Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

Available online at www.sciencedirect.com

Metabolism
www.metabolismjournal.com

Reproductive Physiology

The hypothalamus-pituitary-gonad axis:


Tales of mice and men

Athina Kaprara a,⁎, Ilpo T. Huhtaniemi b


a
Unit of Reproductive Endocrinology, Medical School, Aristotle University of Thessaloniki, Greece
b
Imperial College, London, UK

A R T I C LE I N FO AB S T R A C T

Article history: Reproduction is controlled by the hypothalamic-pituitary-gonadal (HPG) axis.


Received 17 July 2017 Gonadotropin-releasing hormone (GnRH) neurons play a central role in this axis through
Accepted 29 November 2017 production of GnRH, which binds to a membrane receptor on pituitary gonadotrophs and
stimulates the biosynthesis and secretion of follicle-stimulating hormone (FSH) and
Keywords: luteinizing hormone (LH). Multiple factors affect GnRH neuron migration, GnRH gene
GnRH expression, GnRH pulse generator, GnRH secretion, GnRH receptor expression, and
Hypothalamus–pituitary–gonadal gonadotropin synthesis and release. Among them anosmin is involved in the guidance of
axis the GnRH neuron migration, and a loss-of-function mutation in its gene leads to a failure of
GnRH receptors their migration from the olfactory placode to the hypothalamus, with consequent anosmic
GnRH pulse generator hypogonadotropic hypogonadism (Kallmann syndrome). There are also cases of
hypogonadotropic hypogonadim with normal sense of smell, due to mutations of other
genes. Another protein, kisspeptin plays a crucial role in the regulation of GnRH pulse
generator and the pubertal development. GnRH is the main hypothalamic regulator of the
release of gonadotropins. Finally, FSH and LH are the essential hormonal regulators of
testicular functions, acting through their receptors in Sertoli and Leydig cells, respectively.
The main features of the male HPG axis will be described in this review.
© 2017 Elsevier Inc. All rights reserved.

Abbreviations: HPG, hypothalamic-pituitary-gonadal; GnRH, gonadotropin-releasing hormone; FSH, follicle-stimulating hormone; LH,
luteinizing hormone; CNS, central nervous system; HH, hypogonadotropic hypogonadism; PSA-NCAM, polysialic acid form of neural
adhesion molecule; NELF, nasal embryonic LHRH factor; GABA, gamma-aminobutyric acid; FGF8, fibroblast growth factor 8; FGFR1,
fibroblast growth factor receptor 1; PROK2, prokineticin 2; HGF, hepatocyte growth factor; KS, Kallmann syndrome; aa, amino acid;
DREAM, downstream regulatory element antagonist modulator; Dlx2, distalless homeobox 2; PKC, protein kinase C; RARa, retinoic acid
receptor-a; RXRa, retinoid X receptor-a; ER, estrogen receptor; KiSS1R, kisspeptin receptors; DHEAS, dehydroepiandrosterone sulfate; IGF-
1, insulin-like growth factor 1; MAPK, mitogen-activated protein kinase; Egr-1, early growth response-1; NPY, neuropeptide Y; GPR54, G-
protein coupled receptor 54; IP3, inositol-trisphosphate; ERK, extracellular signal–regulated kinase; JNK, c-Jun N-terminal kinase; AC,
adenylyl cyclase; cAMP, cyclic adenosine-3′:5′-monophosphate; PKA, protein kinase A; CREB, cAMP response element-binding protein;
EGFR, epidermal growth factor receptor; GPCR, G protein-coupled receptor; GnIH, gonadotropin-inhibitory hormone; LRR, leucine-rich
repeats; hCG, human chorionic gonadotropin; DSD, disorder of sexual development; TGF-β, transforming growth factor-β.
⁎ Corresponding author at: Unit of Reproductive Endocrinology, Medical School, Aristotle University of Thessaloniki, Papageorgiou
General Hospital, Ring Road, Municipality of Pavlos Meloas Area of N. Efkarpia, Thessaloniki 56403, Greece.
E-mail addresses: athkap@yahoo.gr (A. Kaprara), ilpo.huhtaniemi@imperial.ac.uk (I.T. Huhtaniemi).

https://doi.org/10.1016/j.metabol.2017.11.018
0026-0495/© 2017 Elsevier Inc. All rights reserved.
4 ME TA BOL I SM C LI N I CA L A ND E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
2. Embryonic Development and Anatomical Distribution of GnRH Neurons . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3. GnRH and GnRH Genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.1. Regulation of GnRH Gene Expression. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2. GnRH Pulse Generator . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. Kisspeptin, Leptin, Neurokinin B and Dynrophin Regulating GnRH Production . . . . . . . . . . . . . . . . . . . . . . . 7
4.1. GnRH Receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.2. Gonadotropin-Inhibitory Hormone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
4.3. Gonadotropins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
. Conflicts
Conflicts of Interest
of Interest. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12

1. Introduction neurons to migrate properly results in hypogonadotropic


hypogonadism (HH) and disrupts sexual maturation in mice
The hypothalamic–pituitary–gonadal (HPG) axis is the humoral and men [8].
component of an intercommunicating neural and endocrine Multiple factors play a critical role in the guidance of the GnRH
system that functions in the regulation of fertility. In 1937, a neuron migration process. Among them, there are adhesion
medical student at Cambridge, Geoffrey Harris, was the first one molecules, such as anosmin, polysialic acid form of neural
to provide evidence that the pituitary–gonadal axis is under adhesion molecule (PSA-NCAM) [9] and cell surface
control of the central nervous system (CNS) [1]. The isolation of glycoconjugate [10]. Furthermore, gamma-aminobutyric acid
a hypothalamic substance that, through the hypophysial portal (GABA) [11], cholecystokinin [12], fibroblast growth factor 8
circulation, regulates the synthesis and secretion of gonado- (FGF8) [13], prokineticin 2 (PROK2) [14–16] and transcription
tropins was attained later by two different groups, of Andrew factors, such as Ebf2 modulate GnRH neuronal migration [17].
Schally and Roger Guillemin, from porcine and ovine brain, Moreover, there are guidance cues, such as ephrins [18] and nasal
respectively. The active molecule was a decapeptide, now embryonic LHRH factor (NELF) [19]. There are factors that guide
referred to as gonadotropin-releasing hormone (GnRH). Two GnRH neurons toward the forebrain and factors that guide GnRH
years earlier, in 1969, the group of Ernest Knobil had provided neurons through the cribriform plate to the hypothalamus. The
evidence for an episodic manner of luteinizing hormone (LH) first category includes semaphorins, plexins [8] and reelin [20].
secretion, something that was empirically demonstrated in The second category includes hepatocyte growth factor (HGF)
ovariectomized ewes by Clarke and Cummins in 1982 [2,3]. The [21], Axl, Tyro3, and chemokine attractants [8].
purpose of this review is to review the role of GnRH in Anosmin is the product of KAL1 gene, which is located on the
regulation of the HPG axis in the male. Xp22.3 region; it was cloned in 1991 [22]. It consists of 680 amino
acids and has a molecular weight of 85–100 kDa [23]. It is
expressed extracellularly with high-affinity binding to cell
2. Embryonic Development and Anatomical membrane heparin sulfate proteoglycans, and its best-
Distribution of GnRH Neurons characterized mechanism of action is its interaction with
fibroblast growth factor receptor 1 (FGFR1) [24]. Anosmin is
GnRH neurons comprise a unique neuronal population that involved in normal GnRH migration and a failure of this
originates outside the CNS. In mice, during development, GnRH migration leads to Kallmann syndrome (KS), which is a combi-
neurons migrate from the medial olfactory placode of the nation of HH and deficient sensation of smell [25]. Analysis of
developing nasal cavity across the nasal septum, together with patients with KS showed loss-of-function mutation in KAL1
vomeronasal axons, and enter into the forebrain with the (frameshifts, deletions and premature terminations) [26].
nervus terminalis, arching into the septal-preoptic area and The NELF gene, located on chromosome 9q34.3, encodes
hypothalamus. The exact final location differs among species the nasal embryonic LHRH factor. It is detected in the
[4,5]. From the hypothalamus, processes are extended to the developing olfactory and GnRH migratory pathway. NELF
median eminence, and GnRH is released into capillaries of the mutations can lead to both KS and normosmic HH [19].
hypophysial portal system in order to reach the anterior The FGF8 gene, located on chromosome 10q24, encodes
pituitary gland and to modulate its gonadotropin synthesis FGF8. The FGFR1 gene, located on 8p11.22-p11.23, encodes
and secretion. A typical GnRH neuron in the adult hypothala- FGFR1. FGF8 and its receptor FGFR1 affect the migration of
mus has two dendritic projections which extend a distance of GnRH neurons. FGF8 and FGFR1 mutations can lead to both KS
2–3 mm from their cell body [6]. In humans, there are 1000–1500 and normosmic HH. Moreover, FGF8 and FGFR1 loss-of-
GnRH neurons and their co-location with multiple central function mutations can cause cleft lip or palate, whereas
regulators allows the interaction between GnRH and other gain-of-function mutations can lead to craniosynostosis. The
neuroendocrine and metabolic inputs [7]. Failure of GnRH FGF17 gene, located on chromosome 8p2.3, encodes FGF17
ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7 5

and its mutations have been described in patients with KS Table 1 – Genes, encoded proteins and phenotypes
and normosmic HH [13]. associated to hypogonadotropic hypogonadism.
The PROK2 gene, located on chromosome 3p21.1, encodes Gene Encoded protein Phenotype
prokineticin-2. The PROK2R2 gene, located on 20p13, encodes
AXL Member of the Tyro3-Axl-Mer (TAM) KS or nHH
prokineticin receptor. PROK2 and its receptor PROK2R2 affect
receptor tyrosine kinase subfamily
GnRH migration. PROK2 and PROKR2 mutations can lead to CCDC141 KS
both KS and normosmic HH. Moreover, PROK2 and PROK2R2 CHD7 Chromodomain helicase DNA binding KS or nHH
loss-of-function mutations can cause obesity, epilepsy, sleep protein 7
disorders and synkinesia [14,15]. DMXL2 nHH
The SEMA3A gene, located on chromosome 7q21.11, DUSP6 Member of the dual specificity protein KS or nHH
phosphatase subfamily
encodes semaphorine 3A. It plays a role in the development
FEZF1 Transcriptional repressor that belongs to KS
of the olfactory system and SEMA3A loss-of-function is the zinc finger double domain protein
involved in KS [27]. family
KS represents approximately 60% of the cases of HH, FGF8 Fibroblast growth factor 8 KS or nHH
whereas the rest 40% are associated with a normal sense of FGF17 Fibroblast growth factor 17 KS or nHH
smell. Variants of > 25 genes are associated with about half of FGFR1 Fibroblast growth factor receptor KS or nHH
FLRT3 Member of the fibronectinleucine rich KS
all cases of HH, whereas the cause of the rest is still unknown.
transmembrane protein family
HH can be inherited in an X-linked, autosomal dominant, or
GNRH1 Preproprotein that is proteolytically nHH
autosomal recessive manner [27–29]. Variants in genes listed processed to generate a peptide that is a
in Table 1 are associated with HH. member of the gonadotropin-releasing
hormone family of peptides
GNRHR Receptor for type 1 gonadotropin- nHH
releasing hormone
3. GnRH and GnRH Genes
HESX1 Conserved homeobox protein that is a KS
transcriptional repressor in the
Mammalian GnRH (termed GnRH-I) is a 10-amino acid neuro-
developing forebrain and pituitary gland
peptide (pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-GlyNH2) that HS6ST1 Member of the heparan sulfate KS or nHH
was first isolated from porcine hypothalamus. It plays a key biosynthetic enzyme family
role in the function of reproductive axis. It is produced by IL17RD Interleukin-17 receptor KS
hypothalamic neurosecretory cells (GnRH neurons), and KAL1 Anosmin-1 KS
released in a pulsatile manner into the hypothalamo- KISS1 Kisspeptin nHH
KISS1R Kisspeptin receptor nHH
hypophyseal portal circulation, subsequently reaching the
LEP Leptin nHH
anterior pituitary gland, where it stimulates LH and follicle-
LEPR Leptin receptor nHH
stimulating hormone (FSH) synthesis and release [2]. Its NELF Nasal embryonic LHRH factor KS or nHH
prohormone is synthesized as a larger 92–amino acid (aa) NR0B1 nHH
precursor protein which consists of 23-aa signal peptide, the OTUD4 nHH
functional GnRH decapeptide, an amidation⁄proteolytic pro- PCSK1 Member of the subtilisin-like nHH
cessing signal (Gly-Lys-Arg), and a 56-GnRH-associated peptide proproteinconvertase family
PNPLA6 Phospholipase that deacetylates nHH
[30]. GnRH is also found in cells outside the brain, but its role is
intracellular phosphatidylcholine to
not yet well understood. Maybe GnRH-I act as autocrine and/or produce glycerophosphocholine
paracrine regulator in extrapituitary tissues such as placenta, POLR3B RNA polymerase III subunit B nHH
immune system, gonads, and endocrine tumors [31–35]. PROK2 Prokineticin-2 KS or nHH
According to aa sequence, localization and embryonic PROKR2 Prok receptor KS or nHH
origin, numerous GnRHs have been found from vertebrates RNF216 Ring finger protein 216 nHH
SEMA3A Semaphorine 3A KS
to protochordates [7]. Most vertebrates have at least two, and
SEMA3E Semaphorin 3E KS
usually three, forms of GnRH. In humans, in addition to
SEMA7A Semaphorin 7A KS or nHH
GnRH-I a second subtype, called GnRH-II has been found. SOX10 SRY-related HMG-box transcription factor KS
GnRH-II was first identified from chicken hypothalamus SPRY4 Sprouty RTK signaling antagonist 4 KS or nHH
[36,37]. It is a decapeptide that is found in the midbrain region SRA1 Steroid receptor RNA activator 1 nHH
and serves as a neurotransmitter, rather than as a pituitary TAC3 Tachykinin 3 nHH
releasing factor. Nevertheless, it has been shown that GnRH-II TACR3 Tachykinin receptor 3 nHH
WDR11 Member of the WD repeat protein family KS or nHH
can stimulate gonadotropin release in vivo through its
binding to the type I GnRH receptor [38]. GnRH-I and GnRH-II KS = Kallmann syndrome, nHH = normosmic hypogonadotropic
are found in phylogenetically diverse species, from fish to hypogonadism.
mammals, but whereas the expression of GnRH-I is higher in
the brain, GnRH-II is expressed mostly in other organs [39,40].
A third form of GnRH, GnRH-III, has been identified in
neurons of the telencephalon in teleost fish. it is located on chromosome 8p11.2-p21, and is composed of
In mammals, two genes encoding GnRH have been four exons separated by three introns [40]. The second,
identified. The first gene GnRH1 encodes GnRH-I and it was GnRH2, encodes GnRH-II, and it is mapped to chromosome
first cloned by Seeburg and Adelman in 1984 [41]. In humans, 20p13 [37].
6 ME TA BOL I SM C LI N I CA L A ND E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

Fig. 1 – Presentation of the GnRH1 gene 5′-region in rat, mouse and human.

The 5′-flanking region of GnRH1 is highly homologous signaling pathway [40]. Studies in rats revealed that GnRH1
between human, mouse and rat (Fig. 1). gene expression is also under autoregulation, through an
In the rat GnRH1, a proximal promoter and a distal ultrashort loop feedback mechanism [47].
enhancer have been found. Both of them are important for The effect of steroids on GnRH neurons is a contentious
gene transcription. The promoter is within 173 bp upstream issue. Since Shivers et al. [48] showed a lack of estrogen
of the transcription start site and the 300 bp enhancer is receptor (ER) immunoreactivity in GnRH neurons, a direct role
located at − 1863 to −1571 bp relative to the transcription start of estradiol on the GnRH neuron has been dismissed. Τhere
site. are studies that suggest a direct effect of estrogen at the level
The previous key regions are highly conserved between rat of GnRH neurons [49–52]. Immunoreactive studies [53] have
and mouse. In the mouse GnRH1, enhancer elements are demonstrated that ERs are highly expressed in the hypothal-
located between − 5500 and −2100 bp, whereas a 400 bp region amus, not in GnRH neurons but in other neurons, afferent to
(− 2100 to − 1700 bp) is found to be a critical enhancer region GnRH neurons [54,55], such as kisspeptin neurons. The last
for the GnRH gene in mouse brain [40]. ones express ERα, while GnRH neurons do not [56]. It is likely
In humans, the promoter is located at − 551 to + 1 bp region that estrogen binds to ERα on kisspeptin neurons, and
of the GnRH1 gene. It was also found that the region between subsequently inhibits kisspeptin and GnRH release. Indeed,
− 3832 and + 8 bp gives high levels of expression of GnRH genes in gonadectomised animals and humans, kisspeptins, GnRH
in Gt1–7 neuronal cells. The − 3036 to − 2923 bp, − 2766 to − and gonadotropin levels increased, whereas sex steroid levels
2539 bp and −1775 to − 1552 bp fragments are similar to declined. Moreover, antagonism of kisspeptin receptors
sequences in the rat GnRH promoter. Moreover, the region (KiSS1R) in rodents prevented the rise of gonadotropins [57].
between − 343 and + 8 bp has marked homology to the region Ιn the rat, low (picomolar) estrogen concentrations through
between − 332 and + 96 bp of the rat GnRH [40,42,43] (Fig. 1). ERα exert negative feedback on GnRH release. On the other
hand, higher (nanomolar) estradiol concentrations acting
3.1. Regulation of GnRH Gene Expression through ERβ, have the opposite effect, and evoke a positive
regulation of GnRH secretion [58]. Although most studies have
GnRH1 gene is expressed in a pulsatile manner in the focused on the act of estradiol, it is also evident that
hypothalamus, and this is closely associated with the progesterone functions together with estradiol to regulate
pulsatile manner of its secretion. A specific region between kisspeptin, because both receptors are needed for positive
− 2012 and − 1597 bp was found to be responsible for this feedback of the LH surge, and both have been indentified in
pulsatility [43]. The involvement of Ca2+ and a Ca2 + − binding kisspeptin neurons, but neither are found in GnRH neurons
protein, downstream regulatory element antagonist modula- [59]. Testosterone exerts negative feedback effects on the
tor (DREAM), in GnRH1 pulsatility has also been found, playing release of GnRH from the hypothalamus. Immunohistochem-
a possible role in the communication between cytoplasm and ical analyses in rats demonstrated that hypothalamic GnRH
nucleus [44]. A variety of hormones and second messengers neurons do not express the androgen receptor, thus, the
regulate GnRH gene expression. In the rat, the promoter effects of testosterone may be mediated by higher-level
region contains two Octamer-binding transcription factor-1 afferent neuronal populations, such as kisspeptin neurons
binding sites that play a critical role in the regulation of rat [60], or through its conversion to estradiol. It was found that
GnRH1 transcription [45]. Within the enhancer, two POU (Pit-1, glucocorticoids repress GnRH1 gene expression and release
Oct-1/2 and Unc-86) homeoprotein Octamer-binding tran- [61], and dehydroepiandrosterone sulfate (DHEAS) inhibits
scription factor-1-binding sites were also identified [46]. Two GnRH gene expression in male rats [62].
other homeodomain proteins, Msx1 and distalless homeobox Concerning other physiological regulators, it is known that
2 (Dlx2), the first of which is a repressor and the second an starvation inhibits the normal function of the reproductive
activator, have been found to be responsible for GnRH1 gene axis. It was found that insulin and insulin-like growth factor 1
regulation. Other studies have revealed that GnRH1 gene (IGF-1) stimulate GnRH gene expression by inducing mitogen-
regulation is also mediated through the protein kinase C (PKC) activated protein kinase (MAPK) pathway. Moreover, insulin-
ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7 7

dependent stimulation of the GnRH gene is mediated by a mapped to human chromosome 19p13.3 and has five exons,
transcription factor, early growth response-1 (Egr-1) [63,64]. encoding a 398-amino acid protein with seven trans-
Melatonin downregulates GnRH mRNA levels [65], and nitric membrane domains [77]. The KiSS1R is found throughout
oxide suppress the expression of the GnRH gene [66]. About the body, such as to brain, pituitary gland, placenta, gonads,
retinoic acid it was found that all-trans-retinoic acid stimu- gastrointestinal tract, liver, and vascular system [78]. After
lates GnRH1 gene expression, whereas 9-cis-retinoic acid binding the ligand, KiSS1R activates phospholipase C and
represses it [67]. Finally, it has been shown that GABA induces recruits intracellular messengers, inositol triphosphate and
rat GnRH gene expression in mature rat GnRH neurons, diacylglycerol, which in turn lead to the release of calcium
through hyperpolarization of the neurons, but in embryonic and PKC activation [79].
rat GnRH neurons, GABA reduce rat GnRH gene expression, In the human hypothalamus, kisspeptin neurons are
through depolarization of the neurons [68]. localized in the rostral preoptic area and infundibular
nucleus. While the localization in the infundibular/arcuate
3.2. GnRH Pulse Generator nucleus is preserved in all species, the population in the
rostral area is species specific [80]. Studies in humans and
GnRH pulse generator is activated in the fetus after the animals have shown the stimulating role of kisspeptin on
completion of GnRH neuron migration to the hypothalamus. gonadotropin secretion [81–83]. Kisspeptin axons compose
Hypothalamic secretion of GnRH increases in early postnatal pericapillary plexuses in the human infundibular stalk, where
life, also leading to temporary activation of gonadal steroido- the GnRH is secreted [80], and GnRH neurons express KiSS1R
genesis, especially in the male. During childhood, gonads can [84]. Some studies support that, beyond the indirect stimula-
respond to exogenous gonadotropin stimulation but remain tion which kisspeptin evokes in the secretion of gonadotro-
normally quiescent because the pulsatile GnRH release is pins through the action on GnRH, there is a directly
suppressed until its re-awakening at the onset of puberty. The stimulating action on the pituitary gonadotrophs to release
mechanism responsible for the quiescent infantile and FSH and LH. This was based on the expression of kisspeptin
prepubertal period is still not fully understood [3]. gene and peptide in gonadotrophs [85]. The stimulatory effect
GnRH secretion follows two distinct modes: pulsatile and of kisspeptin on FSH is less marked and less consistent than
surge [69]. The surge mode occurs only in females. Pulsatile on LH in humans and rodents [86,87].
mode refers to episodic release of GnRH, where there are Hypothalamic expression of kisspeptin and its receptor is
distinct pulses of GnRH secretion into the portal circulation, increased at puberty in rodents and primates [88,89]. The role
with undetectable GnRH concentrations during inter-pulse of kisspeptin in the reproductive system of humans became
intervals. It was first demonstrated in ovariectomised rhesus apparent from patients with pubertal disorders. In patients
monkeys, and subsequently in humans [70]. with HH and impaired pubertal development, inactivating
GnRH neurons co-ordinate their activity, and show intrin- point mutations and deletions in KiSS1R were identified
sic electrical activity, that depends on intracellular signaling [90,91]. Missense mutations in KISS1 gene have also been
and mechanisms. Episodic multi-unit electrical activity at the reported in three unrelated children with central precocious
medial basal hypothalamus suggests that the ‘GnRH pulse puberty. These mutant peptides demonstrated more resis-
generator’ is anatomically located there [71]. Functionally, the tance to in vitro degradation but normal affinity to KiSS1R,
‘GnRH pulse generator’ relies on complex relations between thus suggesting increased bioavailability as the mechanism of
neurons that contain norepinephrine, dopamine, serotonin, precocious puberty [92]. A correlation was found between
GABA, glutamate, neuropeptide Y (NPY) and galanin. Gluta- energy status and kisspeptin system. There is a possibility
mate and norepinephrine stimulate the reproductive axis, that decreased kisspeptin secretion is a putative mechanism
whereas GABA inhibits it. Moreover, other elements, such as for HH in obese and diabetic patients, because the low levels
the kisspeptin-neurokinin B-opioid pathway, have a crucial of hypothalamic kisspeptin mRNA, gonadotropins and sex
role in the regulation of GnRH pulsatility [72]. steroids found in the diabetic rat model was corrected by the
administration of kisspeptin [93,94]. Moreover, a disruption in
pubertal development in states of negative energy balance
4. Kisspeptin, Leptin, Neurokinin B and has been observed and kisspeptin mRNA and gonadotropin
Dynrophin Regulating GnRH Production secretion is reduced in mice, rats and monkeys during fasting
[95–97]. A putative mechanism is through leptin.
Kisspeptin is encoded by KiSS1, which was first discovered in Leptin is a product of adipose tissue and it was first
Hershey in 1996 as a suppressor of metastasis in human isolated in 1994 [98]. It is coded by the lep gene, which is
malignant melanoma, and took its name after the famous expressed in adipocytes. Leptin acts via its receptors LepR,
chocolates ‘Kisses’ produced in the town [73]. The KiSS1 gene whose long isoform mediates most of its actions. Leptin levels
is located on chromosome 1q32, including four exons, of are higher in the obese people and they are correlated with fat
which the first two are not translated [74], and it encodes a mass. Leptin decreases appetite and increases energy expen-
precursor 145 amino acid peptide, which is cleaved to a 54 diture. Moreover, abnormal levels of leptin have been
amino acid peptide. This peptide can be further cleaved to 14, associated with HH and fertility problems in males and
13 and 10 amino-acid peptides, all sharing the C-terminal rodents [99]. Leptin stimulates the HPG axis, through the
sequence of Arg-Phe-NH2 [75]. The orphan G-protein coupled increase of the expression of hypothalamic GnRH and the
receptor 54 (GPR54) was identified, in 2001, as the receptor for stimulation on LH pulsatility [100]. This stimulatory effect is
kisspeptins. It was first described in the rat brain [76]. It is not a direct one because leptin receptor is not present in
8 ME TA BOL I SM C LI N I CA L A ND E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

Fig. 2 – Regulation of the hypothalamic-pituitary-gonadal axis. Diagram of neuroanatomy of the GnRH pathway showing
neural systems that regulate GnRH secretion and feedback mechanisms. (+): stimulatory, (−): inhibitory, Era: estrogen receptor
a, PR: progesterone receptor, GABA: gamma-aminobutyric acid, GnRH: gonadotropin-releasing hormone, GnIH: gonadotropin-
inhibitory hormone.

GnRH neurons but is present in 40% of kisspeptin neurons in of gonadotropins in various biochemical processes, such as
the mouse arcuate nucleus [101]. hormone secretion, its receptor binding and rate of degrada-
In 2009, a role for neurokinin B in hypothalamic regulation tion [105].
was also demonstrated when it was reported that missense
mutations in TAC3 (which encodes neurokinin B) and TAC3R 4.1. GnRH Receptors
(which encodes the neurokinin B receptor) resulted in HH
[102]. Two years earlier, another group had demonstrated GnRH binds to a membrane receptor on pituitary
that, in a subset of neurons in the arcuate nucleus of the ewe, gonadotrophs and stimulates the biosynthesis and secretion
kisspeptin and neurokinin were co-localized, and in addition of LH and FSH. The GnRH receptor is a member of the
a third peptide was co-expressed on the same neurons. This rhodopsin-like G protein-coupled receptor superfamily
peptide was called dynorphin, which is an endogenous opioid (GPCR), which is characterized by the seven-transmembrane
[103]. Based on the first letter of each of these peptides, these domain structure. An important feature of the mammalian
neurons called by the acronym KNDy. In contrast to GnRH receptor is the absence of the cytoplasmic C-terminal
neurokinin B, that stimulates GnRH release, dynorphin tail [106], which is present in non-mammalian GnRH recep-
inhibits GnRH release [104] (Fig. 2). tors [107]. The gene of the human GnRH receptor is composed
GnRH pulse frequency influences the ratio of FSH to LH of three exons separated by two introns, and it has been
secretion. A slower pulse frequency leads to a decrease in LH identified as a single copy on chromosome 4q21.2. The
secretion and to an increase in FSH secretion. In that way, the human gene has some additional features that do not exist
ratio of FSH to LH secretion is elevated. The ratio of FSH to in other species, such as the location of the transcription start
LH secretion is also affected by the extent of FSH and LH sites and the presence of TATA boxes [108]. GnRH receptors
glycosylation, the localization of N-glycans and their and splice variants have been identified in normal human
branching. Variations in glycosylation affect the participation pituitary and pituitary adenomas [109]. In normal
ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7 9

adenohypophysis the receptor is expressed in gonadotrophs, entry stimulates gonadotropin release [127]. The MAPK plays an
thyrotrophs, and somatotrophs [110]. GnRH receptors have important role in the intracellular signaling, providing a crucial
also been identified in the cytotrophoblast and link for the transmission of signals from the cell surface to the
syncytiotrophoblast cell layers of human placenta, human nucleus, thus affecting gonadotropin transcription. Activation of
corpus luteum, luteinized granulose cells, epithelial ovarian extracellular signal–regulated kinase (ERK)-one of the MAPK
carcinoma, ovarian cancer cell lines, endometrial carcinoma cascades - by GnRH in these cells depends mainly on the
cell lines, normal and neoplastic prostatic tissues, prostatic phosphorylation of Raf1 by PKC, supported by a pathway
cancer cell lines, breast carcinoma, melanoma, mammary involving c-Src, dynamin, and Ras. On the other hand, the
cancer lines, liver, heart, skeletal muscle, kidney and periph- activation of c-Jun N-terminal kinase (JNK), another MAPK
eral blood mononuclear cells [35,106]. Ligand binding assays cascade, involves PKC, c-Src, CDC42/Rac1, and probably MEKK1.
for GnRH receptors in human ovary and testis yielded The GnRH receptor can also couple Gs, leading to the activation of
negative results [111], but a more recent study demonstrate adenylyl cyclase (AC) and a rise in cyclic adenosine
GnRH and GnRH receptor mRNA in human testis tissue [112]. monophosphate (cAMP) levels and the activation of protein
It is well documented that the expression of GnRH receptor in kinase A (PKA) and cAMP response element-binding protein
the pituitary is regulated by GnRH-I. When a decline in GnRH-I (CREB) [128,129] (Fig. 3). However, the signaling mechanism of the
stimulation to the pituitary is observed, as occurs in multiple GnRH receptor in some human reproductive tract tumors differs
conditions including lactation, undernutrition, or seasonal pe- significantly. In them, the transmission of GnRH receptor signal
riods of reproductive quiescence, the number of GnRH receptors mainly occurs via Gi, epidermal growth factor receptor (EGFR), c-
on pituitary gonadotrophs declines, but subsequent exposure of Src, and is not dependent on PKC [130,131].
the pituitary to GnRH-I induces and restores the levels of After the activation of GPCR follow their desensitization
receptors via calcium mobilization. This is known as upregula- and internalization. For many GPCRs phosphorylation within
tion or self-priming. This augmentation of GnRH receptor the C-terminal tail of the receptor facilitates binding to β-
production is differentially controlled by varying GnRH-I pulse arrestins, leading to arrestin-dependent receptor desensitiza-
frequencies and differs among species [113]. In cultured rat tion and internalization [132]. In this process a crucial role is
pituitary cells the maximal augmentation of GnRH receptor played by the carboxyl-terminal receptor tail that is absent in
production occurs at a frequency of every 30 min [114], and in the mammalian GnRH receptor [133]. As a result, they do not
human ovarian cancer and peripheral blood mononuclear cells a undergo agonist-induced phosphorylation and they are
biphasic effect of GnRH-I on GnRH receptor expression has been therefore resistant to receptor desensitization, and internal-
observed [115,116]. On the other hand, continuous exposure to ize slowly. The underlying mechanism is not well known, but
GnRH brings about down-regulation of GnRH receptors and, may involve post-receptor adaptive processes such as IP3
subsequently, the inhibition in LH and FSH synthesis and receptor down-regulation and desensitization of Ca2+ mobi-
secretion, called desensitization. Clinical applications of the lization in pituitary cells [134].
down-regulation include the therapy for precocious puberty of It must be mentioned that there are two major types of
hypothalamic origin and endocrine treatment of prostate cancer. receptors in vertebrates, but the pattern of distribution is
The use of a long-acting GnRH agonist down-regulates pituitary unusual. Type I GnRH receptors are not restricted to mam-
GnRH receptors, and suppress the premature activation of mals, but can be present in the lobe-finned and cartilaginous
reproductive axis [117]. On the other hand, the effect of GnRH-II fishes. Skate and coelacanth are the only examples of animals
or its analog significantly inhibits GnRH receptor expression, with both type I and II GnRH receptors [135]. In many
regardless of the concentration used [118]. mammalian species, type II GnRH receptor genes are
Estradiol suppresses GnRH receptor expression both at the disrupted or deleted. Humans belong to this category,
pituitary and extrapituitary tissues, via an ERα dependent possessing a gene encoding type II GnRH receptor but lacking
mechanism [119,120]. Similarly, progesterone inhibits GnRH a functional type II GnRH receptor [136]. The type II GnRH
receptor expression in the pituitary [121]. Activin stimulates the receptor possesses the carboxyl-terminal tails and shows
synthesis of GnRH receptor in rat pituitary cultures and its rapid desensitization and internalization [134].
antagonist follistatin decrease the basal expression of the gene
[122]. Finally, it was found that human chorionic gonadotropin 4.2. Gonadotropin-Inhibitory Hormone
downregulates the GnRH receptor in the rat testis [123], but
increases GnRH receptor gene transcription in choriocarcinoma It seems that GnRH is not the only hypothalamic regulator of
JEG-3 cells [124], indicating a tissue specific effect. the release of gonadotropins. In 2000 a new hypothalamic
Mutations of the GnRH receptor gene lead to idiopathic HH, neuropeptide, that inhibits pituitary gonadotropin secretion,
which is an autosomal inherited disorder and, in contrast was discovered [137]. It is a decapeptide, now referred to as
with KS, is not associated with anosmia [125]. gonadotropin-inhibitory hormone (GnIH). It was originally
The nature of G protein-coupled signaling mediating GnRH identified in birds and afterwards in mammals and other
responses depends on the cellular context. There is evidence that vertebrates [138]. GnIH neurons are located in the
GnRH receptors can couple to multiple G proteins. In the pituitary paraventricular nucleus of the hypothalamus [139]. GnIH
gonadotrophs the primary pathway involves Gq/11 [126]. After inhibits the release and the synthesis of LHβ- and FSHβ-
the coupling phospholipase C is activated, and this results in the subunits, by acting through a GPCR (GPR 147) on the pituitary
formation of diacylglycerols and inositol-trisphosphate (IP3), and on GnRH neurons of birds and mammals [138]. Moreover,
which leads to the activation of several PKC isoforms and the the presence of GnIH and GnIH receptor in the gonads, along
mobilization of intracellular Ca2+, respectively. Increased calcium with the expression of them in cells related to steroid
10 ME TA BOL I SM C LI N I CA L A ND E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

Fig. 3 – GnRH signaling cascade. AC: adenylylcyclase, cAMP: cyclic adenosine monophosphate, PKA: protein kinase A, CREB:
cAMP response element-binding protein, PLC: phospholipase C, DAG: diacylglycerol, PKC: protein kinase C, JNK: c-Jun N-
terminal kinase, ERK: extracellular signal–regulated kinase.

biosynthesis and gamete maturation, reveals the regulatory covalently to a hormone-specific β-subunit (FSHβ and LHβ)
role of GnIH on steroidogenesis and gametogenesis [140]. In [144]. The activation of the α-subunit synthesis is Ca2+
hamsters GnIH was found in seminiferous tubules and GnIH dependent. This process is enhanced by PKC and requires
receptor expression was confined to spermatocytes and ERK1/2 and c-SRC [145]. An essential role of JNK has been
spermatides [141]. In macaque GnIH is expressed in Leydig found by others [146]. The activation of the β-subunit
cells, Sertoli cells, spermatogonia and spermatocytes [140]. synthesis requires the Ca2+, PKC and MAPC signaling path-
The physiological role of GniH in mammals remains still ways [147]. α- and β-subunits are synthesized as a precursor,
largely unresolved. GnIH suppresses LH secretion in juvenile with a signal peptide preceding the authentic subunit
and prepubertal mice, but it is unlikely to have a major role in sequence. In the endoplasmic reticulum proteins undergo
the timing, progression or duration of puberty [142]. It was cleavage and N-glycosylation, leading to immature α/β
found that GnIH could interrupt kisspeptin-induced activa- heterodimers. Along passing the Golgi apparatus their glyco-
tion of GnRH neurons [143]. GnIH may also act as a mediator sylation and conformation take place, and subsequently they
of stress-induced reproductive disruption [138] (Fig. 2). will be stored in separate granules. GnRH-II is also capable to
stimulate FSH and LH secretion acting through the GnRH type
4.3. Gonadotropins I receptor [38].
FSH and LH are essential as regulators of ovarian and
As referred to above GnRH-I stimulates the synthesis and testicular function. In men, LH stimulate testosterone pro-
release of FSH and LH. They are heterodimeric glycoprotein duction of Leydig cells, which in turn exerts sexual and
hormones that consisted of a common α-subunit bound non- anabolic actions, and in addition participates in the
ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7 11

maintenance of spermatogenesis through its paracrine action FSH increases cAMP levels of inhibin in testes. In human adult
on Sertoli cells. The latter also forms the target of FSH. Studies inhibin levels are inversely correlated with serum FSH and
in rats and mice have shown that the role of testosterone is positively correlated with Sertoli cell number, sperm count,
crucial for spermatogenesis, especially its distal part, and sperm concentration, and testicular volume [159].
relatively independent of FSH. Moreover, testosterone is Activins are dimers of the two β-subunits, βA and βB,
essential for male genital differentiation and growth [148]. giving rise to three activins, A (βΑβΑ), ΑΒ (βΑβΒ) and B (βBβΒ).
FSH stimulates Sertoli cell proliferation in prepuberty and Two other β-subunits were also found (βC and βE) but their
defines the size of their population, including the size of the function is still uknown [160]. Activins are gonadotroph cell-
testes and the quantity of spermatogenesis, and acting as a derived ligands which act through a hetero-oligomeric serine/
factor constraining apoptosis [149]. Hence, the main action of threonine receptor complex and intracellular Smad proteins
FSH is, alongside testosterone, the augmentation of the sperm [161].
production [148]. Inhibin down-regulates FSH release from the anterior
Human LH receptor and FSH receptor are encoded by pituitary, whereas activin stimulates it. Inhibin acts primarily
single copy genes on the short arm of chromosome 2 and in gonadotrophs, where it reduces the levels of FSHβ mRNA
separated by 200 kb. They belong to the rhodopsin/2 adren- and FSH protein by blocking activin-stimulated transcription.
ergic receptor-like family A of GPCRs [150]. The rat LH receptor Antagonism of activin action by inhibin is caused by
gene is encoded by a 70 kb genomic DNA and contain 11 interaction of the inhibin β-subunit with the activin receptor,
exons. The FSH receptor is encoded by a 190 kb genomic DNA that prevents the initiation of the intracellular activin
and contains 10 exons. Both receptors consist of an N- signaling cascade. Because inhibin has a lower affinity to
terminal extracellular domain that includes a number of activin receptors than activin itself, a collateral protein acting
irregular leucine-rich repeats (LRR) where the ligands bind, as a coreceptor is necessary. One such protein, betaglycan,
the seven transmembrane domain responsible for signal also known as the type III TGFβ receptor, functions as an
transduction, and a short intracellular C-terminal tail. The inhibin coreceptor, and potentiates antagonistic action of
LH receptor binds both LH and human chorionic gonadotropin inhibin on activin [160,162].
(hCG) with high affinity, whereas the FSH receptor only binds Follistatin is a cysteine-rich, glycosylated, monomeric
FSH. protein [163]. It modulates the bioactivity of activin dimers,
Νaturally occurring activating and inactivating mutations being their functional antagonist. Follistatin binds to the
have been reported in gonadotropin receptor genes. The first activin subunits, blocking their interactions with receptors.
identified mutation was a constitutively activating mutation It has an inhibitory effect on pituitary FSH release. FSH
of LH receptor in a male patient with precocious puberty [151]. stimulates the gonadotrophs to produce follistatin, and
Since then similar mutations have been reported by others follistatin inactivates activin locally [164]. Follistatin expres-
[152]. Asp578His has been found to be responsible for sion is found in gonadal tissue and in pituitary gonadotrophs.
hyperplasia and Leydig cell adenoma [153]. LH receptor In addition, it is found in non-reproductive tissue suggesting a
defects in human 46,XY individuals result in insufficient paracrine or autocrine role [160] (Fig. 2). Activin A and
testosterone production during the period of fetal sex follistatin are also expressed in the hypothalamus.
differentiation, leading to disorders of sex development
(DSD) characterized by a variety of phenotypes, from com-
plete feminisation of external genitalia, to minor hypospadias
or even just micropenis [154,155]. In knockout mice for LH and 5. Conclusions
LH receptor, normal prenatal and pre-pubertal sexual differ-
entiation and gonadal development was found, but the male In this review, beginning from the embryonic development
animals failed to undergo puberty and showed a diminished and anatomic distribution of GnRH neurons, we follow the
sex steroid production [156]. Loss of the FSH receptor function pathways of the HPG axis in various species. The HPG axis
by knockout leads to decreased numbers of Sertoli and functions as a complex unit in the regulation of reproduction
somewhat reduced spermatogenesis, but the animals remain and fertility. There are numerous studies regarding the
fertile [157]. factors that influence the migration of GnRH neurons,
Activin, inhibin and follistatin regulate gonadotroph func- because GnRH plays a particular critical role in the function
tion. They are expressed at all levels of the HPG axis, and in of the reproductive axis, and acts as the intermediate factor
several nonreproductive tissue, suggesting a possible biolog- between hypothalamus and hypophysis. Several GnRHs have
ical role in various physiologic functions [158]. been found from protochondrates to vertebrates. The pulsa-
Inhibin is a glycoprotein hormone that belongs to the tile manner of their secretion is essential for optimal
transforming growth factor-β (TGF-β) superfamily. It is gonadotropin synthesis and secretion in mice to males.
mainly produced by the Sertoli cells. Inhibin consists of two Kisspeptin, first described in the rat brain, with species-
disulphide-linked subunits, a common α subunit (20 kDa) and specific localization in hypothalamus, is a potent upstream
a β subunit (13 kDa). The latter exists in two forms, βA and βB, stimulator of GnRH and gonadotropin secretion. GnRHs act
giving rise in two isoforms, inhibin A (αβA) and inhibin B through GnRH receptors, which in mammals differ from the
(αβB). Ιnhibin B is the major form produced in the human lower species by their absence of the cytoplasmic c-terminal
male. Its concentration is high during early postnatal life, tail, which suppresses their desensitization and internaliza-
reaching a still detectable level until the beginning of puberty. tion. GnIH is another hypothalamic regulator of gonadotro-
During puberty, it again increases and declines with ageing. pins, originally identified in birds, and subsequently in
12 ME TA BOL I SM C LI N I CA L A ND E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

mammals and other vertebrates. GnRH stimulates the syn- and reproductive system in mice lacking prokineticin
thesis of FSH and LH, the main tropic regulators of testicular receptor PKR2. Proc Natl Acad Sci U S A 2006;103:4140–5.
https://doi.org/10.1073/pnas.0508881103.
functions in all species.
[15] Dodé C, Teixeira L, Levilliers J, Fouveaut C, Bouchard P,
Kottler ML, et al. Kallmann syndrome: mutations in the
genes encoding prokineticin-2 and prokineticin receptor-2.
Conflicts of Interest PLoS Genet 2006;2:e175. https://doi.org/10.1371/journal.
pgen.0020175.
[16] Martin C, Balasubramanian R, Dwyer AA, Au MG, Sidis Y,
None.
Kaiser UB, et al. The role of the Prokineticin 2 pathway in
human reproduction: evidence from the study of human
and murine gene mutations. Endocr Rev 2011;32:225–46.
REFERENCES https://doi.org/10.1210/er.2010-0007.
[17] Corradi A, Croci L, Broccoli V, Zecchini S, Previtali S, Wurst
W, et al. Hypogonadotropic hypogonadism and peripheral
[1] Fink G. 60 YEARS OF NEUROENDOCRINOLOGY: MEMOIR: neuropathy in Ebf2-null mice. Development 2003;130:
Harris' neuroendocrine revolution: of portal vessels and 401–10.
self-priming. J Endocrinol 2015;226:T13–24. https://doi.org/ [18] Gamble JA, Karunadasa DK, Pape JR, Skynner MJ, Todman
10.1530/JOE-15-0130. MG, Bicknell RJ, et al. Disruption of ephrin signaling
[2] Schally AV, Arimura A, Kastin AJ, Matsuo H, Baba Y, Redding associates with disordered axophilic migration of the
TW, et al. Gonadotropin-releasing hormone: one gonadotropin-releasing hormone neurons. J Neurosci 2005;
polypeptide regulates secretion of luteinizing and follicle- 25:3142–50. https://doi.org/10.1523/JNEUROSCI.4759-04.2005.
stimulating hormones. Science 1971;173:1036–8. [19] Kramer PR, Wray S. Novel gene expressed in nasal region
[3] Plant TM. 60 YEARS OF NEUROENDOCRINOLOGY: the influences outgrowth of olfactory axons and migration of
hypothalamo-pituitary-gonadal axis. J Endocrinol 2015;226: luteinizing hormone-releasing hormone (LHRH) neurons.
T41–54. https://doi.org/10.1530/JOE-15-0113. Genes Dev 2000;14:1824–34.
[4] Schwanzel-Fukuda M, Pfaff DW. Origin of luteinizing [20] Cariboni A, Rakic S, Liapi A, Maggi R, Goffinet A, Parnavelas
hormone-releasing hormone neurons. Nature 1989;338: JG. Reelin provides an inhibitory signal in the migration of
161–4. https://doi.org/10.1038/338161a0. gonadotropin-releasing hormone neurons. Development
[5] Yoshida K, Tobet SA, Crandall JE, Jimenez TP, Schwarting 2005;132:4709–18. https://doi.org/10.1242/dev.02033.
GA. The migration of luteinizing hormone-releasing [21] Giacobini P, Giampietro C, Fioretto M, Maggi R, Cariboni A,
hormone neurons in the developing rat is associated with a Perroteau I, et al. Hepatocyte growth factor/scatter factor
transient, caudal projection of the vomeronasal nerve. J facilitates migration of GN-11 immortalized LHRH neurons.
Neurosci 1995;15:7769–77. Endocrinology 2002;143:3306–15. https://doi.org/10.1210/en.
[6] Herbison AE. Physiology of the adult gonadotropin-releasing 2002-220146.
hormone neuronal network. In: Plant TM, Zeleznik AJ, [22] Legouis R, Hardelin JP, Levilliers J, Claverie JM, Compain S,
editors. Knobil and Neill's physiology of reproduction. 5. San Wunderle V, et al. The candidate gene for the X-linked
Diego, CA, USA: Elsevier Inc.; 2015. p. 399–467. Kallmann syndrome encodes a protein related to adhesion
[7] Millar RP. GnRHs and GnRH receptors. Anim Reprod Sci 2005;88: molecules. Cell 1991;67:423–35.
5–28. https://doi.org/10.1016/j.anireprosci.2005.05.032. [23] Hardelin JP, Julliard AK, Moniot B, Soussi-Yanicostas N,
[8] Wierman ME, Kiseljak-Vassiliades K, Tobet S. Verney C, Schwanzel-Fukuda M, et al. Anosmin-1 is a
Gonadotropin-releasing hormone (GnRH) neuron migration: regionally restricted component of basement membranes
initiation, maintenance and cessation as critical steps to and interstitial matrices during organogenesis: implications
ensure normal reproductive function. Front for the developmental anomalies of X chromosome-linked
Neuroendocrinol 2011;32:43–52. https://doi.org/10.1016/j. Kallmann syndrome. Dev Dyn 1999;215:26–44. https://doi.
yfrne.2010.07.005. org/10.1002/(SICI)1097-0177(199905)215:1<26::AID-DVDY4>3.
[9] Yoshida K, Rutishauser U, Crandall JE, Schwarting GA. 0.CO;2-D.
Polysialic acid facilitates migration of luteinizing hormone- [24] Esteban PF, Murcia-Belmonte V, García-González D, de
releasing hormone neurons on vomeronasal axons. J Castro F. The cysteine-rich region and the whey acidic
Neurosci 1999;19:794–801. protein domain are essential for anosmin-1 biological
[10] Bless E, Raitcheva D, Henion TR, Tobet S, Schwarting GA. functions. J Neurochem 2013;124:708–20. https://doi.org/10.
Lactosamine modulates the rate of migration of GnRH 1111/jnc.12104.
neurons during mouse development. Eur J Neurosci 2006;24: [25] Bick D, Franco B, Sherins RJ, Heye B, Pike L, Crawford J, et al.
654–60. https://doi.org/10.1111/j.1460-9568.2006.04955.x. Brief report: intragenic deletion of the KALIG-1 gene in
[11] Bless EP, Walker HJ, Yu KW, Knoll JG, Moenter SM, Kallmann's syndrome. N Engl J Med 1992;326:1752–5.
Schwarting GA, et al. Live view of gonadotropin-releasing https://doi.org/10.1056/NEJM199206253262606.
hormone containing neuron migration. Endocrinology 2005; [26] Hu Y, Bouloux PM. X-linked GnRH deficiency: role of KAL-1
14:463–8. https://doi.org/10.1210/en.2004-0838. mutations in GnRH deficiency. Mol Cell Endocrinol 2011;346:
[12] Giacobini P, Kopin AS, Beart PM, Mercer LD, Fasolo A, Wray 13–20. https://doi.org/10.1016/j.mce.2011.04.001.
S. Cholecystokinin modulates migration of gonadotropin- [27] Valdes-Socin H, Rubio Almanza M, Tomé Fernández-
releasing hormone-1 neurons. J Neurosci 2004;24:4737–48. Ladreda M, Debray FG, Bours V, Beckers A. Reproduction,
https://doi.org/10.1523/JNEUROSCI.0649-04.2004. smell, and neurodevelopmental disorders: genetic defects
[13] Kim SH, Hu Y, Cadman S, Bouloux P. Diversity in fibroblast in different hypogonadotropic hypogonadal syndromes.
growth factor receptor 1 regulation: learning from the Front Endocrinol (Lausanne) 2014;5:109. https://doi.org/10.
investigation of Kallmann syndrome. J Neuroendocrinol 3389/fendo.2014.00109 [eCollection 2014].
2008;20:141–63. https://doi.org/10.1111/j.1365-2826.2007. [28] Kim SH. Congenital hypogonadotropic hypogonadism and
01627.x. Kallmann syndrome: past, present, and future. Endocrinol
[14] Matsumoto S, Yamazaki C, Masumoto KH, Nagano M, Naito Metab (Seoul) 2015;30:456–66. https://doi.org/10.3803/EnM.
M, Soga T, et al. Abnormal development of the olfactory bulb 2015.30.4.456.
ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7 13

[29] Miraoui H, Dwyer AA, Sykiotis GP, Plummer L, Chung W, and functional analysis in Gt1-7 neuronal cells. Nucleic
Feng B, et al. Mutations in FGF17, IL17RD, DUSP6, SPRY4, and Acids Res 1996;24:3614–20.
FLRT3 are identified in individuals with congenital [44] Leclerc GM, Boockfor FR. Calcium influx and DREAM protein
hypogonadotropic hypogonadism. Am J Hum Genet 2013;92: are required for GnRH gene expression pulse activity. Mol
725–43. https://doi.org/10.1016/j.ajhg.2013.04.008. Cell Endocrinol 2007;267:70–9. https://doi.org/10.1016/j.mce.
[30] Ikemoto T, Park MK. Molecular and evolutionary 2006.12.040.
characterization of the GnRH-II gene in the chicken: [45] Eraly SA, Nelson SB, Huang KM, Mellon PL. Oct-1 binds
distinctive genomic organization, expression pattern, and promoter elements required for transcription of the GnRH
precursor sequence. Gene 2006;368:28–36. https://doi.org/10. gene. Mol Endocrinol 1998;12:469–81. https://doi.org/10.
1016/j.gene.2005.10.004. 1210/mend.12.4.0092.
[31] Sasaki K, Norwitz ER. Gonadotropin-releasing hormone/ [46] Clark ME, Mellon PL. The POU homeodomain transcription
gonadotropin-releasing hormone receptor signaling in the factor Oct-1 is essential for activity of the gonadotropin-
placenta. Curr Opin Endocrinol Diabetes Obes 2011;18:401–8. releasing hormone neuron-specific enhancer. Mol Cell Biol
https://doi.org/10.1097/MED.0b013e32834cd3b0. 1995;15:6169–77.
[32] Chen A, Ganor Y, Rahimipour S, Ben-Aroya N, Koch Y, Levite [47] Han YG, Kang SS, Seong JY, Geum D, Suh YH, Kim K. Negative
M. The neuropeptides GnRH-II and GnRH-I are produced by regulation of gonadotropin-releasing hormone and
human T cells and trigger laminin receptor gene expression, gonadotropin-releasing hormone receptor gene expression by a
adhesion, chemotaxis and homing to specific organs. Nat gonadotrophin-releasing hormone agonist in the rat hypothal-
Med 2002;8:1421–6. https://doi.org/10.1038/nm801. amus. J Neuroendocrinol 1999;11:195–201.
[33] Sakamoto Y, Harada T, Horie S, Iba Y, Taniguchi F, Yoshida [48] Shivers BD, Harlan RE, Morrell JI, Pfaff DW. Absence of
S, et al. Tumor necrosis factor–induced interleukin-8 (IL-8) oestradiol concentration in cell nuclei of LHRH-
expression in endometriotic stromal cells, probably through immunoreactive neurones. Nature 1983;304:345–7.
nuclear factor-B activation: gonadotropin-releasing [49] Kalló I, Butler JA, Barkovics-Kalló M, Goubillon ML, Coen CW.
hormone agonist treatment reduced IL-8 expression. J Clin Oestrogen receptor beta-immunoreactivity in gonadotropin
Endocrinol Metab 2003;88:730–5. https://doi.org/10.1210/jc. releasing hormone-expressing neurones: regulation by
2002-020666. oestrogen. J Neuroendocrinol 2001;13:741–8.
[34] Pierantoni R, Cobellis G, Meccariello R, Cacciola G, Chianese [50] Hrabovszky E, Kalló I, Szlávik N, Keller E, Merchenthaler I,
R, Chioccarelli T, et al. Testicular gonadotropin-releasing Liposits Z. Gonadotropin-releasing hormone neurons
hormone activity, progression of spermatogenesis, and express estrogen receptor-beta. J Clin Endocrinol Metab
sperm transport in vertebrates. Ann N Y Acad Sci 2009;1163: 2007;92:2827–30. https://doi.org/10.1210/jc.2006-2819.
279–91. https://doi.org/10.1111/j.1749-6632.2008.03617.x. [51] Navarro CE, Saeed SA, Murdock C, Martinez-Fuentes AJ,
[35] Pazaitou-Panayiotou K, Chemonidou C, Poupi A, Koureta M, Arora KK, Krsmanovic LZ, et al. Regulation of cyclic
Kaprara A, Lambropoulou M, et al. Gonadotropin-releasing adenosine 3′,5′- monophosphate signaling and pulsatile
hormone neuropeptides and receptor in human breast neurosecretion by Gi-coupled plasma membrane estrogen
cancer: correlation to poor prognosis parameters. Peptides receptors in immortalized gonadotrophin-releasing
2013;42:15–24. https://doi.org/10.1016/j.peptides.2012.12. hormone neurons. Mol Endocrinol 2003;17:1792–804.
016. [52] Roy D, Angelini NL, Belsham DD. Estrogen directly
[36] Miyamoto K, Hasegawa Y, Nomura M, Igarashi M, Kangawa respresses gonadotropin-releasing hormone (GnRH) gene
K, Matsuo H. Identification of the second gonadotropin- expression in estrogen receptor-alpha (ERalpha)- and
releasing hormone in chicken hypothalamus: evidence that ERbeta-expressing GT1-7 GnRH neurons. Endocrinology
gonadotropin secretion is probably controlled by two 1999;140:5045–53. https://doi.org/10.1210/endo.140.11.7117.
distinct gonadotropin-releasing hormone in avian species. [53] Herbison AE, Horvath TL, Naftolin F, Leranth C. Distribution
Proc Natl Acad Sci U S A 1984;81:3874–8. of estrogen receptor-immunoreactive cells in monkey
[37] White RB, Eisen JA, Kasten TL, Fernald RD. Second form of hypothalamus: relationship to neurones containing
gonadotropin-releasing hormone in humans. Proc Natl luteinizing hormone-releasing hormone and tyrosine
Acad Sci U S A 1998;95:305–9. hydroxylase. Neuroendocrinology 1995;61:1–10.
[38] Densmore VS, Urbanski HF. Relative effect of gonadotropin- [54] Dungan HM, Clifton DK, Steiner RA. Minireview: kisspeptin
releasing hormone (GnRH)-I and GnRH-II on gonadotropin neurons as central processors in the regulation of
release. J Clin Endocrinol Metab 2003;88:2126–34. https://doi. gonadotropin-releasing hormone secretion. Endocrinology
org/10.1210/jc.2002-021359. 2006;147:1154–8. https://doi.org/10.1210/en.2005-1282.
[39] Fernald RD, White RB. Gonadotropin-releasing hormone [55] Langub Jr MC, Watson Jr RE. Estrogen receptor-
genes: phylogeny, structure, and functions. Front immunoreactive glia, endothelia, and ependyma in guinea
Neuroendocrinol 1999;20:224–40. https://doi.org/10.1006/ pig preoptic area and median eminence: electron
frne.1999.0181. microscopy. Endocrinology 1992;130:364–72. https://doi.org/
[40] Lee VH, Lee LT, Chow BK. Gonadotropin-releasing hormone: 10.1210/endo.130.1.1727710.
regulation of the GnRH gene. FEBS J 2008;275:5458–78. [56] Herbison AE. Estrogen positive feedback to gonadotropin-
https://doi.org/10.1111/j.1742-4658.2008.06676.x. releasing hormone (GnRH) neurons in the rodent: the case
[41] Seeburg PH, Adelman JP. Characterization of cDNA for for the rostral periventricular area of the third ventricle
precursor of human luteinizing hormone releasing (RP3V). Brain Res Rev 2008;57:277–87. https://doi.org/10.
hormone. Nature 1984;311:666–8. 1016/j.brainresrev.2007.05.006.
[42] Wolfe A, Kim HH, Tobet S, Stafford DE, Radovick S. [57] Chan YM, Broder-Fingert S, Wong KM, Seminara SB.
Identification of a discrete promoter region of the human Kisspeptin/Gpr54-independent gonadotrophin-releasing
GnRH gene that is sufficient for directing neuron-specific hormone activity in Kiss1 and Gpr54 mutant mice. J
expression: a role for POU homeodomain transcription Neuroendocrinol 2009;21:1015–23. https://doi.org/10.1111/j.
factors. Mol Endocrinol 2002;16:435–49. https://doi.org/10. 1365-2826.2009.01926.x.
1210/mend.16.3.0780. [58] Krsmanovic LZ, Hu L, Leung PK, Feng H, Catt KJ. The
[43] Kepa JK, Spaulding AJ, Jacobsen BM, Fang Z, Xiong X, hypothalamic GnRH pulse generator: multiple regulatory
Radovick S, et al. Structure of the distal human mechanisms. Trends Endocrinol Metab 2009;20:402–8.
gonadotropin releasing hormone (hGnrh) gene promoter https://doi.org/10.1016/j.tem.2009.05.002.
14 ME TA BOL I SM C LI N I CA L A ND E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

[59] Rudolph LM, Bentley GE, Calandra RS, Paredes AH, Tesone [75] Kotani M, Detheux M, Vandenbogaerde A, Communi D,
M, Wu TJ, et al. Peripheral and central mechanisms involved Vanderwinden JM, Le Poul E, et al. The metastasis
in the hormonal control of male and female reproduction. J suppressor gene KiSS-1 encodes kisspeptins, the natural
Neuroendocrinol 2016;28. https://doi.org/10.1111/jne.12405. ligands of the orphan G protein-coupled receptor GPR54. J
[60] Bliss SP, Navratil AM, Xie J, Roberson MS. GnRH signaling, Biol Chem 2001;37:34631–6. https://doi.org/10.1074/jbc.
the gonadotrope and endocrine control of fertility. Front M104847200.
Neuroendocrinol 2010;31:322–40. https://doi.org/10.1016/j. [76] Ohtaki T, Shintani Y, Honda S, Matsumoto H, Hori A,
yfrne.2010.04.002. Kanehashi K, et al. Metastasis suppressor gene KiSS-1
[61] Attardi B, Tsujii T, Friedman R, Zeng Z, Roberts JL, Dellovade encodes peptide ligand of a G-protein-coupled receptor.
T, et al. Glucocorticoid repression of gonadotropin-releasing Nature 2001;6837:613–7. https://doi.org/10.1038/35079135.
hormone gene expression and secretion in morphologically [77] Muir AI, Chamberlain L, Elshourbagy NA, Michalovich D,
distinct subpopulations of GT1-7 cells. Mol Cell Endocrinol Moore DJ, Calamari A, et al. AXOR12, a novel human G
1997;131:241–55. protein-coupled receptor, activated by the peptide KiSS-1. J
[62] Li S, Garcia de Yebenes E, Pelletier G. Effects of Biol Chem 2001;31:28969–75. https://doi.org/10.1074/jbc.
dehydroepiandrosterone (DHEA) on GnRH gene expression M102743200.
in the rat brain as studied by in situ hybridization. Peptides [78] Clarke SA, Dhillo WS. Kisspeptin across the human lifespan:
1995;16:425–30. evidence from animal studies and beyond. J Endocrinol
[63] DiVall SA, Radovick S, Wolfe A. Egr-1 binds the GnRH 2016;229:R83-8. https://doi.org/10.1530/JOE-15-0538.
promoter to mediate the increase in gene expression by [79] Constantin S, Caligioni CS, Stojilkovic S, Wray S. Kisspeptin-
insulin. Mol Cell Endocrinol 2007;270:64–72. https://doi.org/ 10 facilitates a plasma membrane-driven calcium
10.1016/j.mce.2007.02.007. oscillator in gonadotropin-releasing hormone-1 neurons.
[64] Zhen S, Zakaria M, Wolfe A, Radovick S. Regulation of Endocrinology 2009;3:1400–12. https://doi.org/10.1210/en.
gonadotropin-releasing hormone (GnRH) gene expression 2008-0979.
by insulin-like growth factor I in a cultured GnRH- [80] Hrabovszky E, Ciofi P, Vida B, Horvath MC, Keller E, Caraty A,
expressing neuronal cell line. Mol Endocrinol 1997;11: et al. The kisspeptin system of the human hypothalamus:
1145–55. https://doi.org/10.1210/mend.11.8.9956. sexual dimorphism and relationship with gonadotropin-
[65] Roy D, Angelini NL, Fujieda H, Brown GM, Belsham DD. releasing hormone and neurokinin B neurons. Eur J
Cyclical regulation of GnRH gene expression in GT1-7 GnRH- Neurosci 2010;11:1984–98. https://doi.org/10.1111/j.1460-
secreting neurons by melatonin. Endocrinology 2001;142: 9568.2010.07239.x.
4711–20. https://doi.org/10.1210/endo.142.11.8464. [81] Clarkson J, Han SY, Piet R, McLennan T, Kane GM, Ng J, et al.
[66] Belsham DD, Wetsel WC, Mellon PL. NMDA and nitric oxide Definition of the hypothalamic GnRH pulse generator in
act through the cGMP signal transduction pathway to mice. Proc Natl Acad Sci U S A 2017. https://doi.org/10.1073/
repress hypothalamic gonadotropin-releasing hormone pnas.1713897114 [pii: 201713897].
gene expression. EMBO J 1996;15:538–47. [82] Dhillo W, Chaudhuri O, Patterson M, Thompson E, Murphy
[67] Cho S, Chung J, Han J, Ju LB, Han KD, Rhee K, et al. 9-cis- K, Badman M, et al. Kisspeptin-54 stimulates the
Retinoic acid represses transcription of the gonadotropin- hypothalamic-pituitary-gonadal axis in human males. J Clin
releasing hormone (GnRH) gene via proximal promoter Endocrinol Metab 2005;90:6609–15. https://doi.org/10.1210/
region that is distinct from all-trans-retinoic acid response jc.2005-1468.
element. Brain Res Mol Brain Res 2001;87:214–22. [83] Thompson E, Patterson M, Murphy K, Smith K, Dhillo W,
[68] Funabashi T, Daikoku S, Suyama K, Mitsushima D, Sano A, Todd J, et al. Central and peripheral administration of
Kimura F. Role of gamma-aminobutyric acid neurons in the kisspeptin-10 stimulates the hypothalamic–pituitary–
release of gonadotropin-releasing hormone in cultured rat gonadal axis. J Neuroendocrinol 2004;16:850–8. https://doi.
embryonic olfactory placodes. Neuroendocrinology 2002;76: org/10.1111/j.1365-2826.2004.01240.x.
193–202. https://doi.org/10.1159/000065950. [84] Messager S, Chatzidaki EE, Ma D, Hendrick AG, Zahn D,
[69] Maeda K, Ohkura S, Uenoyama Y, Wakabayashi Y, Oka Y, Dixon J, et al. Kisspeptin directly stimulates
Tsukamura H, et al. Neurobiological mechanisms gonadotropinreleasing hormone release via G protein-
underlying GnRH pulse generation by the hypothalamus. coupled receptor 54. Proc Natl Acad Sci U S A 2005;5:1761–6.
Brain Res 2010;1364:103–15. https://doi.org/10.1016/j. https://doi.org/10.1073/pnas.0409330102.
brainres.2010.10.026. [85] Richard N, Galmiche G, Corvaisier S, Caraty A, Kottler ML.
[70] Antunes JL, Carmel PW, Housepian EM, Ferin M. Luteinizing KiSS-1 and GPR54 genes are co-expressed in rat
hormone-releasing hormone in human pituitary blood. J gonadotrophs and differentially regulated in vivo by
Neurosurg 1978;49:382–6. https://doi.org/10.3171/jns.1978. oestradiol and gonadotrophin-releasing hormone. J
49.3.0382. Neuroendocrinol 2008;3:381–93. https://doi.org/10.1111/j.
[71] Wilson RC, Kesner JS, Kaufman JM, Uemura T, Akema T, 1365-2826.2008.01653.x.
Knobil E. Central electrophysiologic correlates of pulsatile [86] George JT, Veldhuis JD, Roseweir AK, Newton CL, Faccenda
luteinizing hormone secretion in the Rhesus monkey. E, Millar RP, et al. Kisspeptin-10 is a potent stimulator of LH
Neuroendocrinology 1984;39:256–60. and increases pulse frequency in men. J Clin Endocrinol
[72] Ezzat A, Pereira A, Clarke IJ. Kisspeptin is a component of Metab 2011;8:E1228–36. https://doi.org/10.1210/jc.2011-0089.
the pulse generator for gonadotropin releasing hormone [87] Navarro VM, Castellano JM, Fernandez-Fernandez R, Tovar
(GnRH) secretion in female sheep but not THE pulse S, Roa J, Mayen A, et al. Effects of KiSS-1 peptide, the natural
generator. Endocrinology 2015;156:1828–37. https://doi.org/ ligand of GPR54, on follicle-stimulating hormone secretion
10.1210/en.2014-1756. in the rat. Endocrinology 2005;4:1689–97. https://doi.org/10.
[73] Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman BE, 1210/en.2004-1353.
et al. KiSS-1, a novel human malignant melanoma metastasis- [88] Navarro VM, Castellano JM, Fernandez-Fernandez R,
suppressor gene. J Natl Cancer Inst 1996;88:1731–7. Barreiro ML, Roa J, Sanchez-Criado JE, et al. Developmental
[74] West A, Vojta PJ, Welch DR, Weissman BE. Chromosome and hormonally regulated messenger ribonucleic acid
localization and genomic structure of the KiSS-1 Metastasis expression of KiSS-1 and its putative receptor, GPR54, in rat
Suppressor Gene (KISS1). Genomics 1998;54:145–8. https:// hypothalamus and potent luteinizing hormone-releasing
doi.org/10.1006/geno.1998.5566.
ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7 15

activity of KiSS-1 peptide. Endocrinology 2004;10:4565–74. neurokinin B. Endocrinology 2007;148:5752–60. https://doi.


https://doi.org/10.1210/en.2004-0413. org/10.1210/en.2007-0961.
[89] Shahab M, Mastronardi C, Seminara SB, Crowley WF, Ojeda [104] Cheng G, Coolen LM, Padmanabhan V, Goodman RL,
SR, Plant TM. Increased hypothalamic GPR54 signaling: a Lehman MN. The kisspeptin/neurokinin B/dynorphin
potential mechanism for initiation of puberty in primates. (KNDy) cell population of the arcuate nucleus: sex
Proc Natl Acad Sci U S A 2005;6:2129–34. https://doi.org/10. differences and effects of prenatal testosterone in sheep.
1073/pnas.0409822102. Endocrinology 2010;151:301–11. https://doi.org/10.1210/en.
[90] de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, 2009-0541.
Milgrom E. Hypogonadotropic hypogonadism due to loss of [105] Fares F. The role of O-linked and N-linked oligosaccharides
function of the KiSS1-derived peptide receptor GPR54. Proc on the structure-function of glycoprotein hormones:
Natl Acad Sci U S A 2003;19:10972–6. https://doi.org/10.1073/ development of agonists and antagonists. Biochim Biophys
pnas.1834399100. Acta 1760;2006:560–7. https://doi.org/10.1016/j.bbagen.2005.
[91] Seminara SB, Messager S, Chatzidaki EE, Thresher RR, 12.022.
Acierno Jr JS, Shagoury JK, et al. The GPR54 gene as a [106] Cheng CK, Leung PC. Molecular biology of gonadotropin-
regulator of puberty. N Engl J Med 2003;17:1614–27. https:// releasing hormone (GnRH)-I, GnRH-II, and their receptors in
doi.org/10.1056/NEJMoa035322. humans. Endocr Rev 2005;26:283–306. https://doi.org/10.
[92] Silveira LG, Noel SD, Silveira-Neto AP, Abreu AP, Brito VN, 1210/er.2003-0039.
Santos MG, et al. Mutations of the KISS1 gene in disorders of [107] Lethimonier C, Madigou T, Munoz-Cueto JA, Lareyre JJ, Kah
puberty. J Clin Endocrinol Metab 2010;5:2276–80. https://doi. O. Evolutionary aspects of GnRHs, GnRH neuronal systems
org/10.1210/jc.2009-2421. and GnRH receptors in teleost fish. Gen Comp Endocrinol
[93] Castellano JM, Navarro VM, Roa J, Pineda R, Sanchez-Garrido 2004;135:1–16.
MA, Garcia-Galiano D, et al. Alterations in hypothalamic [108] Kakar SS. Molecular structure of the human
KiSS-1 system in experimental diabetes: early changes and gonadotropinreleasing hormone receptor gene. Eur J
functional consequences. Endocrinology 2009;2:784–94. Endocrinol 1997;137:183–92.
https://doi.org/10.1210/en.2008-0849. [109] Grosse R, Schoneberg T, Schultz G, Gudermann T. Inhibition
[94] George JT, Millar RP, Anderson RA. Hypothesis: kisspeptin of gonadotropin-releasing hormone receptor signaling by
mediates male hypogonadism in obesity and type 2 expression of a splice variant of the human receptor. Mol
diabetes. Neuroendocrinology 2010;4:302–7. https://doi.org/ Endocrinol 1997;11:1305–18. https://doi.org/10.1210/mend.
10.1159/000299767. 11.9.9966.
[95] Castellano JM, Navarro VM, Fernandez-Fernandez R, [110] Sanno N, Jin L, Qian X, Osamura RY, Scheithauer BW,
Nogueiras R, Tovar S, Roa J, et al. Changes in hypothalamic Kovacs K, et al. Gonadotropin-releasing hormone and
KiSS-1 system and restoration of pubertal activation of the gonadotropin-releasing hormone receptor messenger ribo-
reproductive axis by kisspeptin in undernutrition. nucleic acids expression in nontumorous and neoplastic
Endocrinology 2005;9:3917–25. https://doi.org/10.1210/en. pituitaries. J Clin Endocrinol Metab 1997;82:1974–82. https://
2005-0337. doi.org/10.1210/jcem.82.6.3976.
[96] Roa J, Garcia-Galiano D, Varela L, Sanchez-Garrido MA, [111] Clayton RN, Huhtaniemi IT. Absence of gonadotropin-
Pineda R, Castellano JM, et al. The mammalian target of releasing hormone receptors in human gonadal tissue.
rapamycin as novel central regulator of puberty onset via Nature 1982;299:56–9.
modulation of hypothalamic Kiss1 system. Endocrinology [112] Bahk JY, Hyun JS, Chung SH, Lee H, Kim MO, Lee BH, et al.
2009;11:5016–26. https://doi.org/10.1210/en.2009-0096. Stage specific identification of the expression of GnRH
[97] Wahab F, Ullah F, Chan YM, Seminara SB, Shahab M. mRNA and localization of the GnRH receptor in mature rat
Decrease in hypothalamic Kiss1 and Kiss1r expression: a and adult human testis. J Urol 1995;154:1958–61.
potential mechanism for fasting-induced suppression of the [113] Tsutsumi M, Laws SC, Rodic V, Sealfon SC. Translational
HPG axis in the adult male rhesus monkey (Macaca mulatta). regulation of the gonadotropin-releasing hormone receptor
Horm Metab Res 2011;2:81–5. https://doi.org/10.1055/s-0030- in T3–1 cells. Endocrinology 1995;136:1128–36. https://doi.
1269852. org/10.1210/endo.136.3.7867566.
[98] Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, [114] Kaiser UB, Jakubowiak A, Steinberger A, Chin WW.
Friedman JM. Positional cloning of the mouse obese gene Differential effects of gonadotropin-releasing hormone
and its human homologue. Nature 1994;372:425–32. https:// (GnRH) pulse frequency on gonadotropin subunit and GnRH
doi.org/10.1038/372425a0. receptor messenger ribonucleic acid levels in vitro.
[99] Jahan S, Bibi R, Ahmed S, Kafeel S. Leptin levels in infertile Endocrinology 1997;138:1224–31. https://doi.org/10.1210/
males. J Coll Physicians Surg Pak 2014;21:393–707.2011/ endo.138.3.4968.
JCPSP.393397. [115] Kang SK, Choi KC, Cheng KW, Nathwani PS, Auersperg N,
[100] Quennell JH, Mulligan AC, Tups A, Liu X, Phipps SJ, Kemp CJ, Leung PC. Role of gonadotropin-releasing hormone as an
et al. Leptin indirectly regulates gonadotropin-releasing autocrine growth factor in human ovarian surface
hormone neuronal function. Endocrinology 2009;150: epithelium. Endocrinology 2000;141:72–80. https://doi.org/
2805–12. https://doi.org/10.1210/en.2008-1693. 10.1210/endo.141.1.7250.
[101] Smith JT, Acohido BV, Clifton DK, Steiner RA. KiSS-1 [116] Chen HF, Jeung EB, Stephenson M, Leung PC. Human
neurones are direct targets for leptin in the ob/ob mouse. J peripheral blood mononuclear cells express gonadotropin-
Neuroendocrinol 2006;4:298–303. https://doi.org/10.1111/j. releasing hormone (GnRH), GnRH receptor, and interleukin-
1365-2826.2006.01417.x. 2 receptor -chain messenger ribonucleic acids that are
[102] Topaloglu AK, Reimann F, Guclu M, Yalin AS, Kotan LD, regulated by GnRH in vitro. J Clin Endocrinol Metab 1999;84:
Porter KM, et al. TAC3 and TACR3 mutations in familial 743–50. https://doi.org/10.1210/jcem.84.2.5440.
hypogonadotropic hypogonadism reveal a key role for [117] Lahlou N, Carel JC, Chaussain JL, Roger M. Pharmacokinetics
Neurokinin B in the central control of reproduction. Nat and pharmacodynamics of GnRH agonists: clinical
Genet 2009;41:354–8. https://doi.org/10.1038/ng.306. implications in pediatrics. J Pediatr Endocrinol Metab 2000;
[103] Goodman RL, Lehman MN, Smith JT, Coolen LM, de Oliveira 13(Suppl. 1):723–37.
CV, Jafarzadehshirazi MR, et al. Kisspeptin neurons in the [118] Kang SK, Tai CJ, Nathwani PS, Leung PC. Differential
arcuate nucleus of the ewe express both dynorphin A and regulation of two forms of gonadotropin-releasing hormone
16 ME TA BOL I SM C LI N I CA L A ND E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7

messenger ribonucleic acid in human granulosa-luteal cells. [133] Ferguson SS. Evolving concepts in G protein-coupled
Endocrinology 2001;142:182–92. https://doi.org/10.1210/ receptor endocytosis: the role in receptor desensitization
endo.142.1.7895. and signaling. Pharmacol Rev 2001;53:1–24.
[119] Quinones-Jenab V, Jenab S, Ogawa S, Funabashi T, Weesner [134] McArdle CA, Franklin J, Green L, Hislop JN. Signaling, cycling
GD, Pfaff DW. Estrogen regulation of gonadotropin-releasing and desensitization of gonadotropin-releasing hormone
hormone receptor messenger RNA in female rat pituitary receptors. J Endocrinol 2002;173:1–11.
tissue. Mol Brain Res 1996;38:243–50. [135] Roch GJ, Busby ER, Sherwood NM. GnRH receptors and
[120] Cheng CK, Chow BK, Leung PC. An activator protein 1-like peptides: skating backward. Gen Comp Endocrinol 2014;209:
motif mediates 17beta-estradiol repression of 118–34. https://doi.org/10.1016/j.ygcen.2014.07.025.
gonadotropin-releasing hormone receptor promoter via an [136] Stewart AJ, Katz AA, Millar RP, Morgan K. Retention and
estrogen receptor alpha-dependent mechanism in ovarian silencing of prepro-GnRH-II and type II GnRH receptor genes
and breast cancer cells. Mol Endocrinol 2003;17:2613–29. in mammals. Neuroendocrinology 2009;90:416–32. https://
https://doi.org/10.1210/me.2003-0217. doi.org/10.1159/000233303.
[121] Laws SC, Beggs JM, Webster JC, Miller WL. Inhibin increases [137] Tsutsui K, Saigoh E, Ukena K, Teranishi H, Fujisawa Y,
and progesterone decrease receptor for gonadotropin- Kikuchi M, et al. A novel avian hypothalamic peptide
releasing hormone in ovine pituitary cultures. inhibiting gonadotropin release. Biochem Biophys Res
Endocrinology 1990;127:373–80. https://doi.org/10.1210/ Commun 2000;275:661–7. https://doi.org/10.1006/bbrc.2000.
endo-127-1-373. 3350.
[122] Fernandez-Vazquez G, Kaiser UB, Albarracin CT, Chin [138] Tsutsui K, Ubuka T, Bentley GE, Kriegsfeld LJ. Gonadotropin-
WW. Transcriptional activation of the gonadotropin- inhibitory hormone (GnIH): discovery, progress and
releasing hormone receptor gene by activin A. Mol prospect. Gen Comp Endocrinol 2012;177:305–14. https://doi.
Endocrinol 1996;10:356–66. https://doi.org/10.1210/mend. org/10.1016/j.ygcen.2012.02.013.
10.4.8721981. [139] Ukena K, Ubuka T, Tsutsui K. Distribution of a novel avian
[123] Botte MC, Lerrant Y, Lozach A, Berault A, Counis R, Kottler gonadotropin-inhibitory hormone in the quail brain. Cell
ML. LH down-regulates gonadotropin-releasing hormone Tissue Res 2003;312:73–9. https://doi.org/10.1007/s00441-
(GnRH) receptor, but not GnRH, mRNA levels in the rat 003-0700-x.
testis. J Endocrinol 1999;162:409–15. [140] McGuire NL, Bentley GE. Neuropeptides in the gonads: from
[124] Cheng KW, Leung PC. Human chorionic gonadotropin- evolution to pharmacology. Front Pharmacol 2010;1:114.
activated cAMP pathway regulates human placental GnRH https://doi.org/10.3389/fphar.2010.00114.
receptor gene transcription in choriocarcinoma JEG-3 cells. J [141] Zhao S, Zhu E, Yang C, Bentley GE, Tsutsui K, Kriegsfeld LJ.
Clin Endocrinol Metab 2002;87:3291–9. https://doi.org/10. RFamide -related peptide and messenger ribonucleic acid
1210/jcem.87.7.8650. expression in mammalian testis: association with the
[125] Janovick JA, Goulet M, Bush E, Greer J, Wettlaufer DG, Conn spermatogenic cycle. Endocrinology 2010;151:617–27.
PM. Structure-activity relations of successful pharmacologic https://doi.org/10.1210/en.2009-0978.
chaperones for rescue of naturally occurring and [142] Poling MC, Kauffman AS. Regulation and function of RFRP-3
manufactured mutants of the gonadotropin-releasing (GnIH) neurons during postnatal development. Front
hormone receptor. J Pharmacol Exp Ther 2003;305:608–14. Endocrinol (Lausanne) 2015;6:150. https://doi.org/10.3389/
https://doi.org/10.1124/jpet.102.048454. fendo.2015.00150.
[126] Grosse R, Schmid A, Schoneberg T, Herrlich A, Muhn P, [143] Wu M, Dumalska I, Morozova E, van den Pol AN, Alreja M.
Schultz G, et al. Gonadotropin-releasing hormone receptor Gonadotropin inhibitory hormone inhibits basal forebrain
initiates multiple signaling pathways by exclusively vGluT2-gonadotropin-releasing hormone neurons via a
coupling to Gq/11 proteins. J Biol Chem 2000;275:9193–200. direct postsynaptic mechanism. J Physiol 2009;587(Pt 7):
[127] Stojilkovic SS, Reinhart J, Catt KJ. Gonadotropin-releasing 1401–11. https://doi.org/10.1113/jphysiol.2008.166447.
hormone receptors: structure and signal transduction [144] Hamernik DL. Molecular biology of gonadotropins. J Reprod
pathways. Endocr Rev 1994;15:462–99. https://doi.org/10. Fertil Suppl 1995;49:257–69.
1210/edrv-15-4-462. [145] Harris D, Chuderland D, Bonfil D, Kraus S, Seger R, Naor Z.
[128] Cheng KW, Nathwani PS, Leung PC. Regulation of human Extracellular signal-regulated kinase and c-Src, but not Jun
gonadotropin-releasing hormone receptor gene expression Nterminal kinase, are involved in basal and gonadotropin-
in placental cells. Endocrinology 2000;141:2340–9. https:// releasing hormone-stimulated activity of the glycoprotein
doi.org/10.1210/endo.141.7.7543. hormone -subunit promoter. Endocrinology 2003;144:
[129] Perrett RM, McArdle CA. Molecular mechanisms of 612–22. https://doi.org/10.1210/en.2002-220690.
gonadotropin-releasing hormone signaling: integrating [146] Yokoi T, Ohmichi M, Tasaka K, Kimura A, Kanda Y,
cyclic nucleotides into the network. Front Endocrinol Hayakawa J, et al. Activation of the luteinizing hormone
(Lausanne) 2013;4:180. https://doi.org/10.3389/fendo.2013. promoter by gonadotropin-releasing hormone requires
00180. c-Jun N-terminal protein kinase. J Biol Chem 2000;275:
[130] Kraus S, Naor Z, Seger R. Intracellular signaling pathways 21639–47. https://doi.org/10.1074/jbc.M910252199.
mediated by the gonadotropin-releasing hormone (GnRH) [147] Vasilyev VV, Pernasetti F, Rosenberg SB, Barsoum MJ, Austin
receptor. Arch Med Res 2001;32:499–509. DA, Webster NJ, et al. Transcriptional activation of the ovine
[131] Grundker C, Volker P, Emons G. Antiproliferative signaling follicle-stimulating hormone- gene by gonadotropin-
of luteinizing hormone-releasing hormone in human releasing hormone involves multiple signal transduction
endometrial and ovarian cancer cells through G protein pathways. Endocrinology 2002;143:1651–9. https://doi.org/
i-mediated activation of phosphotyrosine phosphatase. 10.1210/endo.143.5.8771.
Endocrinology 2001;142:2369–80. https://doi.org/10.1210/ [148] Huhtaniemi I. A short evolutionary history of FSH-
endo.142.6.8190. stimulated spermatogenesis. Hormones (Athens) 2015;14:
[132] Finch AR, Sedgley KR, Armstrong SP, Caunt CJ, McArdle CA. 468–78. https://doi.org/10.14310/horm.2002.1632.
Trafficking and signalling of gonadotrophin-releasing [149] Sharpe RM, McKinnell C, Kivlin C, Fisher JS. Proliferation and
hormone receptors: an automated imaging approach. Br J functional maturation of Sertoli cells, and their relevance to
Pharmacol 2010;159:751–60. https://doi.org/10.1111/j.1476- disorders of testis function in adulthood. Reproduction
5381.2009.00413.x. 2003;125:769–84.
ME T ABOL I SM C L IN I CA L A N D E XP E RI ME N TAL 86 ( 20 1 8 ) 3 – 1 7 17

[150] Dias JA, Cohen BD, Lindau-Shepard B, Nechamen CA, receptordeficient mice, but not FSHbeta-deficient mice:
Peterson AJ, Schmidt A. Molecular, structural, and cellular role for constitutive FSH receptor activity.
biology of follitropin and follitropin receptor. Vitam Horm Endocrinology 2003;144:138–45. https://doi.org/10.1210/en.
2002;64:249–322. 2002-220637.
[151] Shenker A, Laue L, Kosugi S, Merendino Jr JJ, Minegishi T, [158] Schneider O, Nau R, Michel U. Comparative analysis of
Cutler Jr GB. A constitutively activating mutation of the follistatin-, activin beta A- and activin beta B-mRNA
luteinizing hormone receptor in familial male precocious steady-state levels in diverse porcine tissues by multiplex
puberty. Nature 1993;365:652–4. https://doi.org/10.1038/ S1 nuclease analysis. Eur J Endocrinol 2000;142:537–44.
365652a0. [159] Iliadou PK, Tsametis C, Kaprara A, Papadimas I, Goulis DG.
[152] Menon KM, Menon B. Structure, function and regulation of The Sertoli cell: novel clinical potentiality. Hormones
gonadotropin receptors - a perspective. Mol Cell Endocrinol (Athens) 2015;14:504–14. https://doi.org/10.14310/horm.
2012;356:88–97. https://doi.org/10.1016/j.mce.2012.01.021. 2002.1648.
[153] Boot AM, Lumbroso S, Verhoef-Post M, Richter-Unruh A, [160] Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WP,
Looijenga LH, Funaro A, et al. Mutation analysis of the LH Schwartz NB, et al. Inhibin at 90: from discovery to clinical
receptor gene in Leydig cell adenoma and hyperplasia and application, a historical review. Endocr Rev 2014;35:747–94.
functional and biochemical studies of activating mutations https://doi.org/10.1210/er.2014-1003.
of the LH receptor gene. J Clin Endocrinol Metab 2011;96: [161] Attisano L, Wrana JL. Signal transduction by the TGF-β
E1197–205. https://doi.org/10.1210/jc.2010-3031. superfamily. Science 2002;296:1646–7. https://doi.org/10.
[154] Qiao J, Han B, Liu BL, Chen X, Ru Y, Cheng KX, et al. A splice 1126/science.1071809.
site mutation combined with a novel missense mutation of [162] Lewis KA, Gray PC, Blount AL, MacConell LA, Wiater E,
LHCGR cause male pseudohermaphroditism. Hum Mutat Bilezikjian LM, et al. Betaglycan binds inhibin and can
2009;30:E855-5. https://doi.org/10.1002/humu.21072. mediate functional antagonism of activin signalling. Nature
[155] Themmen APN, Huhtaniemi IT. Mutations of gonadotropins 2000;404:411–4. https://doi.org/10.1038/35006129.
and gonadotropin receptors: elucidating the physiology and [163] Esch FS, Shimasaki S, Mercado M, Cooksey K, Ling N, Ying S,
pathophysiology of pituitary-gonadal function. Endocr Rev et al. Structural characterization of follistatin: a novel
2000;21:551–83. https://doi.org/10.1210/edrv.21.5.0409. follicle-stimulating hormone release-inhibiting polypeptide
[156] Jonas KC, Oduwole OO, Peltoketo H, Rulli SB, Huhtaniemi IT. from the gonad. Mol Endocrinol 1987;1:849–55. https://doi.
Mouse models of altered gonadotrophin action: insight into org/10.1210/mend-1-11-849.
male reproductive disorders. Reproduction 2014;148:R63-0. [164] Kaiser UB, Lee BL, Carroll RS, Unabia G, Chin WW, Childs GV.
https://doi.org/10.1530/REP-14-0302. Follistatin gene expression in the pituitary: localization in
[157] Baker PJ, Pakarinen P, Huhtaniemi IT, Abel MH, Charlton gonadotropes and folliculostellate cells in diestrous rats.
HM, Kumar TR, et al. Failure of normal Leydig cell Endocrinology 1992;130:3048–56. https://doi.org/10.1210/
development in follicle-stimulating hormone (FSH) endo.130.5.1572312.

You might also like