-linked N-telopeptide (NTX), col- cancer displays an unusual preference for the
lagen I carboxy-terminal telopeptide (CTX), spine. This has been attributed to anatomical
pyridinolines (PYD), and deoxypyridinolines access via Batsons plexus. It is more likely that
(DPYD). Both types of markers are increased in specic gene expression patterns characteristic
prostate cancer patients with osteoblastic of this tumor and of the vertebral bones are the
metastases. Markers of resorption may increase molecular basis for the preference. Breast cancer
when bone loss is the result of adjuvant and metastases are sometimes found in the poste-
chemotherapy and cannot be used alone to rior clinoid processes of the base of the
diagnose bone metastases. NTX is not as skull. The basis for this site preference is not
sensitive as a bone scan for diagnosing bone understood.
metastases, but may be used as an auxiliary Metastasis of human tumors to specic sites
diagnostic index [60]. Biochemical markers are has been modeled in immunocompromised
useful to monitor response to bisphosphonate rodents. Tumor cells are often entrapped within
treatment, with NTX the most sensitive [10]. the capillary beds of lung or liver when injected
This area of diagnostics is still under active into the venous circulation. However, many of
development. Radionuclide complexes with the same tumor cell lines will form lesions
bone-seeking bisphosphonates provide the limited to bone if the cells are inoculated
basis for scintigraphic bone scans, an effective through the left cardiac ventricle to avoid pul-
approach for localizing osteoblastic metastases monary entrapment [3]. This animal model has
[7]. Bone scans are not impaired in patients been used to study metastases to bone with a
that have been treated with bisphosphonates variety of tumor cell lines, in particular the
[166]. human breast cancer cell line MD-MB-231
Skeletal Complications of Malignancy: Central Role for the Osteoclast 159

[228]. Similar animal models of osteoblastic by osteolytic resorption activate pain receptors
metastases have recently been described, in on nerve endings in the skeleton. Inhibitors of
which three human breast cancer cell lines were bone resorption effectively palliate bone pain.
found to metastasize to bone [226]. The serious paraneoplastic syndrome of bone
pain has only recently received scientic atten-
tion. A model of bone pain, based on the rodent
Clinical Consequences of Local model of Arguello et al. [3], has been used to
Metastatic Disease demonstrate that antiresorptive treatment can
decrease metastatic bone pain [85]. Studies
Bone pain is the most common symptom of with the model suggest that the mechanisms of
cancer metastasis to bone and may be the initial pain in bone are specic [123]. Thus, bone pain
presenting symptom. Pathological fractures caused by cancer is fundamentally different
may occur in patients with advanced disease. from inammatory pain and shows much less
Vertebral fracture as the initial presentation in response to morphine [118]. Further study of
an elderly patient with metastatic breast cancer bone pain is likely to lead to approaches that
may be misdiagnosed as age-related osteoporo- would markedly improve the quality of life of
sis. Bone marrow suppression and leukopenia patients with advanced breast cancer.
can occur with sufcient tumor burden within
bone. Hypercalcemia caused by excessive bone Steps in Tumor Cell Metastasis
destruction used to occur in up to 30% of breast
cancer patients with bone involvement, but the Extravasation and Migration
incidence of this condition has decreased as a
result of the utilization of effective bisphospho- Initial angiogenesis followed by tumor cell
nate antiresorptive agents. Spinal cord com- migration through extracellular matrix and
pression can occur as a consequence of extravasation into the bloodstream are general
osteoblastic lesions or vertebral fractures and properties of all metastatic cancer cells. These
may be devastating for the patient. Since breast steps require the expression of enzymes to
cancer patients with metastases limited to the degrade extracellular matrix and of autocrine
skeleton have a median survival of 24 years, factors to stimulate tumor cell motility [52, 124,
these patients have a high cumulative risk of 218]. Metastasis to bone is likely controlled by
skeletal-related events (SREs). Approximately interactions subsequent to the exit of tumor
60% of these patients will suffer pathological cells from the primary site. Matrix metallopro-
fractures, 20% will develop hypercalcemia, and teinases, MMPs, form a large family of proteins
10% will have spinal cord compression [117]. with roles in tumor cell invasion [120]. MMP-1,
Many patients with terminal metastatic interstitial collagenase, is necessary for the
disease suffer from severe weight loss. This initiation of bone resorption [231]. MMP
cachexia involves the loss of muscle and fat inhibitors have been tested in clinical trials, with
mass and can occur independently of anorexia disappointing results to date [38, 146]. The lack
or intestinal malabsorption [200]. Cancer of efcacy of these inhibitors may reect the
cachexia is an area that has received insufcient complexity and redundancy of the MMP family
attention from researchers. Rapid onset members and not their value as targets for
cachexia often occurs in animals with breast treatment.
cancer bone metastases [228], and weight loss
correlates with tumor burden in bone [226]. The
results suggest that bone metastases release
Chemotaxis and Homing to Bone
factors into the peripheral circulation that then Bone produces factors that stimulate primary
stimulate wasting of skeletal muscle. Effective tumor cells, such as those that originate in the
anti-cachectic therapies improve the quality of breast, to home to bone [126, 145]. The best-
life and may increase the survival of patients characterized example of such a factor is the
with advanced, metastatic disease. chemokine stromal-derived factor, SDF-1,
Bone cancer pain, whether constant or inter- which is released by bone, and its tumor-
mittent, is often intractable. In the case of oste- expressed receptor CXCR-4 [132]. The ligand-
olytic metastases, products released from bone receptor pair mediates the attraction of breast
160 Bone Resorption

and other cancer cells to specic metastatic sites described in this chapter use an animal model
including, but not limited to, bone. CXCR-4 is a in which human tumor cells are inoculated into
co-receptor for HIV. Small molecule inhibitors immuno-decient mice. Careful tumor admin-
of CXCR-4 are under development and may also istration directly into the left cardiac ventricle
function to decrease breast cancer metastases. can result in 100% incidence of bone metastases
CXCR-4 expression is increased by the vascular with many tumor cell lines. Osteolytic lesions
endothelial growth factor, VEGF [4]. The mech- are detected by X-ray as early as three weeks and
anisms of adhesion of tumor cells to bone [124, can be quantied by image analysis. Osteoblas-
126] and to bone marrow endothelium [36] have tic lesions may take up to six months to develop
been studied extensively. The surface of bone is in nude mice, and the lesions cannot be quanti-
rich in extracellular matrix components, includ- ed radiographically.
ing integrin substrates. The receptors on tumor
cells for these molecules offer opportunities for Development of Skeletal Lesions
inhibition of metastases to bone [27, 228]. The
avb3 integrin plays an important role in osteo- The growth of bone metastases is usually slow
clast binding and activation in bone and can [180], as is usually the progression from micro-
regulate breast cancer metastases [50]. Antago- scopic lesion to initial diagnosis. The initial
nists of avb3 integrin may have a therapeutic steps in the development of bone lesions have
role as inhibitors of metastasis to bone [196]. not been investigated in depth in animal
models.
Colonization of Bone Tumor cells secrete proteolytic enzymes and
acid in abundance and therefore can destroy
The initiation of a metastasis is believed to be a bone matrix in vitro [32]. During the establish-
rare and inefcient process [52], although ment of metastases, however, bone is resorbed
tumor cells are often detectable in patient bone by osteoclasts rather than by tumor cells.
marrow samples when analyzed by sensitive Histological analysis and scanning electron
PCR techniques [147]. The basic mechanisms of microscopy of osteolytic bone metastases indi-
cancer metastases to specic sites continue to cate that the bone destruction is mediated by
evolve. Cancer cells continue to mutate in vivo. the osteoclast [18, 195].
The ability of cancer cells to metastasize is
characteristic of advanced disease and could
occur only after the gradual accumulation of a Cellular Mechanisms of Bone Destruction:
necessary set of pro-metastatic mutations [52]. Bone Stromal Cells Control Osteoclast
However, recent experiments suggest that a Formation and Activity by Expression of
constellation of expressed genes necessary for
metastasis to bone preexists within the primary
RANK Ligand and Osteoprotegerin
breast tumor. [98]. The results strongly support Although the osteolytic factor, parathyroid
a multi-factorial mechanism underlying organ- hormone-related protein, PTHrP, is clearly a
specic metastases. Thus, bone-specic metas- potent stimulator of bone resorption, tumor-
tases are the consequence of the selection of produced PTHrP does not have direct actions
variants from a heterogeneous population of on cells of the osteoclastic lineage. Thomas et al.
cells within the primary tumor, plus changes in [199] showed that a recently identied osteo-
gene expression induced by bone factors. The clast differentiation factor indirectly mediates
results of Kang et al. [98] are conrmed by gene the effects PTHrP on bone resorption. The
array analyses of micro-dissected patient tumor factor acts on cells of the osteoblast
samples, comparing primary and metastatic lineage, which in turn regulate the formation,
breast cancer cells [176]. Cells of the osteoblas- activity, and lifetime of osteoclasts derived from
tic lineage appear to be the main targets of hematopoietic progenitors. The regulation of
tumor-secreted factors. In turn, bone-derived osteoclasts by osteoblasts has been recognized
transforming growth factor-b [TGFb] is a major for over 15 years, but its molecular basis has
factor regulating of tumor cell behavior in bone. been understood only in the last ve years.
There are no convenient animal models Stromal cell-expressed RANK ligand, in combi-
where primary tumors reproducibly metasta- nation with macrophage colony-stimulating
size to bone. Many of the preclinical ndings factor (M-CSF) can generate osteoclasts from
Skeletal Complications of Malignancy: Central Role for the Osteoclast 161

hematopoietic precursors in the absence of cells ity and bone destruction caused by breast
or other factors [192]. RANK ligand is a member cancer [78, 227]. As noted earlier approximately
of the TNF family and is a membrane-bound 60% of human primary breast cancers express
molecule. Opposing the actions of membrane- PTHrP [21, 22, 186]. Some of these retrospective
bound RANK ligand is soluble osteoprotegerin studies suggested that women with PTHrP-
(Opg), a secreted TNF receptor family member positive tumors were more likely to develop
whose overexpression in mice resulted in bone metastases [21, 22, 186]. However, PTHrP
osteopetrosis [181]. Conversely, mice decient expression by breast cancer at metastatic sites
in OPG are osteoporotic. OPG functions as a differs signicantly from that of the primary
soluble inhibitory binding protein for RANK site. PTHrP is expressed by greater than 90% of
ligand [192]. Both RANK ligand and OPG are breast cancer metastases to bone compared with
made by osteoblastic cells, which regulate osteo- only 17% of non-bone metastases [152, 186,
clast formation and activity by the ratio of the 211]. These observations suggest two possible
two opposing factors. Interleukin 1 and tumor explanations: that PTHrP, as a bone-resorbing
necrosis factor-a have RANK-independent factor, favors tumor growth in bone or that the
effects on osteoclastogenesis, but the quantita- bone microenvironment enhances the produc-
tive importance of these pathways is unclear, tion of PTHrP. The explanations are not mutu-
and their contributions to bone metastasis are ally exclusive. Interpretation of the clinical
unknown. A variety of organic mediators ndings is limited by the data on PTHrP expres-
(prostaglandin E2, 1,25dihydroxyvitaminD3) sion in metastatic and primary tumor tissue,
and protein factors (PTH, PTHrP, IL-6, IL-11) which were obtained from different patients.
appear to exert their osteoclastogenic actions A prospective study of over 300 breast cancer
primarily via stimulation of RANK ligand patients indicated that those whose primary
expression on the surface of cells in the tumors were PTHrP-negative were more likely
osteoblast lineage. Nearly all of these conclu- to develop bone metastases [83]. Tumor tissue
sions are based on data obtained with mice. The from both bone metastases and primary tumor
role of the RANK ligand pathway in bone was available from three patients. In these cases,
resorption stimulated by IL-1, IL-6, and TNFa the bone metastases were PTHrP-positive and
may be different in humans compared to mice the respective primary tumors were PTHrP-
[84]. The components of the central osteoclast- negative. This is the only published study to
regulating pathway, RANK, RANK ligand, and compare PTHrP expression between primary
OPG offer new opportunities for molecular and metastatic tumor in the same patient. Data
therapies against metastatic bone disease. from this small subset of patients suggest that
tumor PTHrP expression is enhanced in the
Tumor-Produced Factors Which bone microenvironment. Studies with larger
numbers of patients are needed to conrm these
Stimulate Osteoclasts observations.
Many factors that stimulate bone resorption do The clinical studies suggested a role for
so not by acting directly on osteoclasts or their PTHrP as a local mediator of bone destruction
precursors, but by indirect actions on bone by metastatic breast cancer and provided the
marrow stromal cells in the osteoblastic lineage, rationale for the following experimental studies.
involving regulation of the ratio of RANK ligand A neutralizing monoclonal antibody to PTHrP-
to osteoprotegerin [166]. Pertinent to the mo- (134) inhibited the development of breast
lecular mechanisms of osteolytic metastases cancer metastases to bone by the human breast
is experimental evidence that breast cancer- cancer cell line, MDA-MB-231, which produces
produced PTHrP mediates osteolysis via increas- small amounts of PTHrP [80]. This monoclonal
ing RANK ligand and decreasing OPG expression antibody has been humanized and placed in
by the osteoblast. Breast cancer cells express OPG clinical trials against humor hypercalcemia of
and RANK, but not RANK ligand [199]. malignancy. Examination of the long bones
from tumor-bearing mice revealed signicantly
fewer osteoclasts at the tumor-bone interface
Role of Parathyroid Hormone-related Protein and less tumor burden in animals treated with
The tumor-secreted factor PTHrP is often the PTHrP antibody compared with the con-
responsible for the osteoclast-stimulating activ- trols. Similar results were obtained with PTHrP-
162 Bone Resorption

secreting lung cancer cells [91]. Neutralizing [209]. A number of PTHrP-independent factors
PTHrP decreased not only osteoclastic bone have also been reported, including IL-8 [6].
resorption, but also tumor burden in bone. Kang et al. [144] compared less- and more-
Experimental overexpression of PTHrP by metastatic variants of a breast cancer cell line by
MCF-7 breast cancer cells, which do not express gene expression proling. They identied
PTHrP and do not cause osteolytic metastases, mRNAs whose expression strongly correlated
induced marked bone destruction and with increased bone metastasis. Five of the
increased osteoclast formation, compared to the mRNAs encoded IL-11, matrix metallopro-
controls which caused no bone metastases teinase [MMP]-1 osteopontin, connective tissue
[199]. growth factor [CTGF], and CXCR-4. MMP-1 is
The data together suggest that tumor pro- an interstitial collagenase made by osteoblasts.
duction of PTHrP is important for the estab- It cleaves collagen at a site resistant to osteo-
lishment and progression of osteolytic bone clastic enzyme hydrolysis and may be rate-
metastases. Unaddressed was the possibility limiting in normal bone resorption [231].
that the bone microenvironment induces Osteopontin plays a complex role in metastasis,
PTHrP expression. The regulation of PTHrP is including modulation of anti-tumor immune
complex, and factors such as prolactin, estrogen, responses [25, 215]. CTGF is a potent osteoblast-
epidermal growth factor (EGF), insulin, IGFs 1 stimulatory factor [171] and is expressed by
and 2, TGFa, TGF-b, angiotensin II, stretch, and tumor cells. CXCR4 is the receptor for the
src have been shown to increase expression, chemokine SDF-1 and functions in the attrac-
while glucocorticoids and the active form of tion of breast cancer cells to specic metastatic
vitamin D3 decrease it [78]. PTHrP plays a local, sites including, but not limited to bone [132].
paracrine role in breast cancer bone metastases, Kang et al. [98] found that the pro-metastatic
which occur in animals in the absence of gene set was coordinately increased in cells that
detectable increases in plasma PTHrP concen- pre-existed in the original cell population. The
trations or malignant hypercalcemia [80]. authors attempted to convert low-metastatic
Osteoclastic resorption of bone releases high MDA-MB-231 breast cancer cell line clones into
concentrations of ionized calcium and phos- ones highly metastatic to bone by overexpress-
phate from the dissolution of the bone mineral. ing each of the ve individual factors. They
The calcium-sensing receptor is a G-protein found that conversion of the cells to a pheno-
coupled, seven-transmembrane domain recep- type of aggressive bone metastasis required co-
tor, which responds to small variations in the transduction of combinations of four of the ve
concentration of extracellular calcium [222]. factors. The results strongly support a multifac-
The calcium-sensing receptor is expressed by torial mechanism underlying organ-specic
breast cancer cells and regulates tumor secre- metastases. These factors may provide addi-
tion of PTHrP [20, 170], an effect which is tional targets for therapeutic intervention to
enhanced by TGFb. Thus, the high concentra- reduce breast cancer bone metastases.
tions of ionized calcium in bone may contribute
to the vicious cycle by increasing PTHrP pro- Actions of Bone-Derived Factors on
duction and osteolysis. Small molecule agonists
and antagonists of the receptor have been
Tumor Cells
developed and are in clinical trials [141]. Such The effects of bone-derived factors on tumor
agents may be effective against breast cancer cells remain understudied. Van der Pluim et al.
bone metastasis. [209] elegantly demonstrated that several
mRNAs are increased in bone compared with
non-bone sites of human breast cancer metas-
Roles of Other Osteolysis-Stimulating Factors tases in nude mice. RNA abundances were
PTHrP cannot be the only factor responsible determined by species-specic RT-PCR. PTHrP,
for bone metastases, and a number of other VEGFs, and M-CSF were increased specically
proteins play either contributory or PTHrP- in bone, while several mouse markers of host
independent roles [29]. Candidate factors that angiogenesis were similarly increased. These
may contribute to PTHrP-induced osteolytic experiments did not identify the factor(s)
lesions are interleukin [IL]-11, macrophage responsible for the bone-specic mRNA induc-
colony-stimulating factor [M-CSF], and VEGF tion. Hauschka et al. [82] found that insulin-like
Skeletal Complications of Malignancy: Central Role for the Osteoclast 163

growth factors [IGFs]-1 and -2, were most abun- pathways, such as the MAP kinases, and extra-
dant in bone matrix. The second most abundant cellular mediators, such as TGFb [46], offer
was TGFb, whereas bone morphogenetic pro- further targets to be tested for inhibition of
teins (BMPs), broblast growth factors-1 and skeletal metastases.
-2, and platelet-derived growth factor were at
lower concentrations. Only TGFb has been
shown to play a direct role in stimulating tumor Interactions Between Tumor And
cells, although IGF signaling may regulate breast Bone The Vicious Cycle
cancer cells that metastasize to bone [95]. TGFb
is growth-inhibitory in the early stages of Preclinical animal models have established that
tumorigenesis. Advanced cancers lose growth bone metastases involve a vicious cycle between
inhibition but retain TGFb regulation of metas- tumor cells and the skeleton (Figure 9.1). The
tasis-promoting genes [214], such as CTGF and cycle is fueled by four contributors: the tumor
IL-11, [98], and PTHrP [227]. In the MDA-MB- cells, bone-forming osteoblasts, bone-destroy-
231 model of breast cancer metastasis to bone, ing osteoclasts, and organic bone matrix. Osteo-
detailed experiments showed that tumor cell clast formation and activity is regulated by the
expression of PTHrP is the major target of TGFb osteoblast, adding complexity to the vicious
and that TGFb is the most important regulator cycle. The mineralized matrix of bone is a vast
of PTHrP [227]. These experiments also showed storehouse of growth factors, such as insulin-
that dual pathways in the tumor cells transmit like and transforming growth factors [82].
TGFb signaling to the nucleus: through p38 These are synthesized by osteoblasts and
MAP kinase and through the Smad proteins released by osteoclastic bone resorption. The
[97]. Osteoclastic bone resorption specically factors reach high local concentrations in the
activates TGFb from its stored form in bone bone microenvironment and can act on tumor
matrix [40]. This step may be another point at cells to attract tumor cells [145] or to encourage
which the efcacy of bisphosphonates against metastatic growth. In turn, cancer cells secrete
bone metastases is exerted [47]. Signaling many factors that act on bone cells. Therapies

Cancer Cells

Osteoblastic Factors
Bone-
Osteolytic
derived Growth Factors
Factors
Growth tur atio n
Factors Obl Ma

New Bone

Osteoblasts
Osteoclasts Mineralized Bone Matrix

Figure 9.1 The vicious cycle between tumor cells and bone which fuels the growth of bone metastases. The major components
of this vicious cycle include the cancer cells, osteoclasts, osteoblasts, and mineralized bone matrix, a major source of immobilized
growth factors. Tumor products, such as ET-1, adrenomedullin, and PTHrP123, stimulate osteoblasts (Obl) proliferation, while full-
length PTHrP, IL-11, and IL-8 stimulate osteoclastic bone resorption. Immature osteoblasts respond to osteolytic cytokines, such as
PTHrP and IL-11, by expressing RANK ligand. RANK ligand stimulates osteoclastic bone resorption by binding its cognate receptor
RANK (Y) on osteoclast precursors (shown here on mature osteoclasts). This bone resorption causes release of growth factors (TGFb)
and Ca++ from mineralized matrix. Mature osteoblasts synthesize growth factors, which are incorporated into bone and also enrich
the local microenvironment. Growth factors stimulate tumor cells. Osteoblasts lose RANK ligand expression during maturation. The
balance of osteoblast proliferation vs maturation, plus tumor production of osteolytic factors like PTHrP and IL-11, determines
whether bone metastases are osteoblastic or osteolytic.
164 Bone Resorption

targeting the vicious cycle can decrease metas- should therefore be effective therapy against all
tases by lowering the concentrations of growth types of bone metastases, whereas endothelin
factors in bone. receptor antagonists may be useful in the treat-
ment of osteoblastic metastases.
Osteoblastic and Mixed Metastases Other factors responsible for osteoblastic
metastases still need to be tested. Such factors
Histomorphometric studies of prostate cancer should possess at least these two characteristics:
samples indicate that osteoblastic metastases the ability to stimulate osteoblastic new bone
are due to tumor-produced factors that stimu- formation and to be expressed by cancer cells.
late bone formation [26, 107]. The seed and soil The bone morphogenetic proteins are obvious
analogy of Paget can be applied to osteoblastic candidates, but appear not to play a causal role
metastases, which share a similar pathophysiol- in bone metastases. CTGF, identied in the
ogy with osteolytic metastases. The tumor cells, experiments of Kang et al. [98], is a factor that
the seeds, secrete factors that stimulate stimulates osteoblasts [169]. Adrenomedullin, a
osteoblast activity and bone formation. The 52-amino acid vasoactive peptide with potent
bone microenvironment is enriched with bone-stimulatory actions [37], is produced by
osteoblast-derived growth factors, which many cancers [232]. Unpublished data (Guise
enhance the local growth of tumor cells. et al.) with lung and prostate cancer cell lines
suggest that adrenomedullin increases bone
metastases in vivo. Adrenomedullin is also an
Role of Endothelin-1 autocrine growth factor for breast cancer cells
Endothelin-1, ET-1, is a 21-amino acid peptide [128] and is transcriptionally regulated in
that is a potent vasoconstrictor. It is expressed endometrial cells by tamoxifen [144]. Its role in
by many tissues and binds to two G-protein- bone metastases induced by breast cancer has
coupled receptors. It is also a potent osteoblast- not been investigated.
stimulatory factor through its activation of the Mixed osteolytic-osteoblastic metastases are
ET A receptor [79]. Many receptor-selective characteristic of both breast and prostate
endothelin receptor antagonists have been cancers. The effects of the combined expression
developed by the pharmaceutical industry for of osteolytic and osteoblastic factors on bone
the treatment of cardiovascular conditions have not been studied, so the net response of
[158]. Endothelin plays an important role in bone at the metastatic site is unpredictable. As
cancer and on osteoblastic bone metastases in noted, osteolytic factors such as PTHrP and IL-
particular [139]. 11 act on osteoblasts to increase expression of
An animal model of osteoblastic metastases RANK ligand. We tested the effects of introduc-
that uses three standard human breast cancer ing the osteoblastic factor ET-1 into the PTHrP-
cell lines, ZR-75.1, T47D, and MCF7, has been secreting MDA-MB-231 breast cancer cell line.
established. All three lines are estrogen recep- Instead of converting the bone response from
tor-positive and express ET-1 but not PTHrP. osteolytic to osteoblastic, the bone-destructive
The ZR75.1 cell line was studied in the greatest effects were enhanced by ET-1 (Yin, Chirgwin,
detail. It stimulates new bone formation in and Guise, unpublished). Some of this effect
organ culture and causes osteoblastic metas- may be caused by autocrine responses of the
tases in vivo. Bone formation and metastases tumor cells to ET-1. Osteoblastic factors can
were effectively blocked with a selective antag- stimulate osteoblast proliferation, increasing
onist of the endothelin A receptor [226]. This the population of early osteoblasts [31]. The
orally active antagonist, Atrasentan, is in clini- enlarged pool of early osteoblasts responds to
cal trials in men with advanced metastatic osteolytic factors by increased expression of
prostate cancer [163]. The endothelin receptor RANK ligand [43, 68]. Recently Yi et al. [225]
antagonist blocks the activation of osteoblasts overexpressed platelet-derived growth factor B-
by tumor-produced ET-1. It also decreases chain in MDA-MB-231 cells and observed
osteoclastic bone resorption, as indicated by a osteosclerotic rather than osteolytic metastases.
drop in bone resorption markers seen in the A puzzling question has been the role of
patient trials [140]. Bisphosphonates, on the PTHrP in osteoblastic metastases, especially
other hand, effectively reduce skeletal-related those due to prostate cancers, which nearly
events in prostate cancer [47]. Bisphosphonates always express PTHrP. A partial explanation was
Skeletal Complications of Malignancy: Central Role for the Osteoclast 165

provided by the observation that prostate- cellular concentrations in vivo. It is thus contro-
specic antigen [PSA] is a serine proteinase, versial whether they have signicant effects on
which cleaves PTHrP after residue 23 [39, 93]. tumor cells, particularly at non-bone sites.
The resulting fragment fails to activate the clas- Controversy surrounds clinical data on the
sical PTH/PTHrP receptor. It was later observed effects of bisphosphonates on breast cancer
that the inactive fragment PTHrP116 increased tumor burden at extra-skeletal sites [44, 45].
cyclic AMP in cardiomyocytes by activating Although bisphosphonates have direct anti-
the endothelin A receptor. Binding was attrib- tumor actions in vitro, it is unclear whether the
uted to a four amino acid near-identity between compounds reach sufciently high local con-
the two peptides [174]. We have extended centrations in extra-skeletal sites to have such
these observations to bone. PTHrP123, in con- actions in the body. The bisphosphonates have
centrations as low as 1nM, is a potent stimula- the potential to enhance anti-tumor activities of
tor of calvarial new bone formation. PSA is known cytotoxic agents, but further and larger
commonly expressed in breast cancer [12]. The clinical trials are required to address the impor-
results suggest that PSA proteolysis, rather tance of the preclinical data. A major morbidity
than inactivating PTHrP, converts it from an for patients with skeletal metastases is
osteolytic factor to a potent osteoblastic one. intractable bone pain, which is reduced by bis-
The same may occur in breast cancer bone phosphonate treatment [35, 116, 226]. Interrup-
metastases. tion of the vicious cycle with bisphosphonates
Tumor cells also secrete factors that oppose reduces the concentrations of pain-stimulating
the development and progression of bone molecules in the microenvironment surround-
metastases. One such example is interleukin-18, ing metastatic tumor cells. Although numerous
which decreases osteoclast formation [94, 138]. bisphosphonates are in clinical use and several
Clearly these factors may hold new approaches are approved for cancer bone metastases, they
for future anti-metastatic therapies. Similarly, have not been shown to improve patient sur-
the roles of angiogenesis [208] and the immune vival in patients with established solid tumor
system [161] in bone metastases are not well metastases to bone [3335, 116].
known.
Other Bone-targeting Agents
Clinical And Experimental Therapies The afnity of the bisphosphonate chemical
structure for the mineralized matrix of bone
Bisphosphonates constitutes a unique mechanism by which these
Bisphosphonates are approved by the Food and agents can be targeted to bone. An inhibitor of
Drug Administration for treatment of estab- intracellular src signaling was modied by the
lished bone metastases due to breast and addition of a pair of phosphonate groups and
prostate cancer, as well as other solid tumors. shown to inhibit osteoclastic bone resorption
Bisphosphonates have many low-specicity bio- [179]. This may provide a new way to add speci-
chemical actions that may be responsible for city for bone targeting to chemotherapeutic
their effectiveness. They reduce tumor growth and adjuvant compounds.
in vitro [30, 73] and invasiveness [14], as well as Morony et al. [130] treated mice with a mod-
increase apoptosis [33]. Bisphosphonates can ied recombinant version of osteoprotegerin
reduce adhesion of tumor cells to bone [210] (OPG), the soluble, inhibitory binding protein
and decrease angiogenesis [72, 217]. Angiogen- for RANK ligand. OPG inhibits osteoclast for-
esis plays an important role in bone metastases, mation, activity, and survival. OPG, when
and bisphosphonates may affect vascular injected into mice, decreased osteolytic destruc-
endothelial cells and tumor cells in bone. tion and tumor burden in bone, but had no
Osteoblasts bind bisphosphonates, which effect on metastases in soft tissues [130]. OPG
appear to be anti-apoptotic for the cells, while treatment also decreased bone pain [85].
they are pro-apoptotic for osteoclasts. Com- Recombinant OPG has entered initial clinical
pound-specic effects unique to osteoblasts trials [13], but it is unclear whether it will
have been reported [59, 157], but it is unknown prove equal or superior to the widely used and
if these actions are important in vivo. The more convenient bisphosphonate antiresorptive
actions of bisphosphonates are limited by their agents.
166 Bone Resorption

Therapy to neutralize PTHrP or inhibit its Women who have had breast cancer are at
production is currently under investigation to increased risk for osteoporosis. They are more
treat breast cancer metastases to bone. PTHrP likely to undergo early menopause due to
neutralizing antibodies, which effectively chemotherapy-induced ovarian failure or
reduced breast cancer metastases to bone in a oophorectomy. Chemotherapy may also
mouse model, have been changed for human adversely affect bone mineral density.
application and have now entered clinical trials Tamoxifen treatment causes signicant bone
[80]. Similarly, small molecule inhibitors of loss in premenopausal women, yet prevents it in
PTHrP transcription have reduced osteolytic postmenopausal women. In women with breast
bone metastases in the same mouse models and cancer the risk of vertebral fracture is markedly
offer promise as clinical treatment for osteolytic increased [99, 100], with incidence increased
bone metastases [65]. A recent study, designed nearly ve times in women from the time of rst
to inhibit the nuclear promoter responsible diagnosis of breast cancer and 20-fold in women
for PTHrP transcription in tumor cells, used with soft-tissue metastases who had no skeletal
a PTHrP promoter-luciferase reporter in a metastases. Selective estrogen-receptor modula-
high-throughput screen of small molecules tors, SERMS, may play a particularly important
and identied two related molecules as effec- role in the treatment of breast cancer without
tive inhibitors of PTHrP transcription: 6- causing bone loss [34, 57, 159]. The active
thioguanine and 6-thioguanosine. These agents metabolite of vitamin D stimulates osteoblastic
have been used for over a quarter of a century expression of RANK ligand and can increase
against leukemias and several inammatory osteoclastic bone resorption. At the same time,
disorders. When tested in animal models of vitamin D metabolites have powerful anti-
humoral hypercalcemia of malignancy and tumor effects. As with other steroid derivatives,
against breast cancer bone metastases, both selective receptor-modulating vitamin D
were effective [65]. analogs may be effective against breast cancer
Endothelin receptor antagonists may also cells, while sparing bone [49].
prove effective against osteoblastic metastases Since bone resorption directly stimulates the
[139, 140, 163, 226]. Small molecule inhibitors of vicious cycle of bone metastasis (Figure 9.1) it
intracellular signaling in either bone or cancer is possible that treatment-induced bone loss
cells have not been reported to be effective could increase skeletal metastases. This unde-
against bone metastases, although the range of sirable possibility has now been shown to occur
testable targets is very wide. in animal models (Padalecki et al, in prepara-
tion), but may be treatable with antiresorptive
Bone Loss in the Patient with agents. If this is true, then it would be desirable
to inhibit treatment-related bone loss, thus
Cancer and Effects of Therapy on preserving skeletal health and decreasing bone
the Skeleton metastases.

Bone metastases are not the only source of


skeletal morbidity in cancer patients. Patients
with prostate and breast cancer are often sex-
Acknowledgments
steroid decient due to treatment. This state
results in accelerated osteoclastic bone resorp- Work in the authors laboratory was supported
tion, bone loss, and increased risk of fracture in part by funding from the Gerald D. Aurbach
[42]. Sex steroids have complex, gender-specic endowment, the Mellon Institute of the
effects on bone through both genomic and non- University of Virginia Cancer Center, and
genomic mechanisms [106, 122, 187] and also research grants from the NIH (R01 CA69158
regulate the tumor production of bone-active and P01 CA44035) and the U.S. Army
factors such as PTHrP [63] and ET-1. The stan- (DAMD17991-9401) to T.A.G and the
dard of care for patients with advanced prostate Veterans Administration Research Service
cancer, for example, is androgen ablation. (Merit Award) and the U.S. Army (DAMD17
However, hypogonadism causes male osteo- 981-8245 and DAMD17021-0586) to J.M.C.
porosis, and bisphosphonate treatment prevents The authors thank Mr. Cliff Martin for assis-
bone loss or can increase bone mass [184, 185]. tance in preparing the gures.
Skeletal Complications of Malignancy: Central Role for the Osteoclast 167

carcinoma related bone metastases. Cancer 97(3


References Suppl):88792.
14. Boissier S, Ferreras M, Peyruchaud O, Magnetto S,
Ebetino FH, Colombel M, Delmas P, Delaisse JM,
1. Abou-Samra AB, Juppner H, Force T, Freeman MW, Clezardin P (2000) Bisphosphonates inhibit breast and
Kong XF, Schipani E, et al. (1992) Expression cloning prostate carcinoma cell invasion, an early event in the
of a common receptor for parathyroid hormone and formation of bone metastases. Cancer Res 60:2949
parathyroid hormone-related peptide from rat 54.
osteoblast-like cells: a single receptor stimulates 15. Botella A, Rekik M, Delvaux M, Davicco MJ, Barlet JP,
intracellular accumulation of both cAMP and inositol Frexinos J, et al. (1994) Parathyroid hormone (PTH)
trisphosphates and increases intracellular free and PTH-related peptide induce relaxation of smooth
calcium. Proc Natl Acad Sci USA 89(7):27326. muscle cells from guinea pig ileum: interaction with
2. Adams JS, Sharma OP, Gacad MA, Singer FR (1983) vasoactive intestinal peptide receptors. Endocrinology
Metabolism of 25-hydroxyvitamin D3 by cultured pul- 135(5):21607.
monary alveolar macrophages in sarcoidosis. Journal 16. Boxer DI, Todd CE, Coleman R, Fogelman I (1989)
of Clinical Investigation 72(5):185660. Bone secondaries in breast cancer: the solitary metas-
3. Arguello F, Baggs RB, Frantz CN (1998) A murine tasis. J Nucl Med 30:131820.
model of experimental metastasis to bone and bone 17. Boyce BF, Aufdemorte TB, Garrett IR, Yates AJ, Mundy
marrow. Cancer Res 48:687681. GR (1989) Effects of interleukin-1 on bone turnover in
4. Bachelder RE, Wendt MA, Mercurio AM (2002) Vascu- normal mice. Endocrinology 125(3):114250.
lar endothelial growth factor promotes breast carci- 18. Boyde A, Maconnachie E, Reid SA, Delling G, Mundy
noma invasion in an autocrine manner by regulating GR (1986) Scanning electron microscopy in bone
the chemokine receptor CXCR4. Cancer Res 62:7203 pathology: Review of methods. Potential and applica-
6. tions. Scanning Electron Microscopy IV:153754.
5. Barhoum M, Hutchins L, Fonseca VA (1999) Intractable 19. Breslau NA, McGuire JL, Zerwekh JE, Frenkel EP, Pak
hypercalcemia due to a metastatic carcinoid secreting CY (1984) Hypercalcemia associated with increased
parathyroid hormone-related peptide and interleukin- serum calcitriol levels in three patients with lym-
6: response to octreotide. American Journal of the phoma. Annals of Internal Medicine 100(1):16.
Medical Sciences 318(3):2035. 20. Buchs N, Manen D, Bonjour JP, Rizzoli R (2000)
6. Bendre MS, Gaddy-Kurten D, Mon-Foote T, Akel NS, Calcium stimulates parathyroid hormone-related pro-
Skinner RA, Nicholas RW, Suva LJ (2002) Expression of tein production in Leydig tumor cells through a puta-
interleukin 8 and not parathyroid hormone-related tive cation-sensing mechanism. Eur J Endocrinol 142:
protein by human breast cancer cells correlates with 5005.
bone metastasis in vivo. Cancer Res 62:55719. 21. Bundred NJ, Ratcliffe WA, Walker RA, Coley S,
7. Berruti A, Dogliotti L, Tucci M, Tarabuzzi R, Guercio S, Morrison JM, Ratcliffe JG (1991) Parathyroid hormone
Torta M, Tampellini M, Dovio A, Poggio M, Scarpa RM, related protein and hypercalcaemia in breast cancer.
Angeli A (2002) Metabolic effects of single-dose British Medical Journal 303(6816):15069.
pamidronate administration in prostate cancer pa- 22. Bundred NJ, Walker RA, Ratcliffe WA, Warwick J,
tients with bone metastases. Int J Biol Markers 17: Morrison JM, Ratcliffe JG (1992) Parathyroid hormone
24452. related protein and skeletal morbidity in breast cancer.
8. Bertolini DR, Nedwin GE, Bringman TS, Smith DD, European Journal of Cancer 28(23):6902.
Mundy GR (1986) Stimulation of bone resorption and 23. Burtis WJ, Brady TG, Orloff JJ, Ersbak JB, Warrell RP,
inhibition of bone formation in vitro by human Jr., Olson BR, et al. (1990) Immunochemical character-
tumour necrosis factors. Nature 319(6053):51618. ization of circulating parathyroid hormone-related
9. Binstock ML, Mundy GR (1981) Effect of calcitonin protein in patients with humoral hypercalcemia of
and glutocorticoids in combination on the hypercal- cancer. New England Journal of Medicine 322(16):
cemia of malignancy. Annals of Internal Medicine 110612.
93(2):26972. 24. Burtis WJ, Wu T, Bunch C, Wysolmerski J, Insogna K,
10. Bisaz S, Jung A, Fleisch H (1978) Uptake by bone of Weir E, Broadus AE, Stewart AF (1987) Identication of
pyrophosphate, diphosphonates and their technetium a novel 17,000-dalton parathyroid hormone-like
derivatives. Clin Sci Mol Med 54:26572. adenylate cyclase-stimulating protein from a tumor
11. Black K, Garrett IR, Mundy GR (1991). Chinese associated with humoral hypercalcemia of malig-
hamster ovarian cells transfected with the murine nancy. J Biol Chem 262:71516.
interleukin-6 gene cause hypercalcemia as well as 25. Chambers AF, Groom AC, MacDonald IC (2002) Dis-
cachexia, leukocytosis and thrombocytosis in tumor- semination and growth of cancer cells in metastatic
bearing nude mice. Endocrinology 128(5):26579. sites. Nat Rev Cancer 2:56372.
12. Black MH, Diamandis EP (2000) The diagnostic and 26. Charhon SA, Chapuy MC, Delvin EE, Valentin-Opran
prognostic utility of prostate-specic antigen for A, Edouard CM, Meunier PJ (1983) Histomorphomet-
diseases of the breast. Breast Cancer Res Treat 59:1 ric analysis of sclerotic bone metastases from prosta-
14. tic carcinoma special reference to osteomalacia.
13. Body JJ, Greipp P, Coleman RE, Facon T, Geurs F, Cancer 51:91824.
Fermand JP, Harousseau JL, Lipton A, Mariette X, 27. Chelouche Lev D, Price JE (2002) Therapeutic inter-
Williams CD, Nakanishi A, Holloway D, Martin SW, vention with breast cancer metastasis. Crit Rev
Dunstan CR, Bekker PJ (2003) A phase I study of Eukaryot Gene Expr 12:13750.
AMGN-0007, a recombinant osteoprotegerin con- 28. Chen HL, Demiralp B, Schneider A, Koh AJ, Silve C,
struct, in patients with multiple myeloma or breast Wang CY, et al. (2002) Parathyroid hormone and
168 Bone Resorption

parathyroid hormone-related protein exert both 46. Dumont N, Arteaga CL (2003) Targeting the TGFb
pro- and anti-apoptotic effects in mesenchymal cells. signaling network in human neoplasia. Cancer Cell
Journal of Biological Chemistry 277(22):1937481. 3:5316.
29. Chirgwin JM, Guise TA (2000) Molecular mechanisms 47. Eaton CL, Coleman RE (2003) Pathophysiology of
of tumor-bone interactions in osteolytic metastases. bone metastases from prostate cancer and the role
Crit Rev Eukaryot Gene Exp 10:15978. of bisphosphonates in treatment. Cancer Treat Rev
30. Clezardin P (2002) The antitumor potential of bispho- 29:18998.
sphonates. Semin Oncol 29(6 Suppl 21):3342. 48. Eilon G, Mundy GR (1978) Direct resorption of bone
31. Coffman JA (2003) Runx transcription factors and the by human breast cancer cells in vitro. Nature 276:
developmental balance between cell proliferation and 7268.
differentiation. Cell Biol Int 27:31524. 49. El Abdaimi K, Dion N, Papavasiliou V, Cardinal PE,
32. Coleman RE, Rubens RD (1987) The clinical course of Binderup L, Goltzman D, Ste-Marie LG, Kremer R
bone metastases from breast cancer. Brit J Cancer 55: (2000) The vitamin D analogue EB 1089 prevents skele-
616. tal metastasis and prolongs survival time in nude mice
33. Coleman RE (2002) Bisphosphonates for the preven- transplanted with human breast cancer cells. Cancer
tion of bone metastases. Semin Oncol 29(6 Suppl 21): Res 60:441214.
439. 50. Felding-Habermann B, OToole TE, Smith JW, Fransvea
34. Coleman RE (2003) Current and future status of E, Ruggeri ZM, Ginsberg MH, Hughes PE, Pampori N,
adjuvant therapy for breast cancer. Cancer. 97(3 Suppl): Shattil SJ, Saven A, Mueller BM (2001). Integrin activa-
8806. tion controls metastasis in human breast cancer. Proc
35. Coleman RE (2002) Efcacy of zoledronic acid and Natl Acad Sci USA 98:18538.
pamidronate in breast cancer patients: a comparative 51. Fetchick DA, Bertolini DR, Sarin PS, Weintraub ST,
analysis of randomized phase III trials. Am J Clin Mundy GR, Dunn JF (1986) Production of 1,25-dihy-
Oncol 25(6 Suppl 1):S2531. droxyvitamin D3 by human T cell lymphotrophic
36. Cooper CR, Chay CH, Gendernalik JD, Lee HL, Bhatia virus-I-transformed lymphocytes. Journal of Clinical
J, Taichman RS, McCauley LK, Keller ET, Pienta KJ Investigation 78(2):5926.
(2003) Stromal factors involved in prostate carcinoma 52. Fidler IJ (2003) The pathogenesis of cancer metastasis:
metastasis to bone. Cancer 97(3 Suppl):73947. the seed and soil hypothesis revisited. Nat Rev
37. Cornish J, Naot D, Reid IR (2003) Adrenomedullin-a Cancer 3:16.
regulator of bone formation. Regul Pept 112:7986. 53. Fierabracci P, Pinchera A, Miccoli P, Conte PF, Vignali
38. Coussens LM, Fingleton B, Matrisian LM (2002) Matrix E, Zaccagnini M, et al. (2001) Increased prevalence of
metalloproteinase inhibitors and cancer: trials and primary hyperparathyroidism in treated breast cancer.
tribulations. Science 295:238792. Journal of Endocrinological Investigation 24(5):
39. Cramer SD, Chen Z, Peehl DM (1996) Prostate specic 31520.
antigen cleaves parathyroid hormone-related protein 54. Firkin F, Seymour JF, Watson AM, Grill V, Martin TJ
in the PTH-like domain: inactivation of PTHrP-stimu- (1996) Parathyroid hormone-related protein in hyper-
lated cAMP accumulation in mouse osteoblasts. J Urol calcaemia associated with haematological malignancy.
156:52631. British Journal of Haematology 94(3):48692.
40. Dallas SL, Rosser JL, Mundy GR, Bonewald LF (2002) 55. Fleisch H (1991) Bisphosphonates. Pharmacology and
Proteolysis of latent transforming growth factor-beta use in the treatment of tumour-induced hypercal-
(TGF-beta)-binding protein-1 by osteoclasts. A cellular caemic and metastatic bone disease. Drugs 42(6):
mechanism for release of TGF-beta from bone matrix. 91944.
J Biol Chem 277:2135260. 56. Fontana A, Delmas PD (2000) Markers of bone turn-
41. de la Mata J, Uy HL, Guise TA, Story B, Boyce BF, Mundy over in bone metastases. Cancer 88(12 Suppl):
GR, et al. (1995) Interleukin-6 enhances hypercalcemia 295260.
and bone resorption mediated by parathyroid 57. Fontana A, Delmas PD (2003) Selective estrogen recep-
hormone-related protein. Journal of Clinical Investiga- tors modulators in the prevention and treatment of
tion 95(6):284652. postmenopausal osteoporosis. Endocrinol Metab Clin
42. Delmas PD, Balena R, Confravreux E, Hardouin C, North Am 32:21932.
Hardy P, Bremond A (1997) Bisphosphonate rise- 58. Frith JC, Rogers MJ (2003) Antagonistic effects of dif-
dronate prevents bone loss in women with articial ferent classes of bisphosphonates in osteoclasts and
menopause due to chemotherapy of breast cancer: a macrophages in vitro. J Bone Miner Res 18:20412.
double-blind, placebo-controlled study. J Clin Oncol 59. Fromigue O, Body JJ (2002) Bisphosphonates inuence
15:95562. the proliferation and the maturation of normal human
43. Deyama Y, Takeyama S, Koshikawa M, Shirai Y, osteoblasts. J Endocrinol Invest 25:53946.
Yoshimura Y, Nishikata M, Suzuki K, Matsumoto A 60. Fukumitsu N, Uchiyama M (2002) Correlation of urine
(2000) Osteoblast maturation suppressed osteoclasto- type I collagen-cross-linked N telopeptide levels with
genesis in coculture with bone marrow cells. Biochem bone scintigraphic results in prostate cancer patients.
Biophys Res Commun 274:24954. Metabolism 51:81418.
44. Diel IJ, Solomayer EF, Bastert G (2000) Bisphospho- 61. Funk JL, Lausier J, Moser AH, Shigenaga JK, Huling S,
nates and the prevention of metastasis: rst evidences Nissenson RA, et al. (1995) Endotoxin induces parathy-
from preclinical and clinical studies. Cancer 88(12 roid hormone-related protein gene expression in
Suppl):30808. splenic stromal and smooth muscle cells, not in splenic
45. Diel IJ, Solomayer EF, Bastert G (2000) Treatment of lymphocytes. Endocrinology 136(8):341221.
metastatic bone disease in breast cancer: bisphospho- 62. Funk JL, Shigenaga JK, Moser AH, Krul EJ, Strewler GJ,
nates. Clin Breast Cancer 1:4351. Feingold KR, et al. (1994) Cytokine regulation of
Skeletal Complications of Malignancy: Central Role for the Osteoclast 169

parathyroid hormone-related protein messenger 79. Guise TA, Yin JJ, Mohammad KS (2003) Role of
ribonucleic acid levels in mouse spleen: paradoxical endothelin-1 in osteoblastic bone metastases. Cancer
effects of interferon-gamma and interleukin-4. 97(3 Suppl):77984.
Endocrinology 135(1):3518. 80. Guise TA, Yin JJ, Taylor SD, Kumagai Y, Dallas M, Boyce
63. Funk JL, Wei H (1998) Regulation of PTHrP expression BF, Yoneda T, Mundy GR (1996) Evidence for a causal
in MCF-7 breast carcinoma cells by estrogen and antie- role of parathyroid hormone-related protein in the
strogens. Biochem Biophys Res Commun 251:849 pathogenesis of human breast cancer-mediated osteol-
54. ysis. J Clin Invest 98:15449.
64. Galasko CS (1986) Skeletal metastases. Clin Orthop 81. Guise TA, Yoneda T, Yates AJ, Mundy GR (1993) The
210:1830. combined effect of tumor-produced parathyroid
65. Gallwitz WE, Guise TA, Mundy GR (2002) Guanosine hormone-related protein and transforming growth
nucleotides inhibit different syndromes of PTHrP factor-alpha enhance hypercalcemia in vivo and
excess caused by human cancers in vivo. J Clin Invest bone resorption in vitro. J Clin Endocrinol Metab
110(10):155972. 77(1):405.
66. Gardella TJ, Juppner H (2001) Molecular properties of 82. Hauschka PV, Mavrakos AE, Iafrati MD, Doleman SE,
the PTH/PTHrP receptor. Trends Endocrinol Metab Klagsbrun M (1986) Growth factors in bone matrix.
12:21017. Isolation of multiple types by afnity chromatography
67. Garnero P, Buchs N, Zekri J, Rizzoli R, Coleman RE, on heparin-Sepharose. J Biol Chem 261:1266574.
Delmas PD (2000) Markers of bone turnover for the 83. Henderson M, Danks J, Moseley J, Slavin J, Harris T,
management of patients with bone metastases from McKinlay M, Hopper J, Martin T (2001) Parathyroid
prostate cancer. Br J Cancer 82:85864. hormone-related protein production by breast
68. Geoffroy V, Kneissel M, Fournier B, Boyde A, Matthias cancers, improved survival, and reduced bone metas-
P (2002) High bone resorption in adult aging trans- tases. J Natl Cancer Inst 93:2347.
genic mice overexpressing cbfa1/runx2 in cells of the 84. Hofbauer LC, Heufelder AE (2001) Role of receptor
osteoblastic lineage. Mol Cell Biol 22:622233. activator of nuclear factor-kappaB ligand and osteo-
69. Giuliani N, Pedrazzoni M, Negri G, Passeri G, protegerin in bone cell biology. J Mol Med 79:24353.
Impicciatore M, Girasole G (1998) Bisphosphonates 85. Honore P, Luger NM, Sabino MA, Schwei MJ, Rogers
stimulate formation of osteoblast precursors and SD, Mach DB, Okeefe PF, Ramnaraine ML, Clohisy DR,
mineralized nodules in murine and human bone Mantyh PW (2000) Osteoprotegerin blocks bone
marrow cultures in vitro and promote early osteoblas- cancer-induced skeletal destruction, skeletal pain and
togenesis in young and aged mice in vivo. Bone pain-related neurochemical reorganization of the
22(5):45561. spinal cord. Nat Med 6:5218.
70. Giuliani N, Pedrazzoni M, Passeri G, Girasole G (1998) 86. Horiuchi N, Cauleld MP, Fisher JE, Goldman ME,
Bisphosphonates inhibit IL-6 production by human McKee RL, Reagan JE, et al. (1987) Similarity of syn-
osteoblast-like cells. Scandinavian Journal of Rheuma- thetic peptide from human tumor to parathyroid
tology 27(1):3841. hormone in vivo and in vitro. Science 238(4833):
71. Gkonos PJ, London R, Hendler ED (1984) Hypercal- 15668.
cemia and elevated 1,25-dihydroxyvitamin D levels 87. Hortobagyi GN, Theriault RL, Porter L, Blayney D,
in a patient with end-stage renal disease and active Lipton A, Sinoff C, Wheeler H, Simeone JF, Seaman J,
tuberculosis. New England Journal of Medicine Knight RD, Heffernan M, Reitsma (1996) Efcacy of
311(26):16835. pamidronate in reducing skeletal complications in
72. Green JR, Clezardin P (2002) Mechanisms of bisphos- patients with breast cancer and lytic bone metastases.
phonate effects on osteoclasts, tumor cell growth, and New Engl J Med 335:178591.
metastasis. Am J Clin Oncol 25(6 Suppl 1):S39. 88. Hosking DJ, Cowley A, Bucknall CA (1981) Rehydra-
73. Green JR (2003) Antitumor effects of bisphosphonates. tion in the treatment of severe hypercalcaemia. Quar-
Cancer 97(3 Suppl):8407. terly Journal of Medicine 50(200):47381.
74. Grill V, Ho P, Body JJ, Johanson N, Lee SC, Kukreja SC, 89. Ibbotson KJ, Twardzik DR, DSouza SM, Hargreaves
et al. (1991) Parathyroid hormone-related protein: ele- WR, Todaro GJ, Mundy GR (1985) Stimulation of bone
vated levels in both humoral hypercalcemia of malig- resorption in vitro by synthetic transforming growth
nancy and hypercalcemia complicating metastatic factor-alpha. Science 228(4702):10079.
breast cancer. Journal of Clinical Endocrinology & 90. Iguchi H, Miyagi C, Tomita K, Kawauchi S, Nozuka Y,
Metabolism 73(6):130915. Tsuneyoshi M, et al. (1998) Hypercalcemia caused by
75. Grill V, Martin TJ (2000) Hypercalcemia of malig- ectopic production of parathyroid hormone in a
nancy. Reviews in Endocrine & Metabolic Disorders patient with papillary adenocarcinoma of the thyroid
1(4):25363. gland. Journal of Clinical Endocrinology & Metabo-
76. Gucalp R, Ritch P, Wiernik PH, Sarma PR, Keller A, lism 83(8):26537.
Richman SP, et al. (1992) Comparative study of 91. Iguchi H, Tanaka S, Ozawa Y, Kashiwakuma T, Kimura
pamidronate disodium and etidronate disodium in the T, Hiraga T, Ozawa H, Kono A (1996) An experimental
treatment of cancer-related hypercalcemia. Journal of model of bone metastasis by human lung cancer cells:
Clinical Oncology 10(1):13442. the role of parathyroid hormone-related protein in
77. Guise TA, Garrett IR, Bonewald LF, Mundy GR (1993) bone metastasis. Cancer Res 56:40403.
Interleukin-1 receptor antagonist inhibits the hyper- 92. Ikeda K, Ohno H, Hane M, Yokoi H, Okada M, Honma
calcemia mediated by interleukin-1. Journal of Bone & T, et al. (1994) Development of a sensitive two-site
Mineral Research 8(5):5837. immunoradiometric assay for parathyroid hormone-
78. Guise TA, Mundy GR (1998) Cancer and bone. Endocr related peptide: evidence for elevated levels in plasma
Rev 19:1854. from patients with adult T-cell leukemia/lymphoma
170 Bone Resorption

and B-cell lymphoma. Journal of Clinical Endocrinol- in mice by nongenotropic signaling of sex steroids.
ogy & Metabolism 79(5):13227. Science 298:8436.
93. Iwamura M, Hellman J, Cockett AT, Lilja H, Gershagen 107. Koutsilieris M (1995) Skeletal metastases in advanced
S (1996) Alteration of the hormonal bioactivity of prostate cancer: cell biology and therapy. Crit Rev
parathyroid hormone-related protein (PTHrP) as a Oncol Hematol 18:5164.
result of limited proteolysis by prostate-specic 108. Kovacs CS, Lanske B, Hunzelman JL, Guo J, Karaplis
antigen. Urology 48:31725. AC, Kronenberg HM (1996) Parathyroid hormone-
94. Iwasaki T, Yamashita K, Tsujimura T, Kashiwamura S, related peptide (PTHrP) regulates fetal-placental
Tsutsui H, Kaisho T, Sugihara A, Yamada N, Mukai M, calcium transport through a receptor distinct from the
Yoneda T, Okamura H, Akedo H, Terada N (2002) Inter- PTH/PTHrP receptor. Proc Natl Acad Sci USA
leukin-18 inhibits osteolytic bone metastasis by 93(26):152338.
human lung cancer cells possibly through suppres- 109. Kremer R, Shustik C, Tabak T, Papavasiliou V,
sion of osteoclastic bone-resorption in nude mice. Goltzman D (1996) Parathyroid-hormone-related
J Immunother 25 Suppl 1:S5260. peptide in hematologic malignancies. American
95. Jackson JG, Zhang X, Yoneda T, Yee D (2001) Regula- Journal of Medicine 100(4):40611.
tion of breast cancer cell motility by insulin receptor 110. Kukreja SC, Rosol TJ, Wimbiscus SA, Shevrin DH, Grill
substrate-2 (IRS-2) in metastatic variants of human V, Barengolts EI, Martin TJ (1990) Tumor resection and
breast cancer cell lines. Oncogene 20:731825. antibodies to parathyroid hormone-related protein
96. Johnson RA, Boyce BF, Mundy GR, Roodman GD cause similar changes on bone histomorphometry in
(1989) Tumors producing human tumor necrosis hypercalcemia of cancer. Endocrinology Jul 127(1):
factor induced hypercalcemia and osteoclastic bone 30510.
resorption in nude mice. Endocrinology 124(3):1424 111. Lee J,Weber M, Mejia S, Bone E,Watson P, Orr W (2001)
7. A matrix metalloproteinase inhibitor, batimastat,
97. Kakonen SM, Selander KS, Chirgwin JM, Yin JJ, Burns retards the development of osteolytic bone metastases
S, Rankin WA, Grubbs BG, Dallas M, Cui Y, Guise TA by MDA-MB-231 human breast cancer cells in Balb C
(2002) Transforming growth factor-beta stimulates nu/nu mice. Eur J Cancer 37:10613.
parathyroid hormone-related protein and osteolytic 112. Li H, Seitz PK, Selvanayagam P, Rajaraman S, Cooper
metastases via Smad and mitogen-activated protein CW (1996) Effect of endogenously produced parathy-
kinase signaling pathways. J Biol Chem 277:24571 roid hormone-related peptide on growth of a human
8. hepatoma cell line (Hep G2). Endocrinology 137(6):
98. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, 236774.
Cordon-Cardo C, Guise TA, Massague J (2003) A multi- 113. Li X, Tomita M, Pilbeam CC, Breyer RM, Raisz LG
genic program mediating breast cancer metastasis to (2002) Prostaglandin receptor EP2 mediates PGE2
bone. Cancer Cell 3:53749. stimulated hypercalcemia in mice in vivo.
99. Kanis JA, McCloskey EV, Powles T, Paterson AH, Ashley Prostaglandins Other Lipid Mediat 67(34):17380.
S, Spector T (1999) A high incidence of vertebral 114. Liapis H, Crouch EC, Grosso LE, Kitazawa S, Wick MR
fracture in women with breast cancer. Br J Cancer (1993) Expression of parathyroidlike protein in
79:117981. normal, proliferative, and neoplastic human breast
100. Kanis JA, McCloskey EV, Powles TJ (1999) Clodronate tissues. American Journal of Pathology 143(4):1169
in the treatment of neoplastic bone disease. Program 78.
and Abstracts, Second International Conference on 115. Lin DL, Tarnowski CP, Zhang J, Dai J, Rohn E, Patel AH,
Cancer-Induced Bone Disease 4. Morris MD, Keller ET (2001) Bone metastatic LNCaP-
101. Kanis JA, Urwin GH, Gray RE, Beneton MN, McCloskey derivative C42B prostate cancer cell line mineralizes
EV, Hamdy NA, et al. (1987) Effects of intravenous in vitro. Prostate 47:21221.
etidronate disodium on skeletal and calcium metabo- 116. Lipton A (2003) Bisphosphonates and metastatic
lism. American Journal of Medicine 82(2A):5570. breast carcinoma. Cancer 97(3 Suppl):848853.
102. Klein BY, Ben-Bassat H, Breuer E, Solomon V, Golomb 117. Lipton A (2003) Bone metastases in breast cancer. Curr
G (1998) Structurally different bisphosphonates exert Treat Options Oncol 4:1518.
opposing effects on alkaline phosphatase and mineral- 118. Luger NM, Sabino MA, Schwei MJ, Mach DB, Pomonis
ization in marrow osteoprogenitors. Journal of Cellu- JD, Keyser CP, Rathbun M, Clohisy DR, Honore P,Yaksh
lar Biochemistry 68(2):18694. TL, Mantyh PW (2002) Efcacy of systemic morphine
103. Klein DC, Raisz LG (1970) Prostaglandins: stimulation suggests a fundamental difference in the mechanisms
of bone resorption in tissue culture. Endocrinology that generate bone cancer vs inammatory pain. Pain
86(6):143640. 99:397406.
104. Kohno N, Kitazawa S, Fukase M, Sakoda Y, Kanbara Y, 119. Luparello C, Burtis WJ, Raue F, Birch MA, Gallagher JA
Furuya Y, et al. (1994) The expression of parathyroid (1995) Parathyroid hormone-related peptide and 8701-
hormone-related protein in human breast cancer BC breast cancer cell growth and invasion in vitro: evi-
with skeletal metastases. Surgery Today 24(3):215 dence for growth-inhibiting and invasion-promoting
20. effects. Molecular & Cellular Endocrinology 111(2):
105. Koizumi M, Ogata E (2002) Bone metabolic markers as 22532.
gauges of metastasis to bone: a review. Ann Nucl Med 120. Lynch CC, Matrisian LM (2002) Matrix metallopro-
16:1618. teinases in tumor-host cell communication. Differenti-
106. Kousteni S, Chen JR, Bellido T, Han L, Ali AA, OBrien ation 70:56173.
CA, Plotkin L, Fu Q, Mancino AT, Wen Y, Vertino AM, 121. Major P, Lortholary A, Hon J, Abdi E, Mills G, Menssen
Powers CC, Stewart SA, Ebert R, Partt AM, Weinstein HD, et al. (2001) Zoledronic acid is superior to
RS, Jilka RL, Manolagas SC (2002) Reversal of bone loss pamidronate in the treatment of hypercalcemia of
Skeletal Complications of Malignancy: Central Role for the Osteoclast 171

malignancy: a pooled analysis of two randomized, encoding a secreted form of ODF/TRANCE that
controlled clinical trials. Journal of Clinical Oncology induces osteoclast formation. Biochem Biophys Res
19(2):55867. Commun 269(2):5326.
122. Manolagas SC, Kousteni S, Jilka RL (2002). Sex steroids 138. Nakata A, Tsujimura T, Sugihara A, Okamura H,
and bone. Recent Prog Horm Res 57:385409. Iwasaki T, Shinkai K, Iwata N, Kakishita E, Akedo H,
123. Mantyh PW, Clohisy DR, Koltzenburg M, Hunt SP Terada N (1999) Inhibition by interleukin 18 of oste-
(2002) Molecular mechanisms of cancer pain. Nat Rev olytic bone metastasis by human breast cancer cells.
Cancer 2:2019. Anticancer Res 19:41318.
124. Mareel M, Leroy A (2003) Clinical, cellular, and 139. Nelson J, Bagnato A, Battistini B, Nisen P (2003) The
molecular aspects of cancer invasion. Physiol Rev endothelin axis: emerging role in cancer. Nat Rev
83:33776. Cancer 3:11016.
125. Mason RS, Frankel T, Chan YL, Lissner D, Posen S 140. Nelson JB, Nabulsi AA, Vogelzang NJ, Breul J,
(1984) Vitamin D conversion by sarcoid lymph node Zonnenberg BA, Daliani DD, Schulman CC, Carducci
homogenate. Annals of Internal Medicine 100(1): MA (2003) Suppression of prostate cancer induced
5961. bone remodeling by the endothelin receptor A antag-
126. Mastro AM, Gay CV, Welch DR (2003) The skeleton as onist atrasentan. J Urol 169:11439.
a unique environment for breast cancer cells. Clin Exp 141. Nemeth EF (2002) Pharmacological regulation of
Metastasis 20:27584. parathyroid hormone secretion. Curr Pharm Des
127. Mercier RJ, Thompson JM, Harman GS, Messerschmidt 8:207787.
GL (1988) Recurrent hypercalcemia and elevated 142. Nielsen PK, Rasmussen AK, Feldt-Rasmussen U,
1,25-dihydroxyvitamin D levels in Hodgkins disease. Brandt M, Christensen L, Olgaard K (1996) Ectopic
American Journal of Medicine 84(1):1658. production of intact parathyroid hormone by a
128. Miller MJ, Martinez A, Unsworth EJ, Thiele CJ, Moody squamous cell lung carcinoma in vivo and in vitro.
TW, Elsasser T, Cuttitta F (1996) Adrenomedullin Journal of Clinical Endocrinology & Metabolism
expression in human tumor cell lines. Its potential role 81(10):37936.
as an autocrine growth factor. J Biol Chem 271: 143. Nussbaum SR, Gaz RD,Arnold A (1990) Hypercalcemia
2334551. and ectopic secretion of parathyroid hormone by an
129. Minina E, Wenzel HM, Kreschel C, Karp S, Gafeld W, ovarian carcinoma with rearrangement of the gene for
McMahon AP, et al. (2001) BMP and Ihh/PTHrP parathyroid hormone. New England Journal of Medi-
signaling interact to coordinate chondrocyte pro- cine 323(19):13248.
liferation and differentiation. Development 128(22): 144. Oehler MK, Rees MC, Bicknell R (2000) Steroids and
452334. the endometrium. Curr Med Chem 7:54360.
130. Morony S, Capparelli C, Sarosi I, Lacey DL, Dunstan 145. Orr W, Varani J, Gondex MK, Ward PA, Mundy GR
CR, Kostenuik PJ (2001) Osteoprotegerin inhibits (1978) Chemotactic responses of tumor cells to prod-
osteolysis and decreases skeletal tumor burden in syn- ucts of resorbing bone. Science 203:1769.
geneic and nude mouse models of experimental bone 146. Overall CM, Lopez-Otin C (2002) Strategies for MMP
metastasis. Cancer Res 61:44326. inhibition in cancer: innovations for the post-trial era.
131. Moseley JM, Kubota M, Diefenbach-Jagger H, Nat Rev Cancer 2:65772.
Wettenhall RE, Kemp BE, Suva LJ, et al. (1987) Parathy- 147. Ozbas S, Dafydd H, Purushotham AD (2003) Bone
roid hormone-related protein puried from a human marrow micrometastasis in breast cancer. Br J Surg
lung cancer cell line. Proceedings of the National 90:290301.
Academy of Sciences of the United States of America 148. Paget S (1889) The distribution of secondary growths
84(14):504852. in cancer of the breast. Lancet 1:5712.
132. Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan 149. Philbrick WM, Dreyer BE, Nakchbandi IA, Karaplis AC
ME, McClanahan T, Murphy E, Yuan W, Wagner SN, (1998) Parathyroid hormone-related protein is
Barrera JL, Mohar A, Verastegui E, Zlotnik A (2001) required for tooth eruption. Proc Natl Acad Sci USA
Involvement of chemokine receptors in breast cancer 95(20):1184651.
metastasis. Nature 410:506. 150. Pirola CJ, Wang HM, Strgacich MI, Kamyar A, Cercek
133. Mundy GR, Rick ME, Turcotte R, Kowalski MA (1978) B, Forrester JS, et al. (1994) Mechanical stimuli induce
Pathogenesis of hypercalcemia in lymphosarcoma cell vascular parathyroid hormone-related protein gene
leukemia. Role of an osteoclast activating factor-like expression in vivo and in vitro. Endocrinology
substance and a mechanism of action for gluco- 134(5):22306.
corticoid therapy. American Journal of Medicine 151. Plotkin LI, Weinstein RS, Partt AM, Roberson PK,
65(4):6006. Manolagas SC, Bellido T (1999) Prevention of osteo-
134. Mundy GR, Wilkinson R, Heath DA (1983) Compara- cyte and osteoblast apoptosis by bisphosphonates
tive study of available medical therapy for hypercal- and calcitonin. Journal of Clinical Investigation
cemia of malignancy. American Journal of Medicine 104(10):136374.
74(3):42132. 152. Powell GJ, Southby J, Danks JA, Stillwell RG, Hayman
135. Mundy GR. Hypercalcemia (1995) In: Mundy GR, JA, Henderson MA, Bennett RC, Martin TJ (1991)
editor. Bone remodeling and its disorders. London: Localization of parathyroid hormone-related protein
Martin Dunitz :88103. in breast cancer metastasis: increased incidence in
136. Mundy GR (2002) Metastasis to bone: causes, con- bone compared with other sites. Cancer Res 51:
sequences and therapeutic opportunities. Nat Rev 305961.
Cancer 2:58493. 153. Purohit OP, Radstone CR, Anthony C, Kanis JA,
137. Nagai M, Kyakumoto S, Sato N (2000) Cancer cells Coleman RE (1995) A randomised double-blind com-
responsible for humoral hypercalcemia express mRNA parison of intravenous pamidronate and clodronate in
172 Bone Resorption

the hypercalcaemia of malignancy. British Journal of Its role in osteoblast proliferation and differentiation
Cancer 72(5):128993. in vitro and bone formation in vivo. J Cell Physiol1
154. Ralston SH, Gallacher SJ, Patel U, Campbell J, Boyle IT 96:5162.
(1990) Cancer-associated hypercalcemia: morbidity 170. Sanders JL, Chattopadhyay N, Kifor O, Yamaguchi T,
and mortality. Clinical experience in 126 treated Brown EM (2000) Extracellular calcium-sensing recep-
patients. Annals of Internal Medicine 112(7):499504. tor (CaR) expression and its potential role in parathy-
155. Reddi AH, Roodman D, Freeman C, Mohla S (2003) roid hormone-related peptide (PTHrP) secretion in
Mechanisms of tumor metastasis to the bone: chal- the H-500 rat Leydig cell model of humoral hypercal-
lenges and opportunities. J Bone Miner Res 18:190 cemia of malignancy. Biochem Biophys Res Commun
4. 269:42732.
156. Reinholz GG, Getz B, Pederson L, Sanders ES, 171. Sasaki A, Boyce BF, Story B, Wright KR, Chapman M,
Subramaniam M, Ingle JN, et al. (2000) Bisphospho- Boyce R, Mundy GR, Yoneda T (1995) The bisphos-
nates directly regulate cell proliferation, differentia- phonate residronate reduces metastatic human breast
tion, and gene expression in human osteoblasts. cancer burden in bone in nude mice. Cancer Res
Cancer Research 60(21):60017. 55:35517.
157. Reinholz GG, Getz B, Sanders ES, Karpeisky MY, 172. Sato K, Fujii Y, Kasono K, Ozawa M, Imamura H, Kanaji
Padyukova NSh, Mikhailov SN, Ingle JN, Spelsberg TC Y, Kurosawa H, Tsushima T, Shizume K (1989) Parathy-
(2002) Distinct mechanisms of bisphosphonate action roid hormone-related protein and interleukin-1 alpha
between osteoblasts and breast cancer cells: identity of synergistically stimulate bone resorption in vitro and
a potent new bisphosphonate analogue. Breast Cancer increase the serum calcium concentration in mice in
Res Treat 71:25768. vivo. Endocrinology 124(5):21728.
158. Remuzzi G, Perico N, Benigni A (2003) New therapeu- 173. Sato K, Onuma E, Yocum RC, Ogata E (2003) Treatment
tics that antagonize endothelin: promises and frustra- of malignancy-associated hypercalcemia and cachexia
tions. Nat Rev Drug Discov 1:9861001. with humanized anti-parathyroid hormone-related
159. Riggs BL, Hartmann LC (2003) Selective estrogen- protein antibody. Semin Oncol. 30(5 Suppl 16):16773.
receptor modulators mechanisms of action and 174. Schluter KD, Katzer C, Piper HM (2001) A N-terminal
application to clinical practice. N Engl J Med 348: PTHrP peptide fragment void of a PTH/PTHrP-
61829. receptor binding domain activates cardiac ET(A)
160. Rizzoli R, Pache JC, Didierjean L, Burger A, Bonjour JP receptors. Br J Pharmacol1 32:42732.
(1994) A thymoma as a cause of true ectopic hyper- 175. Schluter KD, Piper HM (1998) Cardiovascular actions
parathyroidism. Journal of Clinical Endocrinology & of parathyroid hormone and parathyroid hormone-
Metabolism 79(3):91215. related peptide. Cardiovascular Research 37(1):3441.
161. Roodman GD (2003) Role of stromal-derived cyto- 176. Schmidt-Kittler O, Ragg T, Daskalakis A, Granzow M,
kines and growth factors in bone metastasis. Cancer Ahr A, Blankenstein TJ, Kaufmann M, Diebold J,
97(3 Suppl):7338. Arnholdt H, Muller P, Bischoff J, Harich D, Schlimok
162. Roodman GD (2001) Biology of osteoclast activation G, Riethmuller G, Eils R, Klein CA (2003) From latent
in cancer. J Clin Oncol 19:356271. disseminated cells to overt metastasis: Genetic analy-
163. Rosenbaum E, Carducci MA (2003) Pharmacotherapy sis of systemic breast cancer progression. Proc Natl
of hormone refractory prostate cancer: new develop- Acad Sci USA 100:773742.
ments and challenges. Expert Opin Pharmacother 177. Seymour JF, Gagel RF, Hagemeister FB, Dimopoulos
4:87587. MA, Cabanillas F (1994) Calcitriol production in
164. Rosenthal N, Insogna KL, Godsall JW, Smaldone L, hypercalcemic and normocalcemic patients with non-
Waldron JA, Stewart AF (1985) Elevations in circulat- Hodgkin lymphoma. Annals of Internal Medicine
ing 1,25-dihydroxyvitamin D in three patients with 121(9):63340.
lymphoma-associated hypercalcemia. Journal of Clin- 178. Seymour JF, Grill V, Martin TJ, Lee N, Firkin F (1993)
ical Endocrinology & Metabolism 60(1):2933. Hypercalcemia in the blastic phase of chronic myeloid
165. Roudier MP, True LD, Higano CS, Vesselle H, Ellis W, leukemia associated with elevated parathyroid
Lange P, Vessella RL (2003). Phenotypic heterogeneity hormone-related protein. Leukemia 7(10):16725.
of end-stage prostate carcinoma metastatic to bone. 179. Shakespeare W,Yang M, Bohacek R, Cerasoli F, Stebbins
Hum Pathol 34:64653. K, Sundaramoorthi R, Azimioara M, Vu C, Pradeepan
166. Roudier MP, Vessella H, True LD, et al. (2003) Bone his- S, Metcalf C 3rd, Haraldson C, Merry T, Dalgarno D,
tology at autopsy and matched bone scintigraphy nd- Narula S, Hatada M, Lu X, van Schravendijk MR,
ings in patients with hormone refractory prostate Adams S,Violette S, Smith J, Guan W, Bartlett C, Herson
cancer: the effect of bisphosphonate therapy on bone J, Iuliucci J, Weigele M, Sawyer T (2000) Structure-
scintigraphy results. Clin Exp Metastasis 20:17180. based design of an osteoclast-selective, nonpeptide src
167. Russell RG, Rogers MJ (1999) Bisphosphonates: from homology 2 inhibitor with in vivo antiresorptive activ-
the laboratory to the clinic and back again. Bone ity. Proc Natl Acad Sci USA 97:93738.
5:97106. 180. Sherry MM, Greco FA, Johnson DH, Hainsworth JD
168. Sabatini M, Boyce B,Aufdemorte T, Bonewald L, Mundy (1986) Metastatic breast cancer conned to the skele-
GR (1988) Infusions of recombinant human inter- tal system. An indolent disease. Am J Med. 81:3816.
leukins 1 alpha and 1 beta cause hypercalcemia 181. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang
in normal mice. Proc Natl Acad Sci USA85(14):5235 MS, Luthy R, Nguyen HQ, Wooden S, Bennett L, Boone
9. T, Shimamoto G, DeRose M, Elliott R, Colombero A,
169. Safadi FF, Xu J, Smock SL, Kanaan RA, Selim AH, Tan HL, Trail G, Sullivan J, Davy E, Bucay N, Renshaw-
Odgren PR, Marks SC Jr, Owen TA, Popoff SN (2003) Gegg L, Hughes TM, Hill D, Pattison W, Campbell P,
Expression of connective tissue growth factor in bone: Boyle WJ (1997) Osteoprotegerin: a novel secreted
Skeletal Complications of Malignancy: Central Role for the Osteoclast 173

protein involved in the regulation of bone density. Cell 196. Teti A, Migliaccio S, Baron R (2002) The role of the
89:30919. alphaVbeta3 integrin in the development of osteolytic
182. Singer FR, Ritch PS, Lad TE, Ringenberg QS, Schiller bone metastases: a pharmacological target for alterna-
JH, Recker RR, et al. (1991) Treatment of hypercal- tive therapy? Calcif Tissue Int 71:2939.
cemia of malignancy with intravenous etidronate. A 197. Thiebaud D, Jacquet AF, Burckhardt P (1990) Fast
controlled, multicenter study. The Hypercalcemia and effective treatment of malignant hypercalcemia.
Study Group. [erratum appears in Arch Intern Med Combination of suppositories of calcitonin and a
(1991) Oct 151(10):2008]. Archives of Internal Medi- single infusion of 3-amino 1-hydroxypropylidene-1-
cine 151(3):4716. bisphosphonate. Archives of Internal Medicine
183. Singletary SE, Walsh G,Vauthey JN, Curley S, Sawaya R, 150(10):21258.
Weber KL, Meric F, Hortobagyi GN (2003) A role for 198. Thiede MA, Daifotis AG, Weir EC, Brines ML, Burtis
curative surgery in the treatment of selected patients WJ, Ikeda K, et al. (1990) Intrauterine occupancy con-
with metastatic breast cancer. Oncologist 8:24151. trols expression of the parathyroid hormone-related
184. Smith MR, Eastham J, Gleason DM, Shasha D, peptide gene in preterm rat myometrium. Proc Natl
Tchekmedyian S, Zinner N (2003) Randomized Con- Acad Sci USA 87(18):696973.
trolled Trial of Zoledronic Acid to Prevent Bone Loss 199. Thomas RJ, Guise TA, Yin JJ, Elliott J, Horwood NJ,
in Men Receiving Androgen Deprivation Therapy for Martin TJ, Gillespie MT (1999) Breast cancer cells
Nonmetastatic Prostate Cancer. J Urol169:200812. interact with osteoblasts to support osteoclast forma-
185. Smith MR (2003) Management of treatment-related tion. Endocrinol 140:44518.
osteoporosis in men with prostate cancer. Cancer Treat 200. Tisdale MJ. Cancer (2002) Cachexia in cancer patients.
Rev 29:21118. Nat Rev 2:86271.
186. Southby J, Kissin MW, Danks JA, Hayman JA, Moseley 201. Tokuda H, Kozawa O, Harada A, Uematsu T (1998)
JM, Henderson MA, et al. (1990) Immunohistochemi- Tiludronate inhibits interleukin-6 synthesis in
cal localization of parathyroid hormone-related osteoblasts: inhibition of phospholipase D activation
protein in human breast cancer. Cancer Research in MC3T3-E1 cells. Journal of Cellular Biochemistry
50(23):771016. 69(3):2529.
187. Spelsberg TC, Subramaniam M, Riggs BL, Khosla S 202. Torring O, Turner RT, Carter WB, Firek AF, Jacobs CA,
(1999) The actions and interactions of sex steroids Health H 3rd (1992) Inhibition by human interleukin-
and growth factors/cytokines on the skeleton. Mol 1 alpha of parathyroid hormone-related peptide effects
Endocrinol 13:81928. on renal calcium and phosphorus metabolism in the
188. Stewart AF, Horst R, Deftos LJ, Cadman EC, Lang R, rat. Endocrinology 131(1):513.
Broadus AE (1981) Biochemical evaluation of patients 203. Truong NU, deB Edwardes MD, Papavasiliou V,
with cancer-associated hypercalcemia: evidence for Goltzman D, Kremer R (2003) Parathyroid hormone-
humoral and nonhumoral groups. New England related peptide and survival of patients with cancer
Journal of Medicine 303(24):137783. and hypercalcemia. Am J Med. 115(2):11521.
189. Strewler GJ, Budayr AA, Clark OH, Nissenson RA 204. Tuttle KR, Kunau RT, Loveridge N, Mundy GR (1991)
(1993) Production of parathyroid hormone by a malig- Altered renal calcium handling in hypercalcemia of
nant nonparathyroid tumor in a hypercalcemic malignancy Journal of the American Society of
patient. Journal of Clinical Endocrinology & Metabo- Nephrology 2(2):1919.
lism 76(5):13735. 205. Ueno M, Ban S, Nakanoma T, Tsukamoto T, Nonaka S,
190. Strewler GJ, Stern PH, Jacobs JW, Eveloff J, Klein RF, Hirata R, et al. (2000) Hypercalcemia in a patient with
Leung SC, et al. (1987) Parathyroid hormonelike pro- renal cell carcinoma producing parathyroid hormone-
tein from human renal carcinoma cells. Structural and related protein and interleukin-6 International Journal
functional homology with parathyroid hormone. of Urology 7(6):23942.
Journal of Clinical Investigation 80(6):18037. 206. Uy HL, Guise TA, De La Mata J, Taylor SD, Story BM,
191. Strewler GJ (2000) The physiology of parathyroid Dallas MR, et al. (1995) Effects of parathyroid hormone
hormone-related protein. New England Journal of (PTH)-related protein and PTH on osteoclasts and
Medicine 342(3):17785. osteoclast precursors in vivo Endocrinology 136(8):
192. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, 320712.
Martin TJ (1999) Modulation of osteoclast differentia- 207. Uy HL, Mundy GR, Boyce BF, Story BM, Dunstan CR,
tion and function by the new members of the tumor Yin JJ, et al. (1997) Tumor necrosis factor enhances
necrosis factor receptor and ligand families. Endocr parathyroid hormone-related protein-induced
Rev 20:34557. hypercalcemia and bone resorption without inhibiting
193. Suki WN, Yium JJ, Von Minden M, Saller-Hebert C, bone formation in vivo Cancer Research 57(15):
Eknoyan G, Martinez-Maldonado M (1970) Actue 31949.
treatment of hypercalcemia with furosemide. New 208. van der Pluijm G, Lowik C, Papapoulos S (2000)
England Journal of Medicine 283(16):83640. Tumour progression and angiogenesis in bone metas-
194. Suva LJ, Winslow GA, Wettenhall RE, Hammonds RG, tasis from breast cancer: new approaches to an old
Moseley JM, Diefenbach-Jagger H, et al. (1987) A problem Cancer Treat Rev 26:1127.
parathyroid hormone-related protein implicated in 209. van der Pluijm G, Sijmons B, Vloedgraven H, Deckers
malignant hypercalcemia: cloning and expression. M, Papapoulos S, Lowik C (2001) Monitoring metasta-
Science 237(4817):8936. tic behavior of human tumor cells in mice with
195. Taube T, Elomaa I, Blomqvist C, Beneton MNC, Kanis species-specic polymerase chain reaction: elevated
JA (1994) Histomorphometric evidence for osteoclast- expression of angiogenesis and bone resorption stim-
mediated bone resorption in metastatic breast cancer. ulators by breast cancer in bone metastases. J Bone
Bone 15:1616. Miner Res 16:107791.
174 Bone Resorption

210. van der Pluijm G, Vloedgraven HJM, Ivanov B, Robey demonstrates that parathyroid hormone-related
FA, Grzesik WJ, Robey PG, Papapoulos SE, Lwik protein is essential for mammary gland development.
CWGM (1996) Bone sialoprotein peptides are potent Development 125(7):128594.
inhibitors of breast cancer cell adhesion to bone. 222. Yamaguchi T, Chattopadhyay N, Brown EM (2000) G
Cancer Res. 56:194855. protein-coupled extracellular Ca2+ (Ca2+o)-sensing
211. Vargas SJ, Gillespie MT, Powell GJ, Southby J, Danks JA, receptor (CaR): roles in cell signaling and control
Moseley JM, Martin TJ (1992) Localization of parathy- of diverse cellular functions. Adv Pharmacol 47:209
roid hormone-related protein mRNA expression and 53.
metastatic lesions by in situ hybridization. J Bone 223. Yamamoto M, Harm SC, Grasser WA, Thiede MA
Miner Res 7:97180. (1992) Parathyroid hormone-related protein in the rat
212. Viereck V, Emons G, Lauck V, Frosch KH, Blaschke S, urinary bladder: a smooth muscle relaxant produced
Grundker C, et al. (2002) Bisphosphonates locally in response to mechanical stretch. Proc Natl
pamidronate and zoledronic acid stimulate osteopro- Acad Sci USA 89(12):532630.
tegerin production by primary human osteoblasts. 224. Yates AJ, Boyce BF, Favarato G,Aufdemorte TB, Marcelli
Biochemical & Biophysical Research Communications C, Kester MB, et al. (1992) Expression of human trans-
291(3):6806. forming growth factor alpha by Chinese hamster
213. Wada S, Yasuda S, Nagai T, Maeda T, Kitahama S, Suda ovarian tumors in nude mice causes hypercalcemia
S, et al. (2001) Regulation of calcitonin receptor by glu- and increased osteoclastic bone resorption. Journal of
cocorticoid in human osteoclast-like cells prepared in Bone & Mineral Research 7(7):84753.
vitro using receptor activator of nuclear factor-kappaB 225. Yi B, Williams PJ, Niewolna M, Wang Y,Yoneda T (2002)
ligand and macrophage colony-stimulating factor. Tumor-derived platelet-derived growth factor-BB
Endocrinology 142(4):14718. plays a critical role in osteosclerotic bone metastasis in
214. Wakeeld LM, Roberts AB (2002) TGF-beta signaling: an animal model of human breast cancer. Cancer Res
positive and negative effects on tumorigenesis. Curr 62:91723.
Opin Genet Dev 12:229. 226. Yin JJ, Mohammad KS, Kknen S-M, Harris S, Wu-
215. Weber GF (2001) The metastasis gene osteopontin: a Wong RJ, Wessale J, Padley RJ, Garrett IR, Chirgwin JM,
candidate target for cancer therapy. Biochim Biophys Guise TA (2003) A causal role for endothelin-1 in the
Acta 1552:6185. pathogenesis of osteoblastic bone metastases. Proc
216. Weissglas M, Schamhart D, Lowik C, Papapoulos S, Vos Natl Acad Sci USA 100(19):105889.
P, Kurth KH (1995) Hypercalcemia and cosecretion 227. Yin JJ, Selander KS, Chirgwin JM, Dallas M, Grubbs BG,
of interleukin-6 and parathyroid hormone related Wieser R, Massague J, Mundy GR, Guise TA (1999)
peptide by a human renal cell carcinoma implanted TGFb signaling blockade inhibits parathyroid
into nude mice. Journal of Urology 153(3 Pt 1):854 hormone-related protein secretion by breast cancer
7. cells and bone metastases development. J Clin Invest
217. Wood J, Bonjean K, Ruetz S, Bellahcene A, Devy L, 103:197206.
Foidart JM, Castronovo V, Green JR (2002) Novel 228. Yoneda T (2000) Cellular and molecular basis of pref-
antiangiogenic effects of the bisphosphonate com- erential metastasis of breast cancer to bone. J Orthop
pound zoledronic acid. J Pharmacol Exp Ther Sci 5:7581.
302:105561. 229. Yoshimoto K, Yamasaki R, Sakai H, Tezuka U,
218. Woodhouse EC, Chuaqui RF, Liotta LA (1997) General Takahashi M, Iizuka M, et al. (1989) Ectopic production
mechanisms of metastasis. Cancer 80(8 Suppl):1529 of parathyroid hormone by small cell lung cancer in
37. a patient with hypercalcemia. Journal of Clinical
219. Wysolmerski JJ, Broadus AE, Zhou J, Fuchs E, Milstone Endocrinology & Metabolism 68(5):97681.
LM, Philbrick WM (1994) Overexpression of parathy- 230. Zehnder D, Bland R,Williams MC, McNinch RW, Howie
roid hormone-related protein in the skin of transgenic AJ, Stewart PM, et al. (2001) Extrarenal expression of
mice interferes with hair follicle development. Proc 25-hydroxyvitamin d(3)-1 alpha-hydroxylase. Journal
Natl Acad Sci USA 91(3):11337. of Clinical Endocrinology & Metabolism 86(2):888
220. Wysolmerski JJ, Carucci JM, Broadus AE, Philbrick 94.
WM (1994) PTH and PTHrP antagonize mammary 231. Zhao W, Byrne MH, Boyce BF, Krane SM (1999) Bone
gland growth and development in transgenic mice. resorption induced by parathyroid hormone is strik-
Journal of Bone & Mineral Research 9(Suppl 1):S121. ingly diminished in collagenase-resistant mutant mice.
221. Wysolmerski JJ, Philbrick WM, Dunbar ME, Lanske B, J Clin Invest 103:51724.
Kronenberg H, Broadus AE (1998) Rescue of the 232. Zudaire E, Martinez A, Cuttitta F (2003) Adren-
parathyroid hormone-related protein knockout mouse omedullin and cancer. Regul Pept 112:17583.
Index

Accessory cell meta-analysis of eleven trials, Apoptosis


control of RANKL expression 145 of osteoblasts, 146147
by, 89 structure of, 129 in glucocorticoid excess,
role in osteoclastogenesis, study 112113
78 of combined therapy with induced by tumor necrosis
Acidication in bone resorption, estrogen, 142143 factor, 78
36 of safety and efcacy of, ten of osteoclasts
Acid phosphatase, tartrate- year study, 135136 effects of estren in, 146147
resistant. See Tartrate- Alkaline phosphatase role of estrogen in, 110
resistant acid phosphatase bone-specic, effect of protecting osteoclasts from
Activation of matrix calcitonin on levels of, by activation of Erk, 4749
metalloproteases, 2728 141142 by macrophage colony-
Activator protein-1 (AP-1) inhibition of stimulating factor, 95
cJUN and JUND, effect of by risedronate, 131132 in response to tumor necrosis
estrogen on, 71 by tumor necrosis factor, 78 factor receptor-1, 7172
phosphorylation of, response to Anabolic effects signaling pathway to, from
activation of mitogen- of parathyroid hormone, tumor necrosis factor or
activated protein kinase, combination with interleukin-1, 76
67 antiresorptive therapies, Arginine-glycine-aspartic acid
transcription factor complex, 143 (RGD) sequences,
c-fos proto-oncogene in, of prostaglandins, 98 extracellular matrix
9394 Androgen deciency proteins containing, 40
tumor necrosis factor receptor- impairment of bone response Arteries, calcication of, in
associated factor (TRAF) to physical loading in, osteoprotegerin-decient
role in activation of 111 mice, 10, 95
ADAM8 meltrin, effect on osteoporosis due to, 14 Arthritis
osteoclast formation, 13 Angiogenesis, in bone metastases, association with estrogen
Adhesion effect of bisphosphonates deciency of menopause,
integrin-mediated, 3940 on, 165 67
of osteoclasts, 36 Annexin II, effect on osteoclast inammatory, periarticular
role of Src kinases in, 41 formation, 15 bone destruction in, 67
Adrenomedullin vasoactive Antigens Ascorbic acid, effect on fusion of
peptide, production of, by macrophage-specic, precursor cells in the
cancers, 164 expression by pre- extracellular matrix, 12
Albers-Schnberg disease, osteoclasts, 2 ATP-bisphosphonate analogues,
119122 stem cell antigen-1 (Sca-1+), 4 cytotoxic, 128
Alendronate, 130131 Antiresorptive therapies, meta- Atrasentan, blocking of endothelin
comparison with raloxifene, analysis of, 145146 A receptor by, 164165
and in combination with Apolipoprotein E gene promoter, Avascular necrosis of the jaw,
parathyroid hormone, control of osteoprotegerin caused by
study, 145 expression by, 1011 bisphosphonates, 136
176 Index

Basic multicellular unit (BMU), interactions with tumors, osteoclast resorptive activity
58, 6061 vicious cycle in bone decrease mediated by,
Basolateral plasma membrane, metastasis, 163164 1112, 14
osteoclast, sodium- loss of postmenopausal osteoporosis
potassium pump of, 37 in cancer patients, 166 study using, 141142
B cell after discontinuing hormone therapy with, for juvenile
B-lymphocyte, effect of RANKL therapy, 143 Pagets disease, 115
gene ablation on, 8 disuse-induced, 6263 Calcitonin receptors
differentiation of Bone disuse, vi expression of, in the osteoclast
role of E2A and EBF gene rate of bone loss due to, precursor stage, 3
products in, 93 6263 inactivation of the osteoclast
transcription factors in Bone marrow by, 37
lymphopoiesis, 92 cells of, as a source of signal transduction from, 4647
in lymphoma, parathyroid- osteoclast precursors, 91 Calcium, role of bone in
related protein levels in, transplantation of, to cure managing stores of, 1
153 osteopetrosis, 12 Camurati-Engelmann disease
B cell lineage specic activation Bone morphogenic proteins (CED), 122
factor (BSAP), gene coding (BMPs), stimulation of Cancellous bone, resorption of,
for, 93 differentiation to 6062
Bicarbonate, active exchange of, osteoblasts by, 7980 Carbonic anhydrase, source of
for chloride, in the Bone remodeling unit (BRU), protons in resorption, 38
basolateral membrane, 38 dened, 58 Carbonic anhydrase II
Bisphosphonates Bone scans, to identify osteolytic deciency of, autosomal
efcacy of, long-term, 135136 and osteoblastic recessive, 121122
inhibition by, of osteoclast metastases, 158 mutation in, leading to
differentiation, 12 Bone resorption, structural osteopetrosis, 36
overview, 128130 implications of, 6264 Cathepsins, v
reduction of skeletal-related Bone structural unit (BSU) B, detecting in osteoclasts,
events in prostate cancer dened, 59 2526
by, 164165 formation of, vi K
suppression of remodeling by, Breast cancer activation of tartrate-
and accumulation of calcium-sensing receptor resistant acid phosphatase
microdamage, 60 expressed by cells of, 162 by, 25
therapy using hypercalcemia and parathyroid degradation of bone matrix
biochemical markers for hormone-related protein proteins by, 100
monitoring response to, associated with, 153 effect on receptor activator
158 metastasis to bone of kB, 147
in breast and prostate frequency of, and survival, gene for, location on
cancers, 165166 157 chromosome 1, 121
in hypercalcemia of studies of cell line MD-MB- knockout mice, vii
malignancy, 155156 231, 158159 osteopetrosis in knockout
in multiple myeloma, 114 BSU. See Bone structural unit mice, 25, 36
BMP. See Bone morphogenic L
proteins Calcication of the arteries, in effects on bone resorption,
BMU. See Basic multicellular unit osteoprotegerin-decient 2527
itBone mice, 10, 95 in osteoclasts, 26
environment of, and Calcineurin, activation of NFATs role in bone resorption, 24
osteoclastogenesis, 2 by, 94 c-Cbl
formation of Calcitonin association with Pyk2
inhibition of, in the tumor combination with pamidronate, downstream of integrins,
necrosis factor signaling to treat hypercalcemia, 4243
pathways, 71 156 in signaling mediated by
role of parathyroid-related meta-analysis of thirty trials, macrophage colony-
protein in, 152 146 stimulating factor, 49
Index 177

CD4+ cells, role in bone loss Cholesterol synthesis, inhibition Collagen genes
caused by estrogen of, by bisphosphonates, interleukin-1 inhibition of
deciency, 69 12 transcription of, 7778
CD4 molecule, expression of, in Chromosomes tumor necrosis factor
virus-induced cell fusion, 1, q21, mutation in regulation of transcription
12 pycnodysostosis, 121 of, 7677
Cell lines, for estrogen-positive 8, mutation in Pagets disease, Collagen/laminin receptor a2b1,
human breast cancer, ZR- 115 of mammalian osteoclasts,
75.1 study, 164165 11, linkage to high bone 40
Cell surface, cytokines, receptors density in the mouse, Colony-forming unit for
and signaling at, 94101 118119 granulocytes and
Cell-to-cell contact, in osteoclast 16, mutation in autosomal macrophages (CFU-GM)
formation, 91 dominant osteopetrosis common myeloid progenitor in
Cellular mechanisms of bone type II, 121 the progression to, 3
destruction, 160161 18 expression of membrane
c-Fms (macrophage colony- mutation in familial tyrosine kinase by, 5
stimulating factor expansile osteolysis, 116 hypercalcemia due to increased
receptor) mutation in Pagets disease, production of, 155
control of expression of, by 113 See also Granulocyte-
PU.1, 92 c-Jun N-terminal kinase macrophage colony
downregulation of, by phosphorylation, effect of forming units
macrophage colony- estrogen on, 14 Colony-forming unit for
stimulating factor, 95 Class II transactivator (CIITA), macrophages (CFU-M),
signaling by, 4749 expression of the gene for, transformation to pre-
c-fms gene, encoding of after ovariectomy, 70 osteoclast, 611. See also
membrane tyrosine kinase Clinical disorders of bone Macrophage colony-
by, 5 resorption stimulating factor
c-fos association with alterations, Colony-stimulating factors (CSFs)
osteopetrosis in -/- mice, 100 108127 -1, and osteoclast apoptosis,
proto-oncogene, in the AP-1 regulatory role in vivii
transcription factor osteoclastogenesis, 75 role in osteoclast development,
complex, 5, 9394 therapies for, in malignancy, 3
transcription factor, for 165166 See also Granulocyte-
activation of genes for Clodronate macrophage-colony
the osteoclast phenotype, osteolysis treated with, studies, stimulating factor;
75 134 Macrophage colony-
Chemotaxis, in tumor cell structure of, 129 stimulating factor
metastasis to bone, Cochrane Collection system, 145 Combination therapies
159160 Collagen anabolic, to prevent bone loss,
Children, treating with degradation of, role of 143146
bisphosphonate, 136 cathepsin K in, 25 antiresorptive, 142146
Chloride channel-7 type 1 Common lymphoid progenitor
acidication of the extracellular in bone, 24 (CLP), differentiating
bone resorbing inhibition of production by from the myeloid
compartment by, 37 tumor necrosis factor and lineage, 4
chromosome 16 location of the interleukin-1, 76 Common myeloid progenitor
gene for, 121 Collagenase-1 (matrix (CMP), progression to
mutation in metalloprotease-1), 27 colony-forming unit for
leading to osteopetrosis, 36, Collagenase-3 (matrix granulocytes and
121 metalloprotease 13), 27 macrophages, 3
malignant osteopetrosis secretion by osteoclasts, 38 Connective tissue growth
resulting from, 117118 Collagen gel, osteoclast migration factor (CTGF), as an
in the rufed border through, effects of matrix osteoblast stimulatory
membrane, 38 metalloproteases on, 28 factor, 162
178 Index

Control of RANKL expression, DAP12 Disintegrase meltrin-alpha, cell


89 formation of a complex with fusion involvement, 12
Cortical bone, resorption of, surface cell receptor Disintegrins, eichistatin,
5860 TREM2, 116 inhibition of
response to disuse, 6364 immunoreceptor tyrosine- multinucleated osteoclasts
Cortical endosteal surfaces, based activation motif by, 12
resorption of, 60 (ITAM) of, 50 Drop-out rate, in a clinical trial of
c-Src, regulation of signals DAP12 -/- FcR-gamma -/- risedronate for treating
mediated by avb3 integrin, double knockout mice, osteoporosis, 131
4142 osteoclast formation in,
c-Src kinases 98 E-cadherin, role in cell fusion, 12
roles in cell adhesion and Deafness, in familial expansile Eichistatin
migration, 41, 4647 osteolysis, 116 effect of, on formation of
roles in cell migration, Demineralized bone, association multinucleated osteoclasts,
response to macrophage- with cathepsin K 12
colony-stimulating factor, deciency, 25 inhibition of bone loss in
95 Dendritic cells, RANK in, 910 hyperparathyroid mice by,
c-Src -/- mice, defects in, effects Differentiation, viiviii 40
on bone phenotype, of cartilage, role of Electron microscopy, to follow
98100 parathyroid-related resorption of cancellous
culture systems, 23 protein in, 152 bone, 62
Cushings syndrome, iatrogenic, of hematopoietic cell lineages, Electrophoretic mobility shift
secondary osteoporosis 92 assays (EMSA), of
due to, 112 myeloid, 4 activation of NF-kB and
Cyclic adenosine monophosphate of osteoblasts, cytokine effects activator protein-1 in
(cAMP) on, 7980 osteoblasts, 73
enhancement of osteoclast of osteoclasts, 123 Encephalopathy in Rett
development by, 3 cytokine effects on, 7476 syndrome, 116117
enhancement of osteoclast differentiation of, stimulators Endocrine disorders resulting in
development by receptors and repressors 1316 osteoporosis, 110113
for, 98 late, 1113 Endothelin-1 (ET-1),
Cyclooxygenase 2 (COX2), macrophage colony- vasoconstrictor, as an
effect of IL-1 and TNF stimulating factor role in, osteoblast-stimulatory
on, 78 49 factor, 164165
Cysteine protease role of macrophage colony- Endothelin receptor antagonists,
inhibition of, to block bone stimulating factor in, 166
resorption, 25 9495 Engineering of the skeleton, 1
in osteoclasts, 100 1,25-Dihydroxyvitamin D Environmental/genetic disorders,
See also Cathepsin K effect of 113114
Cytokines on cathepsin L levels, in Epiphyseal growth plate, effect of
effects of osteoclasts, 2627 tartrate-resistant acid
on osteoblast survival, 80 on RANKL expression, 8 phosphatase disruption
on osteoclastogenesis, 15 enhancement of osteoclast on, 25
inammatory development by, 3 Estradiol
effect on matrix hypercalcemia associated with, effect on bone mass density,
metalloprotease levels, 28 in hematologic clinical trials, 138
role in bone disease, vi, malignancies, 153154 effect on osteoprotegerin, 11
6790 resistance to Estren (4-estren-3a,17-diol),
regulation of osteoblast gene caused by nuclear factor effects on apoptosis of
expression by, 7679 kappa B, 67 osteoclasts and
tumor-necrosis factor-related induced by tumor necrosis osteoblasts, 146147
activation-induced, 6 factor, 8081 Estrogen
Cytoskeletal proteins, recruitment Diseases, involving osteoclast as an anti-resorptive agent on
of, 43 activity extremes, vii bone, 137139
Index 179

effects of fas-activated death domain Genetics


on osteoclast activity, 110111 (FADD), activation of, affecting osteoclast
on tumor necrosis factor triggering apoptotic development and function,
production and action, caspases, 73 91107
6971 Fibroblast growth factor 2 (FGF- inherited diseases with
regulation of interferon-gamma 2), induction of osteoclast dysfunctional osteoclases,
by, 7071 formation by, effect of 117118
trials evaluating therapy using parathyroid hormone on, Giant cell tumors
changes in bone loss, 137 14 human, matrix metalloprotease
ultra-low-dose, 138139 Fibrous dysplasia, interleukin-6 9 mRNA expressed by,
Estrogen deciency association with bone 27
impairment of bone response pathology in, 97 interleukin-6 association with
to physical loading in, 111 Flow cytometry, delineation of bone pathology in, 97
indolent inammatory state progression from stem cell Glucocorticoid receptors (Grs),
associated with, 67 to osteoclast phenotype interaction with nuclear
interaction with macrophage using, 2 factor-kappa B, 81
colony-stimulating factor, Fluorescent-activated cell sorting Glucocorticoids
5 (FACS), for analysis of with calcitonin administration,
stimulation of cytokine unfractionated bone for treating hypercalcemia,
production by, 6768 marrow, 70 156157
Etidronate Focal adhesion kinase (FAK) effect on osteoprotegerin levels,
effect on hypercalcemia of family, Pyk2 of, 4142 11
malignancy, 156 fos transcription factor, role in excess of, osteoporosis
impairment of bone osteoclast differentiation, associated with, 112113
mineralization by, 133134 5 osteoporosis from, treatment
structure of, 129 Fractures with bisphosphonates,
Evolution, selection for osteoclast healing of, in tumor necrosis 130
behaviors that are factor receptor-decient Glycoproteins
structurally benecial, 60 mice, 80 gp-130 activator protein in the
Expansile skeletal pathological, in patients with interleukin-6 receptor
hyperphosphatasia (ESH), advanced cancer, 159 complex, 97
116 gp 160, expression of, in virus-
Experimental therapies, for bone Gallium nitrate, to treat induced cell fusion, 12
metastases, 165166 hypercalcemia of SHPS-1, role in macrophage
Extracellular matrix components, malignancy, 157 fusion, 12
recognition of, by Gelatinase B (matrix Glycosaminoglycans, complex
osteoclasts, 40 metalloprotease 9) with cathepsin K, in
Extravasation, of metastatic secretion by osteoclasts, 38 collagen degradation, 26
cancer cells, 159 substrates of, 27 Golgi complex, lysosomal
Gelsolin, as a phosphatidyl enzymes of, 3738
F-actin, regulation of the motility inositol-3-kinase Gorham-Stout disease (GSD),
of, by gelsolin, 43 dependent target in 113114
Familial expansile osteolysis osteoclasts, 43 interleukin-6 association with
(FEO), 116 Genes bone pathology in, 97
deletion of osteoprotegerin osteoblast, cytokine regulation gp-130 activator protein, 97
coding sequence in, 11 of the expression of, 7679 G protein-coupled receptors,
Farnesyldiphosphate synthase, Pax5, encoding for B cell calcitonin receptor,
inhibition of, by nitrogen- lineage specic activation signaling downstream of,
substituted factor, 93 4647
bisphosphonates, 156 for RANKL, effects of ablation Granulocyte-macrophage colony
Farnesyl pyrophosphatase, in a transgenic mouse, 8 forming units, increased
inhibition of, by nitrogen- in skeletal cells production of
containing IL-1-regulated, 7677 parathyroid-related
bisphosphonates, 128 TNF-regulated, 7677 protein mediated by, 155
180 Index

Granulocyte-macrophage-colony Hormones, effect on osteoclast Insulin-like growth factor-I (IGF-


stimulating factor (GM- differentiation, 1415. See I)
CSF) also Estrogen effect of tumor necrosis factor
role in osteoclast formation, 3 Howships lacunae (resorption on expression of, 78
simulation of secretion from T- cavity), 59 effects on bone density, 11, 14
cells by annexin II, 15 Hydration, to reduce calcium upregulation of RANKL by, 8
Gray-lethal mutation levels in hypercalcemia, Integrin receptors, attachment of
autosomal dominant 155 cell to matrix via, 37
osteopetrosis caused by, Hydrogen ion pump, of lysosome Integrins, gure, 38
118119 membranes, 24 avb3
in malignant osteopetrosis, Hydrogen peroxide, effect on defects in b3 -/- mice,
117118 osteoclast differentiation, 100101
vesicular trafc defect in mice, 15 role in bone resorption, 13,
36 Hydroxyapatite, of bone, 24 100101
Growth plates, lengthened, in Hypercalcemia role in regulation of breast
matrix metalloprotease 9 in breast cancer patients, 159 cancer metastases, 160
knockout mice, 28 in hyperparathyroidism, Integrin signaling, 3940
111112 Interferon-b (INFb), down
Haversian canal. See Bone Hypercalcemia of malignancy, regulation ofc-fos
structural unit 151157 expression by, 48
HEF1, phosphorylation and interleukin-6 association with Interferon-gamma (INF-gamma)
association with FAK and bone pathology in, 97 degradation of TRAF6
paxillin, 4647 Hyperostosis corticalis deformans promoted by, 48
HEK 293 cells juvenilis (juvenile Pagets expression in macrophages,
cell adhesion role of, 4243 disease), remodeling in, after ovariectomy, 70
effect of calcitonin in, 46 114115 suppression of
Helix-loop-helix proteins, in B Hyperparathyroidism, 111112 osteoclastogenesis by, 10
cell differentiation, Hyperthyroidism, bone Interferon-B, induction of, in
encoded by E2A and EBF remodeling in, 112 osteoclast precursor cells,
genes, 93 5
Hematogenous cells, origin of Ibandronate, 132133 Interleukin-1 (IL-1)
osteoclasts in, 12 Immune system, DAP12/TREM2 antiapoptotic action of, 76
Hematologic malignancies, co-receptors for, 50 effect on bone loss, vi
hypercalcemia associated Immunoreceptor tyrosine-based modulation of the renal
with, 153154 activation motif (ITAM) effects of parathyroid
Hematopoietic stem cells (HSC) of DAP12, 50, 98 hormone-related protein
origin of osteoclasts from, 2, 91 signaling pathway leading to by, 152153
pluripotent, progression to osteoclast formation, production of, associated with
colony-forming unit for 9899 estrogen deciency of
macrophages, 4 Inherited diseases with menopause, 67
Hemi-osteonal model, of dysfunctional osteoclasts, regulation by,
cancellous bone 117118 of bone resorption, 96
multicellular units, 60 Inhibition of matrix metalloprotease 1
HIV gp 160 envelope of alkaline phosphatase, by and matrix
glycoprotein, expression tumor necrosis factor, 78 metalloprotease 9 in
of, in virus-induced cell of cathepsin K, by antisense osteoclasts, 27
fusion, 12 molecules, 26 signaling pathways of, 7374
Hodgkins disease, hypercalcemia of collagen synthesis, by tumor stimulation of
in, 153154 necrosis factor, 76 osteoclastogenesis by,
Hormone replacement therapy of mitogen-activated protein requirement for TRAF6,
combination with parathyroid kinase activity by PTP 7273
hormone treatment, 144 epsilon, 45 Interleukin-1a (IL-1a), effect on
gonadal, meta-analysis of fty- of osteoclastic bone resorption cathepsin L levels, in
seven trials, 146 in vitro, 40 osteoclasts, 2627
Index 181

Interleukin-1 receptor accessory Janus kinase-signal transducer Levormeloxifene, clinical trial of,
protein (IRAcP), and activator of withdrawal from
interaction with transcription (JAK-STAT), development, 140141
interleukin-1 receptor, inhibition of, 45 Life-cycle diagram, representing
96 pathway for signal bone multicellular unit
Interleukin-1-regulated genes, in transduction through activities, 60
skeletal cells, 7677 cytokine receptors, 97 Lineage of osteoclasts, 14
Interleukin-6 (IL-6) Juvenile Pagets disease, alterations in differentiation to
effect on cathepsin L levels, in 114115 change bone resorption,
osteoclasts, 2627 37
hypercalcemia potentiated by, Ketoconazole, effect on Lipoxygenase, 12/15-, mutation
155 osteoclasts in culture, 12 resulting in high bone
mediated by parathyroid Kinases density, 118119
hormone-related protein, c-Jun N-terminal, Loading, effect on bone loss,
152153 phosphorylation of, 14 63
regulation of osteoclast focal adhesion kinase(FAK) Low density lipoprotein receptor-
differentiation by, in family, 4142 related protein 5 (LRP5)
murine marrow cultures, 3 interleukin-1 receptor- gene, mutations in
stimulation of bone resorption associated kinase (IRAK), osteopetrosis type I,
by, 97 96 119120
Interleukin-7 (IL-7) interleukin receptor-associated Lung cancer, metastasis to bone,
levels of, response to kinase-1 (IRAK-1), inhibition by a
ovariectomy and bone activation of, 7374 monoclonal antibody,
loss, 68 Janus kinase-signal transducer trial, 162
receptor for, control of and activator of Lymph node structure, effect of
expression by PU.1, 92 transcription, 45 RANKL gene ablation on,
Interleukin-7 receptor a-chain mitogen-activated protein 8
(IL-7Ra), differentiation of kinase MAPK), 67 Lymphocytes
common lymphoid p38, TRAF signaling via, B-lymphocytes, effect of
progenitor from myeloid 7273 RANKL gene ablation
lineage by, 4 P13, activation of, 43 on, 8
Interleukin-11 (IL-11), in Syk kinase, 98 development of, roles of
osteolytic lesions induced See also c-Src kinases; receptor activator nuclear
by parathyroid hormone- Phosphatidyl inositol-3 factor-kappa ligand, 95
related protein, 162 kinase; Protein kinase C; T-lymphocytes
Interleukin-13 (IL-13), effect on Protein tyrosine kinases; modulation of actions of, by
fusion of macrophages, Src kinases; Tyrosine estrogen, 14
12 kinases RANKL expression in, 89
Interleukin-18 (IL-18), decrease in Lysosomal enzymes, synthesis by
osteoclast formation by, Large cell lymphoma, 1,25- the osteoclast, 3738
165 (OH)2D3 dysregulation in, Lysosome, extracellularbone-
Interleukin receptor-associated 154 resorbing compartment as
kinase-1 (IRAK-1) Lasofoxifene, effects of, on bone the functional equivalent
activation of, by interleukin-1 density, 141 of, 39
binding to its receptor, Legumain, inhibition of osteoclast
7374 formation by, 15 Macrophage colony-stimulating
binding of TRAF6 by, 96 Leptin, effect on osteoprotegerin factor (M-CSF)
Internalization, in bone levels, 11 levels of, in b3 -/- mice, 101
resorption, 36 Leukemia, adult T-cell, in osteolytic lesions induced by
Ion transport systems, osteoclast, parathyroid-related parathyroid hormone-
37 protein levels in, 153 related protein, 162
ITAM. See Immunoreceptor Leukemia inhibitory factor(LIF), phosphorylation of
tyrosine-based activation production in response to microphthalmia
motif resorption stimuli, 97 transcription factor by, 13
182 Index

receptor for, expression of in -13, synthesis by osteocytes, 28 Mindronate, structure of, 129
the basolateral plasma -14, substrates of, 27 Mitogen-activated protein kinase
membrane, 37 produced by osteoclasts, v, 78 (MAPK), stimulation of
role in osteoclastogenesis, 35, stimulation by nuclear the system in menopause,
7, 91, 9495, 160161 factor-kappa B, 67 67
signaling by, SHP-1 as a roles of Matrix metalloproteases. See
negative regulator of, in bone resorption, 24, 2729 MMPs
4445 in tumor cell invasion, 159 Molecular mechanisms, of bone
Macrophage fusion receptor M-CFS. See Macrophage colony- resorption, 3439
(SHPS-1), role in stimulating factor entries Monoclonal antibody, to
multinucleation of Measles, association with Pagets parathyroid hormone-
osteoclasts, 12 disease, 113 related protein, to treat
Macrophage metalloelastase Mechanical input, response of hypercalcemia of
(matrix metalloprotease bone to loading and malignancy, 161162
12), elastin as a substrate unloading, 1516 Monocytic cell, RANK : RANKL
of, 27 Mediators, of hypercalcemia of signal transduction in,
Macrophage progenitor, malignancy, 152155 910
differentiation to, 4 Menin gene, mutation of, 112 Mononuclear osteoclast
Macrophages Mental retardation, in carbonic precursors, fusion of, 12
comparison with osteoclasts, 2 anhydrase II deciency, Motheaten (me/me) mutation
modulation of antigen 122 deciency of SHP-1 protein
presentation by estrogen, Meta-analysis, of antiresorptive tyrosine phosphatase in,
70 therapies, 145146 99100
multinucleated, lineage of, 14 Metalloenzymes, zinc, 121122 of the gene for SHP-1, 4445
Magnetic resonance imaging, Metastases, to bone, 157166 Motility of osteoclasts, in
trabecular bone changes classication of, 157158 gelsolin-/- mice, 43
seen using, 63 Methyl-CpG-binding protein-2 Multiple endocrine
Malignancy, skeletal (Mecp2), mutation in the neoplasia I (MEN1),
complications of, viiviii, gene for, in Rett hyperparathyroidism in,
151174 syndrome, 116117 112
Mannose, role in cell-cell binding, Microphthalmia-associated Multiple endocrine neoplasia II
12 transcription factor (MEN2), 112
Mannose-6-phosphate receptors, (MITF) Multiple myeloma, 114
role in bone resorption, 38 activation of tartrate-resistant hypercalcemia in, 153
Marble bone disease, in the acid phosphatase pamidronate for treating, 133
osteopetrotic mouse, 5 expression by, 25 Multiple Outcomes of Raloxifene
Markers cathepsin K as a Evaluation (MORE) study,
for bone formation and bone transcriptional target of, 140
destruction, 158 100 Mutations
for bone turnover, effects of interaction with PU.1 in effect on osteoclastogenesis
estrogen on, 137138 osteoclast differentiation, and osteoclast function,
for early stem cells, 3 92 vivii, 91107
for osteoclastic phenotypic role in osteoclastogenesis, 1213 frameshift, in the macrophage
features, 6 Microphthalmic mouse, mi/mi, colony-stimulating factor
Marrow transplantation osteoclasts of, 1213 gene, 5
to treat carbonic anhydrase II Migration loss-of-function, in DAP12, 116
deciency, 122 inhibition in Src-/- or Cbl-/- Myeloid differentiation, 4
to treat malignant mice, 4243 Myeloid progenitor cells
osteopetrosis, 117 of metastatic cancer cells, 159 identifying characteristics of, 4
Matrix metalloproteases (MMPs) of osteoclasts, 36 origin of osteoclasts in, 91
-1, collagen cleavage by, 162 activation by macrophage Myeloma, 114
-9, levels of, response to colony-stimulating factor bisphosphonates to treat, 156
cathepsin K levels, 100 activation, 49 hypercalcemia in, 153
-12, elastin as a substrate of, 27 roles of Src kinases in, 41 pamidronate for treating, 133
Index 183

Nasu-Hakola disease (polycystic Osteoblastic metastases, 164165 inherited disorders of,


lipomembranous human breast cancer cell lines 114117
osteodysplasia), 116 in animal models of, 159 inhibition of
in DAP12/TREM2 decient Osteoblasts by bisphosphonates,
individuals, 50 in breast cancer metastases, 129130
National Cancer Institute, test of disorganized new bone by estrogen, 137
tamoxifens effect on formation by, 157158 lineage of, 14
breast cancer in high risk decreased proliferation of, in matrix metalloproteases
subjects, 140 disruption of the LRP5 expressed by, 27
N-cadherins, cell surface, effect of gene, 119 tumor-produced factor
tumor necrosis factor on, differentiation of, effects of stimulating, 161
78 cytokines on, 7980 Osteocytes, matrix
Nitric oxide (NO), effects of, on effect on metalloprotease 13
bone remodeling, 15 of bisphosphonates, 156 production by, 2728
Non-Hodgkins lymphoma, of tumor necrosis factor, Osteogenesis imperfecta, effects
hypercalcemia in, 7681 of bisphosphonates in
153154 osteoclast recruitment inhibitor treating, 136
Non-nitrogen-containing of, 130 Osteoid, degradation of, in the
bisphosphonates, recruitment of, in the wake of bone remodeling cycle,
inhibition of ATP- osteoclast activity, 110111 78
dependent intracellular RUNX2 binding elements of, Osteolysis, in bone metastases,
enzymes by, 156 1011 157158
Nuclear factor-kappa B secretion of receptor activator osteolysis-stimulating factors,
(NF-kappa B), of NF-kappa B ligand by, 162
activation of 147 Osteolytic bone disease
by interleukin receptor- stimulation of, by connective metastases, in breast and
associated kinase-1, 73 tissue growth factor, 162 prostate cancers, 164
in menopause, 67 Osteocalcin, of osteoblasts, in myeloma and breast cancer,
Nuclear factor of activated T 7778 bisphosphonates to treat,
cells-2 (NFAT2), role in Osteoclast activity 156
osteoclast formation, 10 and bone resorption, regulation Osteolytic enzymes, 2433
Nuclear factor of activated T of, 3940 Osteopetrosis
cells-c1 (NFATc1) increase in, clinical disorders autosomal dominant,
signaling molecule in associated with, 108117 118119
osteoclastogenic regulation of, 3457 autosomal dominant type II,
responses, 94 Osteoclast-associated receptor 119122
as a target of FcR-gamma and (OSCAR), expression of, 15 bone-marrow transplantation
DAP12-ITAM mediated Osteoclast differentiation factor, 6 to treat, 12
signaling, 98 Osteoclastogenesis in DAP12, FcR = gamma
Nucleus, transcription regulation inhibition by osteoprotegerin, knockout mice, 98
in the, 9294 1011 due to decreased osteoclast
negative regulation of, by activity, 117122
Oncogenes, v-fos, osteosarcoma tyrosine phosphatase SHP- infantile malignant, mutations
caused by, 5. See also 1, 44 in the TCIRG1 subunit of
Proto-oncogenes without RANKL : RANK, 11 the osteoclast vacuolar
Osteitis brosa cystica, in stimulation by TRAF6, 7273 proton pump, 118
hyperparathyroidism, Osteoclast precursors, c-fos malignant, 2, 117118
111112 overexpression in, 5 in mice after tartrate-resistant
Osteoarthritis, hip, in autosomal Osteoclasts acid phosphatase
dominant osteopetrosis attachment to bone, and disruption, 25
type II, 120 subsequent resorption, 24 in Src, Pyk2, or b3 knockout
Osteoblastic cells, regulation of dened, v mice, 4142
osteoclast formation by, functional features dening the in TRAF6 knockout mice, 910,
161 activity of, 3436 9596
184 Index

Osteopetrotic (op/op) mouse Osteosclerosis, in Gorham-Stout Parathyroid hormone (PTH)


resolution of osteopetrosis in, disease, 113114 combination with bisphonates,
with VEGF, 5 Ovarian cancer, ectopic clinical trials, 143144
treatment of, with membrane- parathyroid production in, comparison of combination
bound macrophage 154 with alendronate or
colony-stimulating Ovariectomy, mechanism of raloxifene, study, 145
factor, 5 bone loss caused by, effect of
Osteopontin 6970 on cathepsin L levels, in
dephosphorylation of, by role of interleukin-6, 97 osteoclasts, 2627
tartrate-resistant acid role of tumor necrosis factor, on protegerin levels, 11
phosphatase, 25 9697 on RANKL expression, 8
modulation by, of anti-tumor Oxygen deprivation of marrow on RANKL : OPG
immune responses, 162 culture, osteoclastogenesis equilibrium, 14
Osteoporosis in, 63 enhancement of osteoclast
dened, 108109 development by, 3
involving osteoclast activity p38 kinase hypercalcemia of
in cancellous bone, 6263 activation by RANK-induced malignancy associated
extremes of, vii phosphorylation, 47 with, 154155
in mice with deleted SHIP, activation of mi/Mitf by, 10 Parathyroid hormone-related
4546 TRAF signaling via, 7273 protein
senile, rate of progression of, p55 tumor necrosis factor antibodies to
110 receptor, effect of to inhibit hypercalcemia in
treating deciency of, 9697 animal studies, 157
bisphosphonates for, 130 p65, subunit of nuclear factor- for reducing breast cancer
lasofoxifene trial, 141 kappa B, transrepression metastases, 166
risedronate for, 131 of vitamin D receptor by, hypercalcemia of malignancy
Osteoporosis-pseudoglioma 8081 associated with, 152
syndrome (OPPG), p65/p50 Nf-kappa B, in hematologic malignancies,
mutation of low density transcription factor 153154
lipoprotein receptor- expression, in role in breast cancer metastases
related protein 5 gene in, osteoclastogenesis, 7476 to bone, 161162
119 p75 tumor necrosis factor Pharmaceuticals, targeting the
Osteoprotegerin (OPG), v, 147 receptor, effect of osteoclast, 128150
binding with RANKL, 1011, deciency of, 9697 Phenotypic features of
161 p130Cas signaling adapter, 43 osteoclasts, 611
breast cancer cell expression of, Pagets disease Pheochromocytoma, from ret
161 dened, 113 proto-oncogene mutation,
as a decoy RANK, 34 interleukin-6 association with 112
levels of bone pathology in, 97 Phosphatidyl inositol-3 kinase
in myeloma, 114 juvenile, defective (PI3-K)
response to estrogen, osteoprotegerin in, 11, activation of, avb3 integrin-
110111 114115 dependent, 43
mutation in, in juvenile Pagets pamidronate for treating, 133 in the Rac-mediated migration
disease, 115 RANKL expression in, 8 pathway, 43
osteoclastogenesis by Pain, in bone cancer, treating, 159, relocalization to the cell
prevention of , 67, 95, 165166 membrane, 49
137 Pamidronate Phosphorylation
regulatory role in, 75 hypercalcemic malignancy mediation of, by protein
production of, in response to treated with, 132133, 156 kinases, 47
bisphosphonates, 156 structure of, 129 of microphthalmia
recombinant, for treating bone Papillary adenocarcinoma of the transcription factor, 13
tumors, 165 thyroid, ectopic of tartrate-resistant acid
Osteosarcoma, v-fos oncogene as parathyroid production in, phosphatase, effect on
a cause of, 5 154 bone resorption, 25
Index 185

Phosphotyrosine-binding (PTB) Prostate cancer, metastasis in RANKL. See Receptor activator of


domain, inhibition of to bone, 164165 NF-kappa B ligand;
tyrosine kinases to the spine, 158 RANKL/RANK signaling
associated with, 4243 Proteases, cathepsin K, 121 RANKL/RANK signaling, 611
Plasma membrane, rufed border, Protein kinase C (PKC), TRAF gene expression induced by, 47
proteins of, 37 signaling via, 7273 regulation of osteoclastogenesis
Plicamycin, inhibition of bone Protein kinase C/[Ca2+]i signaling by, 7576
resorption by, to treat pathway, mediation of upregulation associated with c-
hypercalcemia of response to calcitonin, 47 Fms activation, 49
malignancy, 156 Protein tyrosine kinases (PTKs), See also Receptor activator of
Podosomes, in osteoclasts, role in cellular function NF-kappa B
stability of, 4243 regulation, 4344 Receptor activator of NF-kappa B
Polarization, osteoclast, tyrosine Protein tyrosine phosphatases (RANK)
kinase c-Src role in, 41 (PTPs) association of TRAF6 with,
Polycystic lipomembranous epsilon, activation of osteoclast 7273
osteodysplasia with function by, 4445 breast cancer cell expression of,
sclerosing role in cellular function 161
leukoencephalopathy regulation, 4344 effect of cathepsin K on, 147
(PLOSL), 116 Proto-oncogenes expression of, initiating
Polykaryon, osteoclast, fusion of c-Cbl, association of Pyk2 with, intracellular second
committed cells into, 11 4243 messengers, 95
Polyostotic osteolytic dysplasia, c-fos, in activator protein-1 expression of, in the basolateral
116 (AP-1) transcription factor plasma membrane, 37
Postmenopausal complex, 9394 increased signaling by, due to
estrogen/progestin ret, mutation of, 112 estrogen deciency, 110
intervention (PEPI) trial, PTPs. See Protein tyrosine mutation of the signal peptide
137 phosphatases region of, in familial
bone mass density measured PU.1. See Transcription factors, expansile osteolysis, 116
in, 138 PU.1 Receptor activator of NF-kappa B
Precursors, osteoclast, fusion of, Pycnodysostosis ligand (RANKL), v, 611
12 as an autosomal recessive activation of signaling by, 34
Presenile dementia, in polycystic disorder, 121 antibody to, phase 1 trial,
lipomembranous chromosome 1, q21 mutation 147148
osteodysplasia, 116 in pycnodysostosis, 121 effect of matrix
Prevent Recurrence of cathepsin K mutation in, 13, metalloprotease 14 on,
Osteoporotic Fractures 2629, 36, 100 2829
(PROOF) study, of phenotype, possibly exhibited expression of, in multiple
calcitonin use to by Toulouse-Lautrec, 13 myeloma, 114
prevent bone resorption, Pyk2 gene transcription induced by,
141142 association with p130Cas, 43 through c-fos activation, 5
Primitive neuroectodermal regulation of signals mediated interaction with RANKL to
tumor, hypercalcemia by avb3 integrin, 4142 form osteoclast-like cells,
associated with, 154 Pyrophosphate, structure of, 129 91
Prostaglandins levels of, in myeloma, 114
effects on bone remodeling, Raloxifene as an antiresorptive osteoprotegerin as a decoy
1415 comparison with alendronate receptor for, 95
enhancement of osteoclast in combination with protein structure of, 6
development by, 3 parathyroid hormone, regulation of, by estrogen, 137
E-series, association with study, 145 role of
hypercalcemia, 155 comparison with estrogen, in osteoclast differentiation,
stimulation of bone resorption effect on bone-density, 34
by, 97 140 in osteoclast generation from
synthesis of, effect of IL-1 and meta-analysis of seven trials , hematopoietic precursors,
TNF on, 78 146 160161
186 Index

in the pathogenesis of steps in, vvi SHIP. See SH2-containing


hypercalcemia of structural implications of, 6264 inositol-5-phosphatase
malignancy, 155 ret proto-oncogene, mutation of, (SHIP), regulation of
up-regulation of expression of, disorders associated with, osteoclast function by
by estrogen deciency, 69 112 SHP-1(Src homology 2-domain-
Receptors Rett syndrome, 116117 containing phosphatase 1),
calcium-sensing, regulating Rheumatic joints, RANKL down-regulation of
secretion of parathyroid secretion into, by T- osteoclast function by,
hormone-related protein, lymphocytes, 9 4445
161162 RING nger domain, of TRAF6 Side effects
collagen/laminin, 40 protein, 910 of ibandronate, 133
EP2 and EP4, of Risedronate, 131132 of plicamycin, 156157
prostaglandins, 98 meta-analysis of nine clinical Signaling
estrogen, binding of tamoxifen trials, 145146 activation of cascades for, by
to, 139 structure of, 129 RANK, 910
glucocorticoid, 81 study cytokine pathways for, in
gp-130 activator protein, 97 of combined therapy with skeletal cells, 7174
integrin, attachment of cell to estrogen, 142143 by macrophage colony-
matrix via, 37 of efcacy and safety of, long stimulating factor,
macrophage colony-stimulating term, 135 regulation by SHP-1, 4445
factor, expression by the Rufed border, plasma by macrophage colony-
colony forming unit-GM, 5 membrane, 35, 37 stimulating factor
mannose-6-phosphate, 38 RUNX2 receptor, 47
osteoclast-associated, 15 of osteoprotegerin promoter, molecules involved in, vi
p55 tumor necrosis factor 1011 pathways utilized by receptor
receptor, 9697 transcription factor for activator nuclear factor-
p75 tumor necrosis factor osteoblast differentiation, kappa B, 9596
receptor, 9697 8, 7980 by RANK, 47
regulating osteoclast activity, RANK : RANKL, 611
38 Safety, of bisphosphonates, tumor necrosis factor cascade,
TREM-2, on monocyte-derived 135136 causing inammatory
dendritic cells, 50 Sealing zone, dened, 37 gene activation, 7173
vitronectin/bronectin receptor Second messengers, activation Wnt, in osteoblasts, 119
avb3, 40 through membrane Signal transduction
See also Calcitonin receptors; c- tyrosine kinase receptors, by RANK, 6
Fms; Receptor activator of 5 mediation by nuclear factor-
NF-kappa B (RANK) Secretion, in bone resorption, 36 kappa B, 93
Regulatory pathway for osteoclast Selective estrogen-receptor RANK : RANKL, in the
differentiation, 7475 modulators (SERMs), vii monocytic cell, 910
Remodeling and postmenopausal Skeleton
effect of insulin-like growth osteoporosis, 139142 development of lesions of,
factor-I on, 14 role in treatment of breast 160163
modulation by osteoclasts, vi cancer, 166 disordered architecture of,
Renal toxicity of phosphonates, Sequestosome 1 gene (SQSTM1), 108117
136 113 in malignant disease, 158159
Renal tubular acidosis, autosomal Sex hormones, deciency of, roles of, 1
recessive, 121 leading to decrease in Small cell carcinoma of the lung,
Resorption osteoclast formation, ectopic parathyroid
in cancellous bone, 6062 110111 production in, 154
expression of genes Sex steroids, 14 Sodium-potassium pump, of the
contributing to, 78 SH2-containing inositol-5- basolateral plasma
markers of, change in adjuvant phosphatase (SHIP), membrane, 37
therapy and regulation of osteoclast Spinal cord compression, in
chemotherapy, 158 function by, 45 metastasis to the bone, 159
Index 187

Spleen cells, as a source of in c-fos -/- mice, effect on TRANCE (tumor necrosis factor-
osteoclast precursors, 91 osteoclast development, 94 related activation-induced
Squamous cell carcinomas expression of, in the osteoclast cytokine). See Receptor
factors enhancing parathyroid precursor stage, 3 activator of NF-kappa B
hormone-related protein hydrolysis of phosphoproteins ligand (RANKL)
production by, 153 by, 2425 Transcription
hypercalcemia associated with, levels of, in osteopetrosis type of collagen genes
151152 II, 119122 interleukin-1 inhibition of,
of the lung, ectopic parathyroid of lysosome membranes, 2425 7778
production in, 154 regulation by microphthalmia tumor necrosis factor
Src homology 2-domain- transcription factor, 100 regulation of, 7677
containing phosphatase 1 Tartrate-resistant acid regulation of, in the nucleus,
(SHP-1), down-regulation phosphatase (TRAP)- 9294
of osteoclast function by, positive cells, osteoclasts Transcription factors
4445 dened as, 91 AP-1 family of, 56
Src kinases T-cells c-fos
association with Pyk2 clonal expansion of, regulation for activation of genes for
downstream of integrins, by estrogen, 6971 osteoclast phenotype, 75
4243 lymphocytes regulation of osteoclast and
cell adhesion and migration modulation of actions of, by macrophage
roles of, 41 estrogen, 14 differentiation by, 94
Stem cell antigen-1 (Sca-1+), of RANKL expression in, 89 fos, role in osteoclast
the hematopoietic stem role of, in estrogen deciency differentiation, 5
cell, 4 bone loss, 69 nuclear factor-kappa B
Steroids, sex, effects on osteoclast Telopeptide activation in bone during
differentiation, 14. See also C-, levels in calcitonin menopause, 67
Glucocorticoids administration study, 141 activation of genes for
Stromal cells, role of N-, bone turnover markers, 137 osteoclast phenotype, 75
in bone destruction, 160161 6-Thioguanine, for treating p65/p50 nuclear factor-kappa B
in osteoclast differentiation, hypercalcemia of expression, in
34 malignancy and breast osteoclastogenesis, 74
in osteoclastogenesis, 8 cancer metastases, 166 Pax5, for regulation of
Stromal-derived factor (SDF-1), Thromboembolism, venous osteoclast development, 93
receptor CXCR-4 for, in meta-analysis of four raloxifene PU.1
tumors, 159160 trials, 146 interaction with the
Stromelysin (matrix as a risk of raloxifene therapy, microphthalmia
metalloprotease 3), 27 140 transcription factor, 13
Structure Thymoma, ectopic parathyroid null allele. activation of
and bone resorption, 5866 production in, 154 tartrate-resistant acid
of cancellous bone, conversion Thyroid, medullary cancer of, due phosphatase expression
from plates to rods, 63 to ret proto-oncogene by, 25
Syk kinase, dependence of ITAM- mutation, 112 role in B cell development,
signaling on, 98 Thyroid-stimulating hormone 92
Syncytin, stimulation of cell : cell (TSH), role in osteoclast role in macrophage
fusion by, 12 recruitment, 112 development, 56
Tiludronate RUNX2, for osteoblast
Tachyphylaxis, in calcitonin outcome of clinical trial, 134 differentiation, 8, 7980
administration for structure of, 129 SCL/tal-1, role in generation of
treating hypercalcemia, Tissue inhibitors of hematopoietic cells, 4
156 metalloproteases (TIMPs), Transforming growth factor-a
Tamoxifen, 139140 regulation of matrix (TGFa), hypercalcemic
effects of, on bone loss, 166 metalloproteases by, effects of, 155
Tartrate-resistant acid 2728 and parathyroid hormone-
phosphatase (TRAP), v TNF. See Tumor necrosis factor related protein, 152153
188 Index

Transforming growth factor-b of osteoclast precursor role in tumor necrosis factor


(TGFb) differentiation, 96 signaling, 7172
activity of, in regulating tumor relationship with RANKL Tumor necrosis factor-regulated
cell behavior in bone, 160 homology of structures, 7 genes, in skeletal cells,
effect of in osteoclastogenesis, 7476 7677
on osteoclast differentiation, role in estrogen deciency Tumor necrosis factor-related
11 bone loss, 6869 activation-induced
on matrix metalloprotease 9, signal cascade initiated by, cytokine (TRANCE), 67
28 7173 Tumors
on parathyroid hormone- Tumor necrosis factor-a (TNFa) due to overexpression of c-fos,
related protein in renal effect of 5
and squamous cell on cathepsin L levels, in solid, metastasis to bone,
carcinoma, 153 osteoclasts, 2627 157166
on tumor cell secretion of on hypercalcemia due to Tyrosine kinase-binding protein,
parathyroid hormone- parathyroid-related DAP12, loss-of-function
regulating protein, protein, 152153 mutation in Nasu-Hakola
162163 hypercalcemia related to, 155 disease, 116
Transforming growth factor-b1 production of, associated with Tyrosine kinases
(TFG-b1), activity of, in estrogen deciency of membrane, expression by
Camurati-Engelmann menopause, 67 colony-forming unit-GM,
disease, 122 stimulation of osteoclast 5
Transforming growth factor b- differentiation by, 11 ret proto-oncogene as, 112
activated kinase 1 (TAK1), Tumor necrosis factor ligand role for, 4150
activation by TRAF6, 910 family, RANKL, 611 Tyrosine phosphorylation
Transmembrane protein, receptor Tumor necrosis factor receptor- of components of cellular
activator nuclear factor- associated death domain adhesion complexes, 46
kappa B ligand as, 91 (TRADD), direction of of p130Cas adaptor protein, 43
TRAP. See Tartrate-resistant acid information ow by, reversible, and regulation of
phosphatase 7273 cellular functions, 4344
Treatment Tumor necrosis factor receptor-
with bisphosphonate, duration associated factor (TRAF) Ubiquitination, of c-Fms, 49
of, 136 as an intracellular second Ubiquitin ligase, Cbl, and cell
of hypercalcemia of messenger, 95 motility, 43
malignancy, 155157 -5 (TRAF5), deciency of, effect Uteroferrin, porcine homolog of
of malignancy, 165166 on osteoclastogenic tartrate-resistant acid
TREM-2 adaptor, mutation of, in responses, 96 phosphatase, 2425
Nasu-Hakola disease, 116 -6 (TRAF6)
TREM-2 receptor, on monocyte- deciency of, mouse models, Vacuolar proton ATPase
derived dendritic cells, 50 96 (VATPase)
Tumor-associated factors, interaction with IRAK-1, 73 acidication of the extracellular
parathyroid-related in RANK signal compartment by activity
protein modulation by, transduction, gure, 7 of, 38
152153 regulation by, of I kappa B mutation in malignant
Tumor necrosis factor (TNF) kinases and nuclear factor osteopetrosis, 117118
effect of kappa B, 7273 mutation in osteopetrosis, 36
on bone loss, vi Tumor necrosis factor receptor- van Buchem disease, mutations
on osteoblasts, 7681 associated factor proteins in, 117, 119
gene for, estrogen effect on (TRAF proteins), RANK Vanishing bone syndrome, loss of
expression of, 71 interaction with, 910 matrix metalloprotease 2
induction of vitamin D Tumor necrosis factor receptors gene in, 28
resistance by, 8081 osteoprotegerin, effect on bone Vascular endothelial growth
regulation by resorption, 95 factor (VEGF)
of osteoclast differentiation, receptor activator of nuclear increase in CXCR-4 expression
3 factor-kappa B receptor, 91 by, 159160
Index 189

in osteolytic lesions induced by effect of, on bone resorption, 14 Womens Health Initiative (WHI),
parathyroid hormone- resistance to, gure legend, 79 outcomes of estrogen on
related protein, 162 See also 1,25-Dihydroxyvitamin bone fracture risk, vii, 139
support of macrophage/ D
osteoclast differentiation Vitronectin/bronectin receptor Zinc, requirement of matrix
by, 118 avb3, of mammalian metalloproteases for, 27
v-Cbl, binding of, to Src, 4243 osteoclasts, 40 Zolendronic acid
Vertebral fracture, in metastatic bone density changes caused
breast cancer, 159 Weightlessness, effect on RANKL by, clinical study, 134135
Vitamin D expression, 8 for hypercalcemia of
anti-tumor effects of Weight loss, in terminal malignancy treatment, 156
metabolites of, 24 metastatic disease, 159 Zolendronate, structure of, 129

Potrebbero piacerti anche