Sei sulla pagina 1di 7

REVIEW

CURRENT
OPINION Control of puberty: genetics, endocrinology,
and environment
Jin-Ho Choi a,b and Han-Wook Yoo a

Purpose of review
The aim of this review is to summarize recent advances regarding the genetic components of the complex
and coordinated process of puberty, an update of the genes implicated in disorders of puberty, the
endocrinologic changes of puberty, and influences of environment in the light of our current understanding
Downloaded from https://journals.lww.com/co-endocrinology by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3ZRPSXgVoeNmk/yYblKMeKkDzR4ZD1TBUDWoDLiWFSU8= on 09/17/2018

of the mechanism of the onset of puberty.


Recent findings
The timing of puberty varies greatly in the general population among ethnic groups throughout the world,
suggesting the genetic control of puberty. Several studies on the pathological conditions of pubertal onset
provide unique information about the interactions of either the genetic susceptibility of or environmental
influences on hypothalamic control of pubertal onset. However, these findings suggested that no isolated
pathway or external factor is solely responsible for the neuroendocrine control of puberty.
Summary
Puberty is initiated by gonadotropin-releasing hormone from the hypothalamus followed by a complex
sequence of endocrine changes and is regulated by both genetic and environmental factors. New attempts
to use genetics and genomics might enhance our understanding of the spectrum of pubertal development.
Keywords
endocrine disrupting chemical, gonadotropin-releasing hormone, isolated gonadotropin-releasing hormone
deficiency, precocious puberty

INTRODUCTION pathological conditions, these variations can


Puberty is the process of transition from childhood involve different genetic susceptibilities and the
to adulthood and is an important developmental interactions of environmental factors [2]. The study
stage during which secondary sex characteristics of pathological states, such as single gene defects
appear, adolescent growth spurt occurs, reproduc- that result in central precocious puberty (CPP) and
tive capacity is achieved, and profound psychologi- isolated gonadotropin-releasing hormone (GnRH)
cal changes take place [1]. Puberty results from deficiency (IGD), have provided valuable insights
complex, coordinated neuroendocrine mechanisms into the genes that regulate GnRH activity and
involving the maturation and activation of the pathways and modulate pubertal onset and pro-
hypothalamic–pituitary–gonadal (HPG) axis [1,2]. gression, as well as the genetic basis of disorders
The wide variations among the timing and in pubertal timing. Mutations in specific genes
tempo of normal puberty among normal individuals
throughout the world are influenced by both
a
genetic and environmental factors [3–5]. The identi- Department of Pediatrics, Asan Medical Center Children’s Hospital,
fication of genes involving pubertal onset has been University of Ulsan College of Medicine, Seoul, Korea and bReproductive
Endocrine Unit of the Department of Medicine, Harvard Reproductive
difficult because the genetic control of the variance
Endocrine Sciences Center, Massachusetts General Hospital, Boston,
in pubertal timing is regulated by complex poly- Massachusetts, USA
genic traits, and interactions between genetic Correspondence to Jin-Ho Choi, MD, PhD, Department of Pediatrics,
variants and environmental exposures [4,6]. Asan Medical Center Children’s Hospital, University of Ulsan College of
Under physiological conditions, factors affect- Medicine, Seoul, Korea. Tel: +82 2 3010 3991; fax: +82 2 473 3725;
ing the genetic control of hypothalamic functions e-mail: jhc@amc.seoul.kr
are predominant in determining the individual vari- Curr Opin Endocrinol Diabetes Obes 2013, 20:62–68
ations in timing of pubertal onset. However, in DOI:10.1097/MED.0b013e32835b7ec7

www.co-endocrinology.com Volume 20  Number 1  February 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Control of puberty Choi and Yoo

neurokinin B and dynorphin A that are coexpressed


KEY POINTS in Kiss1 neurons (KNDy neurons) [11]. Hypothala-
 The timing and tempo of puberty is controlled by mic levels of Kiss1 and KISS1R mRNA increase
regulatory gene networks and the interactions of dramatically at puberty. Thus, it is clear that acti-
environmental factors. vation of KISS1R by kisspeptins plays a pivotal role
in the onset of puberty [10]. However, it is not yet
 Several genes that play important roles in the
known whether the KISS1/KISS1R system is the
development and regulation of HPG axis have been
identified as causative genes involving precocious initial trigger of puberty or whether it acts as a
puberty and isolated GnRH deficiency. downstream effector of other regulatory factors [12].
Epidemiological observations suggest that the
 Pubertal onset may be related to environmental onset of pubertal timing is highly correlated within
conditions, such as light, geographic location, nutrition,
racial/ethnic groups, within families with CDGP,
chronic diseases, frequent infectious diseases, pollution,
stressful events, interfamilial relationships, and exposure and between monozygotic compared with dizygotic
to endocrine-disrupting chemicals (EDCs). twins [3,4,7,13]. It is interesting to note that most
boys and girls destined to enter puberty late have a
family history of later pubertal development and
follow a characteristic program of linear growth
involved in IGD have been identified but how these throughout childhood, suggesting that variations
genes regulate the timing of puberty in the general in the tempo of growth and puberty may well
population or in populations with constitutional represent the unfolding of a complex mixture of
delay of growth and puberty (CDGP) remains to genetic variables [4]. In a study of 184 pairs of
be determined [7]. It can be assumed that some of monozygotic and dizygotic twin girls, breast devel-
the genes involved in the pathogenesis of delayed or opment and age at menarche showed a high degree
absent puberty may also be candidate genes for of genetic correlation, inferring that a common set
mutations that underlie precocious puberty. of genes control events in puberty [14]. These find-
ings suggest that 50–80% of the variation in puber-
tal timing is regulated by genetic factors [6].
GENETIC CONTROL OF PUBERTY IN Some candidate genes have been found to
NORMAL GENERAL POPULATION exhibit particular single nucleotide polymorphisms
Although environmental and metabolic factors are (SNPs) associated with alterations in pubertal timing
critical regulators of the HPG axis and the timing of by large-scale genome-wide association studies
puberty, genetic background plays an important
&
(GWAS) [15 ]. In Japanese women, early menarche
role in regulating variations in pubertal timing was found to be associated with the A2 polymor-
within the general population at any particular phism of CYP17 controlling androgen biosynthesis
point in time [4,7]. and thereby possibly accounting for increased
The timing and tempo of puberty is controlled serum estradiol levels [16]. However, in American
by regulatory gene networks composed of multiple girls, CYP17 alleles were not associated with early
functional modules operating in overlapping of breast development. Instead, this event was strongly
partially redundant pathways instead of strict hier- associated with the A4 allele of CYP3, an enzyme
archies [8]. Many genes are involved in the control involved in testosterone catabolism [17]. Recently,
of these cellular networks and in the control of the GWAS have identified the first loci with common
pubertal process as a whole. The KISS1/KISS1R sys- variations that were reproducibly associated with
tem is a critical component of the HPG axis and is population variations at the timing of puberty.
necessary for pubertal onset [9]. KISS1 encodes These loci were identified at 6q21 in or near LIN28B
several kisspeptins, which are the ligands that bind and at 9q31.2. Variants of at least 10 other genes are
to the G-protein coupled receptor KISS1R. Studies of also associated with early menarche [6,18–20].
several mammalian species have shown that kiss- However, the specific genetic factors that regulate
peptins stimulate the secretion of gonadotropins the variation in pubertal timing in the general
from the pituitary by stimulating the release of population or in populations with CDGP remain
GnRH from the forebrain after the activation of to be discovered.
KISS1R [10]. Kisspeptin is expressed abundantly in
the arcuate nucleus (ARC) and the anteroventral
periventricular nucleus of the forebrain [10]. Kiss1 GENETIC DEFECTS IN PRECOCIOUS
neurons in the ARC are plausible generators of PUBERTY
GnRH pulses through a system of pulsatile kisspep- Only a few molecular defects including mutations
tin release shaped by the coordinated action of and SNPs of KISS1, KISS1R, NPY, and LIN28B, have

1752-296X ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-endocrinology.com 63

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Growth and development

&&
been identified in patients with CPP [21 ,22]. Fam- to cause autosomal recessive hypogonadotropic
ilial CPP is characterized by an autosomal dominant hypogonadism [28]. However, extensive analyses
mode of transmission with incomplete penetrance suggested that genetic variation in GNRH1 or
affecting mostly girls [23]. A heterozygous missense GNRHR is not a common cause of delayed puberty
mutation in KISS1R, which leads to prolonged in the general population [29]. The KISS1/KISS1R
activation of kisspeptin-responsive intracellular signaling complex was demonstrated as an import-
signaling pathways, has been reported in a girl with ant regulator of the HPG axis in 2003 by two inde-
idiopathic CPP [24]. Heterozygous mutations in pendent groups reporting deletions and inactivating
KISS1, the gene encoding the ligand for KISS1R, have mutations of KISS1R in patients with hypogonado-
also been described as a cause of CPP [25]. Further tropic hypogonadism [9,30]. Recently, mutations in
molecular investigation of familial cases with idio- CHD7, a gene responsible for CHARGE (Coloboma,
pathic CPP might enable us to verify the hypothesis Heart defects, choanal Atresia, Retardation of
regarding the genes involved in CPP and might growth and development, Gonadal defects, and
contribute to the understanding of the pathogenesis Ear/hearing abnormalities, MIM 214800) syndrome,
of CPP. which shares some developmental features with
Single gene defects in peripheral precocious pub- Kallmann syndrome, were identified in patients
erty have been recognized rarely. One condition is with both nIHH and Kallmann syndrome [31].
familial male-limited precocious puberty or testo- IGD accompanied by obesity can result from defects
toxicosis, which is caused by activating mutations in LEP, LEPR, and PC1, which highlights the import-
in LHR. The McCune–Albright syndrome is also ance of nutrition in modulating the HPG axis.
characterized by a peripheral precocious puberty People with a homozygous mutation in LEP or LEPR
associated with postzygotic activating mutations in not only have morbid obesity but also a striking
GNAS, leading to a group of cells with constitutively delay in puberty owing to IGD. However, recent
active adenylate cyclase [26]. association studies have found no substantial associ-
ation between common polymorphisms in LEP or
LEPR and CDGP or age at menarche in the general
GENETIC DEFECTS IN ISOLATED population [29]. Other causes of hypogonadotropic
GONADOTROPIN-RELEASING HORMONE hypogonadism include mutations in genes that are
DEFICIENCY critical to HPG development, including DAX1, SF1,
The study of IGD has led to the identification of and several pituitary transcription factors (HESX1,
several genes that play critical roles in the develop- LHX3, and PROP1) [32].
ment and regulation of the HPG axis and olfactory IGD can result from the combination of
structures, but the specific genetic factors that mutations in different genes [33,34]. One of the
regulate variation in pubertal timing in the general patients in this series was heterozygous for both a
population are just emerging [4,6]. The genes PROKR2 mutation and a KAL1 mutation, suggesting
known to cause IGD are illustrated in Table 1. The a possible digenic mode of inheritance [33]. A
genes implicated in the etiology of IGD vary from heterozygous deletion in NELF and FGFR1 has been
those that are purely neurodevelopmental genes reported as a component of digenic Kallmann syn-
that impair GnRH development and migration drome, but is not clear whether mutations in NELF
(KAL1 and NELF) to those that are assumed to be alone lead to Kallmann syndrome [33]. Cases of
purely neuroendocrine genes (GNRH1, GNRHR, reversible hypogonadotropic hypogonadism have
KISS1, KISS1R, TAC3, and TACR3). Certain genes also been reported [35], further blurring the distinc-
have now been implicated in both developmental tion between IGD and CDGP. These reports likely
and neuroendocrine function (FGF8, FGFR1, represent only the beginning of our understanding
&&
PROK2, PROKR2, and CHD7) [27 ]. Mutations of the roles that multigenic inheritance and modi-
involving neurodevelopmental pathways cause fier genes play in the phenotypic variability within
Kallmann syndrome with anosmia/hyposmia, IGD.
although defects in neuroendocrine pathways lead
to normosmic idiopathic hypogonadotropic hypo-
gonadism (nIHH) [27 ].
&&
ENDOCRINOLOGIC CHANGES DURING
The first identified gene involved in Kallmann PUBERTY
syndrome is KAL1, which encodes anosmin, a Gonadotropin levels are low at birth. A transient
cellular matrix protein required for the normal increase during the first months of life has been
migration of olfactory processes and GnRH neurons termed ‘minipuberty’. Thereafter, levels return to
from their common site of origin, the olfactory a very low, often undetectable range during the
placode [4]. GNRHR was the first gene discovered prepubertal phase as a result of intrinsic restraint,

64 www.co-endocrinology.com Volume 20  Number 1  February 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Control of puberty Choi and Yoo

Table 1. Genes involved in hypogonadotropic hypogonadism

Genes Location Structure Inheritance Phenotypes

KAL1 Xp22.3 14 exons XR KS, unilateral renal agenesis, bimanual synkinesia,


high arched palate
FGFR1 (KAL2) 8p11.2-p11.1 18 exons AD KS, cleft lip/palate, skeletal anomalies (hand and foot),
external ear hypoplasia, dental agenesis
PROKR2 (KAL3) 20p13 2 exons AR, AD KS, nIHH, AHH
PROK2 (KAL4) 3p21.1 4 exons AR, (AD?) KS
CHD7 (KAL5) 8q12.1 38 exons AD<Sporadic CHARGE syndrome, KS, nIHH
FGF8 (KAL6) 10q24 6 exons AD nIHH, AHH, cleft lip/palate, skeletal anomalies (hand and foot),
external ear hypoplasia, dental agenesis
GNRH1 8p21-p11.2 3 exons AR nIHH
GNRHR 4q21.2 3 exons AR nIHH
KISS1 1q32 3 exons AR nIHH
KISS1R 19p13.3 5 exons AR, AD nIHH (AR), Precocious puberty (AD)
TAC3 12q13-q21 7 exons AR nIHH, microphallus, cryptorchidism, reversal of GnRH deficiency
TACR3 4q25 5 exons AR nIHH, microphallus, cryptorchidism, reversal of GnRH deficiency
NELF 9q34.3 15 exons Digenic KS
NR0B1/DAX1 Xp21.3-p21.2 2 exons XR X-linked adrenal hypoplasia congenita, hypogonadotropic
hypogonadism
LEP 7q31.3 3 exons AR nIHH, severe obesity
LEPR 1p31 20 exons AR nIHH, severe obesity
LHb 19q13.32 3 exons AR, (AD) Isolated LH deficiency
FSHb 11p13 3 exons AR Isolated FSH deficiency
PHF6 Xq26.3 11 exons XL Borjeson–Forssman–Lehmann syndrome

AR, autosomal recessive; AD, autosomal dominant; CHARGE, Coloboma, Heart defects, choanal Atresia, Retardation of growth and development, Gonadal
defects, and Ear/hearing abnormalities, MIM 214800; FSH, follicle-stimulating hormone; KS, Kallmann syndrome; LH, luteinizing hormone; nIHH, normosmic
idiopathic hypogonadotropic hypogonadism; XL, X-linked; XR, X-linked recessive.

‘the juvenile pause’ [4]. The onset of puberty is glial component of the system is mostly facilitatory,
characterized by the increase in amplitude of GnRH and it is provided by growth factors and small
pulses and, consequently, of luteinizing hormone diffusible molecules that directly or indirectly
and follicle-stimulating hormone (FSH) pulses, stimulate GnRH secretion [39]. Kisspeptin neurons
initially at night, and then as puberty advances, of the ARC produce two additional peptides: neuro-
throughout a 24-h period [4,36]. kinin B (TAC3), which is encoded by a gene recently
The timing of puberty can be influenced by shown to be required for puberty to occur [40], and
signals involving neurotransmitters and neuro- dynorphin, an opioid peptide that inhibits GnRH
peptides that originate in the hypothalamus, in secretion [41]. These observations indicate that the
addition to peripheral or gonadal signals [3]. The excitatory transsynaptic regulation of GnRH
pubertal activation of GnRH-release requires coor- secretion is provided by neurons that use glutamate,
dinated changes in excitatory and inhibitory inputs kisspeptin, and perhaps neurokinin B for transsy-
to GnRH-secreting neurons. These inputs are pro- naptic communication [39] (Fig. 1).
vided by both transsynaptic and glia-to-neuron Leptin, and insulin-like growth factor-1 (IGF-1)
communication pathways [37]. The transsynaptic have been shown to be involved in the control of
changes consist of a coordinated increase in excit- GnRH secretion, but their role in the timing of pub-
atory inputs and a reduction in inhibitory influ- erty remains controversial [3]. The pubertal growth
ences. The EAP1 protein localizes to neuronal spurt results mainly from the synergistic effect of the
nuclei and is able to transregulate the promoter gonadal sex steroids, growth hormone, and IGF-1, all
activity of genes involved in the transsynaptic con- of which show a significant increase in serum levels
trol of GnRH secretion [38]. GABAergic and opiater- during this period [5]. Both androgens and estrogens
gic neurons provide transsynaptic inhibitory also appear to have a direct growth-modulating effect
control to the system, but GABA neurons also exert on the growth plate by stimulating the local pro-
direct excitatory effects on GnRH neurons [8]. The duction of IGF-1 and other growth factors [5].

1752-296X ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-endocrinology.com 65

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Growth and development

KISS1 neuron AVPN ARC

Mediobasal hypothalamus
GNRH1/KISS1R
GABA
FGF8/FGFR1 (KAL2) Kisspeptin
TAC3/TACR3 Glutamate
PROK2/PROKR2
KAL1/NELF
LEP/LEPR
PCSK1/WDR11
SF1DAX1
Other factors GnRH
IGF-1
Leptin
FGF
Pituitary gland TGFβ
GNRHR
PROP1/HESX1
LHX3/LHX4 Inhibin B
SOX2/SOX3 LH/FSH
SF1/DAX1

Gonads
LHβ/FSHββ
LHR/FSHR Excitatory
Sex steroids
SF1/DAX1 Inhibitory

FIGURE 1. Overview of the hypothalamic–pituitary–gonadal (HPG) axis. Kisspeptin released by neurons in the anteroventral
periventricular nucleus (AVPN) and arcuate nucleus (ARC) stimulates GnRH release, which induces the release of LH and FSH.
The gonads respond to gonadotropins by secreting sex steroids, and sex steroids differentially regulate the expression of Kiss1
mRNA in different nuclei within the forebrain by inhibiting Kiss1 expression in the ARC and inducing its expression in the
AVPN. Mutations in several genes involved in each site have been shown to cause isolated GnRH deficiency in humans.

ENVIRONMENTAL INFLUENCES ON relatively early puberty [46]. A high calorie diet


PUBERTY and accelerated body weight gain during juvenile
It has been well established that pubertal onset period significantly advance the onset of puberty
may be related to environmental conditions, such and serum leptin levels were elevated before early
&
as light, geographic location [3], nutrition, chronic menarche [47 ]. Leptin is considered the link
diseases, frequent infectious diseases, pollution [5], between adipose tissue and growth. Leptin, which
stressful events, interfamilial relationships [42], and is produced and secreted by white adipose tissue, has
exposure to endocrine-disrupting chemicals (EDCs) a crucial role in pubertal onset, the pubertal process,
[43]. Environmental factors may also play a prom- and pubertal growth [7]. The increased levels of
inent role in situations. There are secular trends in leptin just before the onset of puberty might induce
the advancement in the onset of breast develop- both augmented proliferation and differentiation of
ment seen in some industrialized countries and an chondrocytes in the epiphyseal growth plate [48].
increased incidence of precocious puberty in chil- However, leptin itself is not considered a strong
dren migrating to such countries [3]. metabolic trigger for the onset of puberty, although
Environmental effects on the mechanism of it acts as a permissive signal for the onset of puberty
pubertal onset may start during intrauterine life. [49].
Earlier onset of menarche, more rapid progression Migration may interrupt exposure to endocrine
of puberty, and increased prevalence of polycystic disrupters, and precocious puberty might then
ovary disease and premature adrenarche have been result from withdrawal of their negative feedback
described in girls with low birth weight or intra- effect and/or from accelerated hypothalamic matu-
uterine growth retardation [44]. ration. The high incidence of precocious puberty in
Nutrition is likely to play a key role in the timing foreign children migrating to Belgium and the
of puberty and can explain, at least partially, the detection in their plasma of long-lasting 1,1,1-tri-
downward secular trend in the timing of puberty [3]. chloro-2,2-bis(4-chlorophenyl) ethane (DDT) resi-
In a longitudinal assessment of a birth cohort of 156 due suggests the potential role of environmental
girls, it was noted that girls who were light and long EDCs in the early onset of puberty [2]. DDT is able
at birth and had greater circulating fat mass in mid- to promote hypothalamic maturation, exerting an
childhood had earlier ages of menarche [45]. Rapid inhibitory effect at the pituitary level that is most
weight gain early in infancy has been linked to effective in prepubertal individuals [50]. In addition,

66 www.co-endocrinology.com Volume 20  Number 1  February 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Control of puberty Choi and Yoo

11. Navarro VM. New insights into the control of pulsatile GnRH release: the role
early pubertal development and an increased inci- of kiss1/neurokinin B neurons. Front Endocrinol (Lausanne) 2012; 3:48.
dence of precocious puberty have been noticed in 12. Roth CL, Mastronardi C, Lomniczi A, et al. Expression of a tumor-related gene
network increases in the mammalian hypothalamus at the time of female
children migrating to a number of Western Euro- puberty. Endocrinology 2007; 148:5147–5161.
pean countries [3]. These differences in the timing of 13. Palmert MR, Hirschhorn JN. Genetic approaches to stature, pubertal timing,
and other complex traits. Mol Genet Metab 2003; 80:1–10.
puberty seem to result from interactions of environ- 14. van den Berg SM, Setiawan A, Bartels M, et al. Individual differences in
mental factors irrespective of genetic susceptibility, puberty onset in girls: Bayesian estimation of heritabilities and genetic
correlations. Behav Genet 2006; 36:261–270.
as children migrating from several continents and 15. Dvornyk V, Waqar ul H. Genetics of age at menarche: a systematic review.
belonging to several ethnic groups are involved [3]. & Hum Reprod Update 2012; 18:198–210.
This study summarized several large-scale GWAS related to candidate genes
associated with age at menarche.
16. Gorai I, Tanaka K, Inada M, et al. Estrogen-metabolizing gene polymorphisms,
CONCLUSION but not estrogen receptor-alpha gene polymorphisms, are associated with the
onset of menarche in healthy postmenopausal Japanese women. J Clin
There is a great variation in the timing and course of Endocrinol Metab 2003; 88:799–803.
17. Kadlubar FF, Berkowitz GS, Delongchamp RR, et al. The CYP3A41B variant
puberty in the general population, which is influ- is related to the onset of puberty, a known risk factor for the development of
enced by familial, ethnic, and sex patterns of signals breast cancer. Cancer Epidemiol Biomarkers Prev 2003; 12:327–331.
18. Sulem P, Gudbjartsson DF, Rafnar T, et al. Genome-wide association study
or signal receptors in the hypothalamus. Puberty is identifies sequence variants on 6q21 associated with age at menarche. Nat
likely regulated by an intricate and coordinated Genet 2009; 41:734–738.
19. Ong KK, Elks CE, Li S, et al. Genetic variation in LIN28B is associated with the
gene network controlling several physiological timing of puberty. Nat Genet 2009; 41:729–733.
pathways in humans. However, the exact primary 20. He C, Kraft P, Chen C, et al. Genome-wide association studies identify loci
associated with age at menarche and age at natural menopause. Nat Genet
mechanisms that underlie activation of HPG axis 2009; 41:724–728.
maturation and regulate the onset of puberty 21. Teles MG, Silveira LF, Tusset C, Latronico AC. New genetic factors impli-
cated in human GnRH-dependent precocious puberty: the role of kisspeptin
remain to be discovered. Further investigation of &&

system. Mol Cell Endocrinol 2011; 346:84–90.


the maturation of GnRH secretion and pituitary This review summarized the molecular defects that are implicated in GnRH-
dependent precocious puberty.
responsiveness is vital to understanding the mech- 22. Tommiska J, Sorensen K, Aksglaede L, et al. LIN28B, LIN28A, KISS1, and
anisms of the wide spectrum of pubertal develop- KISS1R in idiopathic central precocious puberty. BMC Res Notes 2011;
4:363.
ment. 23. de Vries L, Kauschansky A, Shohat M, Phillip M. Familial central precocious
puberty suggests autosomal dominant inheritance. J Clin Endocrinol Metab
2004; 89:1794–1800.
Acknowledgements 24. Teles MG, Bianco SD, Brito VN, et al. A GPR54-activating mutation in a
None. patient with central precocious puberty. N Engl J Med 2008; 358:709–
715.
25. Silveira LG, Noel SD, Silveira-Neto AP, et al. Mutations of the KISS1 gene in
Conflicts of interest disorders of puberty. J Clin Endocrinol Metab 2010; 95:2276–2280.
26. Dumitrescu CE, Collins MT. McCune-Albright syndrome. Orphanet J Rare Dis
There are no conflicts of interest. 2008; 3:12.
27. Balasubramanian R, Crowley WF Jr. Isolated GnRH deficiency: a disease
&& model serving as a unique prism into the systems biology of the GnRH
neuronal network. Mol Cell Endocrinol 2011; 346:4–12.
This review describes developmental process of the reproductive axis and genetic
REFERENCES AND RECOMMENDED architecture of GnRH deficiency.
READING 28. Layman LC, Cohen DP, Jin M, et al. Mutations in gonadotropin-releasing
Papers of particular interest, published within the annual period of review, have hormone receptor gene cause hypogonadotropic hypogonadism. Nat Genet
been highlighted as: 1998; 18:14–15.
& of special interest 29. Gajdos ZK, Butler JL, Henderson KD, et al. Association studies of common
&& of outstanding interest variants in 10 hypogonadotropic hypogonadism genes with age at menarche.
Additional references related to this topic can also be found in the Current J Clin Endocrinol Metab 2008; 93:4290–4298.
World Literature section in this issue (pp. 78–79). 30. de Roux N, Genin E, Carel JC, et al. Hypogonadotropic hypogonadism due to
loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl
1. Foster DL, Jackson LM, Padmanabhan V. Programming of GnRH feedback Acad Sci U S A 2003; 100:10972–10976.
controls timing puberty and adult reproductive activity. Mol Cell Endocrinol 31. Kim HG, Kurth I, Lan F, et al. Mutations in CHD7, encoding a chromatin-
2006; 254–255:109–119. remodeling protein, cause idiopathic hypogonadotropic hypogonadism and
2. Parent AS, Rasier G, Gerard A, et al. Early onset of puberty: tracking genetic Kallmann syndrome. Am J Hum Genet 2008; 83:511–519.
and environmental factors. Horm Res 2005; 64 (Suppl 2):41–47. 32. Silveira LF, MacColl GS, Bouloux PM. Hypogonadotropic hypogonadism.
3. Parent AS, Teilmann G, Juul A, et al. The timing of normal puberty and the age Semin Reprod Med 2002; 20:327–338.
limits of sexual precocity: variations around the world, secular trends, and 33. Pitteloud N, Quinton R, Pearce S, et al. Digenic mutations account for variable
changes after migration. Endocr Rev 2003; 24:668–693. phenotypes in idiopathic hypogonadotropic hypogonadism. J Clin Invest
4. Palmert MR, Boepple PA. Variation in the timing of puberty: clinical spectrum 2007; 117:457–463.
and genetic investigation. J Clin Endocrinol Metab 2001; 86:2364–2368. 34. Sykiotis GP, Plummer L, Hughes VA, et al. Oligogenic basis of isolated
5. Phillip M, Lazar L. Precocious puberty: growth and genetics. Horm Res 2005; gonadotropin-releasing hormone deficiency. Proc Natl Acad Sci U S A 2010;
64 (Suppl 2):56–61. 107:15140–15144.
6. Gajdos ZK, Henderson KD, Hirschhorn JN, Palmert MR. Genetic determi- 35. Raivio T, Falardeau J, Dwyer A, et al. Reversal of idiopathic hypogonadotropic
nants of pubertal timing in the general population. Mol Cell Endocrinol 2010; hypogonadism. N Engl J Med 2007; 357:863–873.
324:21–29. 36. Grumbach MM. The neuroendocrinology of human puberty revisited. Horm
7. Kaminski BA, Palmert MR. Genetic control of pubertal timing. Curr Opin Res 2002; 57 (Suppl 2):2–14.
Pediatr 2008; 20:458–464. 37. Ojeda SR, Roth C, Mungenast A, et al. Neuroendocrine mechanisms con-
8. Ojeda SR, Dubay C, Lomniczi A, et al. Gene networks and the neuroendocrine trolling female puberty: new approaches, new concepts. Int J Androl 2006;
regulation of puberty. Mol Cell Endocrinol 2010; 324:3–11. 29:256–263.
9. Seminara SB, Messager S, Chatzidaki EE, et al. The GPR54 gene as a 38. Heger S, Mastronardi C, Dissen GA, et al. Enhanced at puberty 1 (EAP1) is a
regulator of puberty. N Engl J Med 2003; 349:1614–1627. new transcriptional regulator of the female neuroendocrine reproductive axis.
10. Dungan HM, Clifton DK, Steiner RA. Minireview: kisspeptin neurons as central J Clin Invest 2007; 117:2145–2154.
processors in the regulation of gonadotropin-releasing hormone secretion. 39. Ojeda SR, Lomniczi A, Loche A, et al. The transcriptional control of female
Endocrinology 2006; 147:1154–1158. puberty. Brain Res 2010; 1364:164–174.

1752-296X ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-endocrinology.com 67

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Growth and development

40. Topaloglu AK, Reimann F, Guclu M, et al. TAC3 and TACR3 mutations in 46. Dunger DB, Ahmed ML, Ong KK. Early and late weight gain and the timing of
familial hypogonadotropic hypogonadism reveal a key role for Neurokinin B puberty. Mol Cell Endocrinol 2006; 254–255:140–145.
in the central control of reproduction. Nat Genet 2009; 41:354–358. 47. Terasawa E, Kurian JR, Keen KL, et al. Body weight impact on puberty: effects
41. Navarro VM, Gottsch ML, Chavkin C, et al. Regulation of gonadotropin- & of high-calorie diet on puberty onset in female rhesus monkeys. Endocrinology
releasing hormone secretion by kisspeptin/dynorphin/neurokinin B 2012; 153:1696–1705.
neurons in the arcuate nucleus of the mouse. J Neurosci 2009; This study demonstrated the importance of juvenile feeding behaviors as an
29:11859–11866. intervention to reduce the prevalence of precocious development.
42. Matchock RL, Susman EJ. Family composition and menarcheal age: antiin- 48. Maor G, Rochwerger M, Segev Y, Phillip M. Leptin acts as a growth factor on
breeding strategies. Am J Hum Biol 2006; 18:481–491. the chondrocytes of skeletal growth centers. J Bone Miner Res 2002;
43. Den Hond E, Schoeters G. Endocrine disrupters and human puberty. Int J 17:1034–1043.
Androl 2006; 29:264–271. 49. Cheung CC, Thornton JE, Nurani SD, et al. A reassessment of leptin’s role in
44. Ibanez L, Ferrer A, Marcos MV, et al. Early puberty: rapid progression and triggering the onset of puberty in the rat and mouse. Neuroendocrinology
reduced final height in girls with low birth weight. Pediatrics 2000; 106:E72. 2001; 74:12–21.
45. Tam CS, de Zegher F, Garnett SP, et al. Opposing influences of prenatal and 50. Krstevska-Konstantinova M, Charlier C, Craen M, et al. Sexual precocity after
postnatal growth on the timing of menarche. J Clin Endocrinol Metab 2006; immigration from developing countries to Belgium: evidence of previous
91:4369–4373. exposure to organochlorine pesticides. Hum Reprod 2001; 16:1020–1026.

68 www.co-endocrinology.com Volume 20  Number 1  February 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Potrebbero piacerti anche