Next Article in Journal
Structural Analysis and Characterization of an Antiproliferative Lectin from Canavalia villosa Seeds
Next Article in Special Issue
The Role of Endocrine Disruptors Bisphenols and Phthalates in Obesity: Current Evidence, Perspectives and Controversies
Previous Article in Journal
Haberlea rhodopensis Extract Tunes the Cellular Response to Stress by Modulating DNA Damage, Redox Components, and Gene Expression
Previous Article in Special Issue
McCune–Albright Syndrome: A Case Report and Review of Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gonadotropin-Releasing Hormone Receptor (GnRHR) and Hypogonadotropic Hypogonadism

by
Pavlos Fanis
1,
Vassos Neocleous
1,
Irene Papapetrou
2,
Leonidas A. Phylactou
1,* and
Nicos Skordis
2,3,*
1
Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
2
School of Medicine, University of Nicosia, Nicosia 1678, Cyprus
3
Division of Paediatric Endocrinology, Paedi Center for Specialized Paediatrics, Nicosia 2024, Cyprus
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(21), 15965; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242115965
Submission received: 4 October 2023 / Revised: 31 October 2023 / Accepted: 2 November 2023 / Published: 4 November 2023
(This article belongs to the Special Issue Hormone Receptors: A 2023 Update)

Abstract

:
Human sexual and reproductive development is regulated by the hypothalamic-pituitary-gonadal (HPG) axis, which is primarily controlled by the gonadotropin-releasing hormone (GnRH) acting on its receptor (GnRHR). Dysregulation of the axis leads to conditions such as congenital hypogonadotropic hypogonadism (CHH) and delayed puberty. The pathophysiology of GnRHR makes it a potential target for treatments in several reproductive diseases and in congenital adrenal hyperplasia. GnRHR belongs to the G protein-coupled receptor family and its GnRH ligand, when bound, activates several complex and tissue-specific signaling pathways. In the pituitary gonadotrope cells, it triggers the G protein subunit dissociation and initiates a cascade of events that lead to the production and secretion of the luteinizing hormone (LH) and follicle-stimulating hormone (FSH) accompanied with the phospholipase C, inositol phosphate production, and protein kinase C activation. Pharmacologically, GnRHR can be modulated by synthetic analogues. Such analogues include the agonists, antagonists, and the pharmacoperones. The agonists stimulate the gonadotropin release and lead to receptor desensitization with prolonged use while the antagonists directly block the GnRHR and rapidly reduce the sex hormone production. Pharmacoperones include the most recent GnRHR therapeutic approaches that directly correct the misfolded GnRHRs, which are caused by genetic mutations and hold serious promise for CHH treatment. Understanding of the GnRHR’s genomic and protein structure is crucial for the most appropriate assessing of the mutation impact. Such mutations in the GNRHR are linked to normosmic hypogonadotropic hypogonadism and lead to various clinical symptoms, including delayed puberty, infertility, and impaired sexual development. These mutations vary regarding their mode of inheritance and can be found in the homozygous, compound heterozygous, or in the digenic state. GnRHR expression extends beyond the pituitary gland, and is found in reproductive tissues such as ovaries, uterus, and prostate and non-reproductive tissues such as heart, muscles, liver and melanoma cells. This comprehensive review explores GnRHR’s multifaceted role in human reproduction and its clinical implications for reproductive disorders.

1. Introduction

Reproduction and development of sexual characteristics, in humans, is under the control of the hypothalamic-pituitary-gonadal (HPG) axis. Gonadotropin-releasing hormone (GnRH) that is produced in the hypothalamic neurosecretory cells is the main regulatory hormone of the HPG axis and acts in a pulsatile manner on the gonadotrope cells of the anterior pituitary gland by binding to GnRH receptors (GnRHRs) [1,2,3]. The stimulated GnRHRs cause the production and secretion of the luteinizing hormone (LH) and the follicle-stimulating hormone (FSH) that are known to control several important functions and have a direct effect on the gonads. Such functions include the gonadal steroid production, gametogenesis, and the gonadal cell proliferation [4,5,6]. Stimulation of the GnRHR by the GnRH takes place through the Gq/11 heterotrimeric protein pathway [4,5,7]. During childhood, GnRH secretion is suppressed and rises during puberty, in which increased production of gonadotropins and gonadal steroids trigger sexual development [8]. Reproduction development is disrupted somewhat by dysfunction of the GnRHR including causative mutations in the GnRHR gene resulting in the delay of puberty and congenital hypogonadotropic hypogonadism (CHH) [9,10]. In humans, GnRHRs are divided into two subtypes, GnRH1R and GnRH2R, with the expression of the latter being controversial [11,12,13]. The GnRH1R receptor is a member of the rhodopsin G protein-coupled receptor (GPCR) family and, in addition to its primary expression in pituitary gonadotrope cells, it was also found to be expressed in the breast, ovaries, prostate cells and lymphocytes [14,15,16,17]. Given its importance in reproductive regulation, the GnRHR has emerged as a potential target for the treatment of infertility and sex steroid-dependent hyperplasia, such as uterine fibroids, endometriosis, and prostate cancer [18,19,20,21]. In these conditions, gonadal steroid secretion is reduced by the delivery of GnRH antagonists or high doses of GnRH agonists, which reduce the expression of the receptor [22,23]. In this review, the main type of human GnRHR receptor is outlined and its main role in regulating reproduction through complex signaling pathways is described. Additionally, the genomic and protein structure of GNRHR along with ligands, mutations, and therapeutic possibilities are also explored. A discussion on tissue expression patterns of GNRHR, extending beyond the pituitary gland, and their possible implication on clinical entities such as hypogonadotropic hypogonadism is also presented.

2. Genomic and Protein Structure of GnRHR

2.1. Genomic Structure of GnRHR

Using cloning and mapping analyses, the human GnRH receptor gene was found to be located on chromosome 4q13.2–13.3 and consists of three exons separated by two introns covering 17.2 kb on the chromosome [24,25,26,27]. Specifically, the chromosomal location is Chromosome 4: 67,737,118–67,754,388 (genome assembly: GRCh38.p14); gene code (Gene: ENSG00000109163.7). The main transcript of the gene is 4402 bp in length and encodes a protein of size 328 aa and predicted unmodified molecular weight of ~38 kDa (Transcript ID: ENST00000226413.5; Refseq: NM_000406.3). Fan et al. showed, due to the size of the gene, that the promoter and the 3′-UTR regions contain multiple transcription initiation sites and polyadenylation signals, respectively [24]. In 1997, Grose et al. showed the presence in the pituitary of a second smaller transcript generated by alternative splicing and encoding a protein of size 249 aa (Transcript ID: ENST00000000420975.2). Expression of this smaller protein was shown to be inhibitory to the mechanism of GnRHR signaling [28]. The 5′ UTR is found in exon 1 as well as the first 522 nucleotides of the coding sequence, which encode the first three transmembrane (TM) domains and a part of the fourth TM domain. Exon 2 encodes the following 220 nucleotides of the coding sequence, which includes the rest of the fourth TM and the fifth TM domain. Finally, exon 3 consists of the remaining 245 nucleotides of the coding sequence and the 3′ UTR [24,25,26] (Figure 1A).

2.2. Protein Structure of GnRHR

The GnRHR, as a member of the GPCR protein family, is composed of seven alpha-helical transmembrane (TM) domains that span the lipid bilayer of the cell membrane. These seven TMs are designated TM1 to TM7 and are connected by three intracellular (IL1–3) and three extracellular loops (EL1–3). The amino-terminal end (NH2) is on the extracellular side and the carboxyl-terminal end is on the intracellular side [14,29] (Figure 1B). TMs form a barrel-like structure that creates a hydrophobic core, allowing the receptor to integrate itself in the lipid bilayer of the cell membrane. This provides the structural framework of the receptor’s function, allowing it to interact extracellularly with GnRH ligands and intracellularly with G proteins [30,31]. The extracellular loops of the GnRHR help in the stability of the ligand-receptor complex. Specific amino acid residues of the extracellular loops of the receptor form a ligand-binding pocket that allows recognition and binding of the GnRH ligand with high specificity and its partial entry to the transmembrane part of the receptor [31]. Specifically, amino acid residues Asp at position 98, Trp at position 101, Asn at position 102, Lys at position 121, and Asp at position 302 were identified as important residues for ligand binding [31,32,33,34,35] (Figure 1B). The GNRHR intracellular loops are involved in G protein coupling. They contain specific sequence motifs that interact with G proteins, facilitating the activation of downstream signaling pathways. Moreover, intracellularly, the GnRHR differs from other GPCRs in a number of ways, including the absence of the carboxyl COOH terminal tail. This domain is regularly anchored to the membrane in other GPCRs [36] and plays an essential part in short-term desensitization caused by ligand-stimulated phosphorylation of Ser/Thr residues [37]. In GnRHR-mediated inositol phosphate production, the immediate desensitization is not observed, which is in line with the absence of a carboxy-terminal tail. Another difference of GnRHR compared to GPCRs is the substitution of Tyr with Ser, at position 140, in the highly conserved GPCR motif Asp-Arg-Tyr found at the junction of TM3 and the second intracellular loop [38,39,40]. This motif’s Asp and Arg residues have been related to the interaction of several GPCRs to their corresponding G proteins [41]. In addition, the substitution of Ser140 for Tyr had no effect on the GnRHR’s coupling to cytoplasmic G proteins [42,43]. The Tyr140 mutant receptor, on the other hand, increased the degree of receptor internalization and agonist-binding affinity, indicating that this substituted residue has mild impact on GNRHR structure [43]. The conserved residues ile135, ile143, and leu147 in the second intracellular loop of GnRHR play an important role in G protein coupling, validating the significance of this loop for the coupling with G proteins [43,44].

2.3. Signal Transduction Pathways of GNRHR

GnRH and its receptor are differently distributed in the various tissues where they are detected. Consequently, the signaling pathways activated by the binding of GnRH to its receptor, in order to transmit extracellular information intracellularly, are strongly influenced by the type of cells in which they occur [45]. In pituitary gonadotrope cells, binding of GnRH to its receptor results in structural changes in both the receptor and the heterotrimeric GTP-binding proteins (G proteins). Heterotrimeric G proteins comprise three subunits: Gα, Gβ, and Gγ [46,47,48,49,50]. G proteins are differentiated into several subclasses based on differences in the structure of the Gα subunit. In mammals, Gα subunits belong to several subtypes: Gαs, Gαq/11, Gα12/13, Gαi/o, and Gαt [51,52]. In pituitary gonadotrope cells, GnRHR is associated with the Gαq/11 subunit and, by few studies, with the Gαs subunit [46]. The structural changes that occur in the heterotrimeric G protein result in an altered affinity for GDP and its replacement by GTP. This change promotes the detachment of the Gα subunit from the heterotrimer and its separation from the Gβγ dimer subunits, which remain as one [49,53]. Initial studies on the GnRH signaling pathway in pituitary gonadotrope cells revealed that the Gαq/11 subunit intracellularly triggers a series of signaling events by binding to phospholipase Cβ (PLCβ) [54] (Figure 2). Further studies showed that the Gβγ dimer could also bind to and activate PLCβ. It was also shown that upon prolonged GnRH stimulation, other factors such as the phospholipase A2 (PLA2) and phospholipase D (PLD) can be stimulated by either Gα or Gβγ [55]. The primary signaling pathway of GnRH’s response is through the phospholipase PLCβ which catalyzes the enzymatic hydrolysis of the membrane phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2), synthesizing the inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG) [46,56]. IP3 activates the release of Ca2+ into the cytosol through its binding to IP3 receptors on the membrane of the endoplasmic reticulum, which act as Ca2+ channels [57,58]. Due to GnRH activation, Ca2+ accumulation together with DAG are the major causes of the production and secretion of gonadotropins in gonadotrope cells by activating protein kinase C (PKC) [59,60] (Figure 2). In gonadotrope cells, as mentioned above, GnRH binding to GnRHR also causes a delayed activation of PLD. PLD hydrolyzes the membrane phosphatidylcholine (PC) generating phosphatidylethanol (PET) and phosphatidic acid (PA) that eventually result in the production of DAG. The production of DAG by this pathway causes a sustained and prolonged activation of PKC, which has its two isoforms α and βII phosphorylate PLD as a positive feedback mechanism [61,62,63] (Figure 2). PKC activation also induces the activation of fibrosarcoma protein kinase 1 (Raf-1), protein tyrosine kinase src, and certain mitogen-activated protein kinases (MAPKs) [64,65,66]. These kinases act through the MAPK signaling pathway in which the end result is the phosphorylation and activation of transcription factors, including Elk-1, Egr-1, c-Fos, and c-Jun, which have a positive effect on the expression of gonadotropin kinase subunit α and PLA2 [56,64,67] (Figure 2). In addition, the PLA2 phospholipase produces arachidonic acid (AA), which is used as a substrate in a number of intracellular signals. AA is converted into leukotrienes by lipoxygenase, which in elevated concentrations are involved in the gene activation of PKCβ and the gonadotropins α subunit [68,69] (Figure 2). In summary, all signaling pathways activated by the binding of GnRH to the GnRHR receptor are interconnected and eventually result in the production and secretion of gonadotropins, LH, and FSH [5]. Moreover, due to the different pulse frequencies of GnRH secretion, different subunits of gonadotropins are produced. Specifically, when gonadotrope cells are exposed to an increased pulse frequency of GnRH, there is an induction in the production and release of the α subunit of gonadotropin and the β subunit of LH. Conversely, when cells are exposed to a low pulse frequency of GnRH, there is induction in the synthesis and release of the β subunit of FSH [70] (Figure 2).

2.4. Tissue Expression of GnRHR in Humans

Pituitary gland is the main tissue with the highest expression of GNRHR. Initial localization studies regarding the GnRHR transcript(s) identified three different transcripts. The primary transcript is correctly spliced and encodes the full-length protein [25,71]. The second transcript contains a 128-nucleotide deletion in exon 2 that causes alternative splicing, resulting in a truncated protein with a change in amino acid 174 and the addition of an extra 75 new amino acids. It is worth noting that this shorter transcript when expressed together with the full-length causes a dominant-negative effect preventing the wild-type protein from normally entering the cell membrane. The third transcript contains a 220-nucleotide deletion at exon 2 encoding a truncate protein with a size of 177 amino acids [28,72]. Immunoreactivity experiments demonstrated that GnRHR is specifically expressed in the gonadotrope, thyrotrope, and somatotrope cells of the pituitary gland [73,74]. GNRHR expression has been identified in other tissues other than the pituitary gland that are related or not to reproduction. Experiments in different ovarian cell types, as well as in several ovarian cancer lines, have shown in addition to GnRHR protein expression, the presence of GnRHR mRNA transcripts and the presence of binding sites for the GnRH ligand [75,76,77,78]. Binding sites for the GnRH ligand have also been found in uterus related cells, as well as in endometrial cancer lines. GnRHR transcripts have also been found in normal and neoplastic uterine cells [79,80]. GnRHR expression has also been observed in various placental cells such as cytotrophoblasts and syncytiotrophoblasts [81]. Moreover, GnRHR expression has also been detected in normal and neoplastic prostate gland cells [82,83] in various breast cancer cell lines [84,85,86] and in cells and cell lines not associated with reproduction, i.e., heart, skeletal muscles, liver, kidney, peripheral blood mononuclear cells, and melanoma cells [45,87,88].

2.5. GnRHR Characterized Mutations

Since 1997 when de Roux et al. identified the first mutation in the GNRHR gene in a family with hypogonadotropic hypogonadism (HH), several mutations in the gene have been described [89]. The mode of inheritance regarding mutations in the GNRHR follows the autosomal recessive manner and, to date, have been found in both the homozygous and compound heterozygous state. Furthermore, patients with HH have been found to carry, in a digenic mode of inheritance fashion, a heterozygous in the GNRHR and a second mutation in the second allele in one of the ANOS1 (KAL1), FGFR1, GNRH1, FGF8, PROK2, PROKR2, KISS1R, CHD7, TAC3, and TACR3 genes. All these genes have recently reported to be associated with HH [90,91,92,93]. It is worth mentioning that the above-mentioned pathogenic variants have also been associated with HH with phenotypes such as partial or total delayed puberty, infertility, and Kallmann syndrome. In HH cases, mutations in GNRHR account for 3.5–16% of sporadic cases and up to 40% of familial cases [91,94]. To date, 58 mutations have been reported including 48 missense, 3 nonsense, 5 frameshift, 1 in-frame, and 1 splice acceptor [89,90,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129] (Table 1). Interestingly, all of the these mutations are localized in all regions of the receptor except the first transmembrane region (TM1), the first intracellular loop (IL1), and the third extracellular loop (EL3) (Figure 3). The mutations have been described as inactivating and cause an alteration in the function of the receptor either by reducing its expression, its localization, impairing ligand binding, and/or affecting signaling. It is worth mentioning that three of the identified mutations, p.Gln106Arg, p.Arg139His, and p.Arg262Gln, show an increased frequency number compared to the others. According to gnomAD browser (https://gnomad.broadinstitute.org/ (accessed on 2 October 2023)), the allele frequency for p.Gln106Arg is 0.002749, for p.Arg139His is 0.0001630, and for p.Arg262Gln is 0.001789, while allele frequencies for the remaining mutations range from 0.0001291 to 0.000004005. Due to this increased frequency, these three mutations have also been found in non-consanguineous families while the remaining mutations, when found in homozygous tissue, typically come from consanguineous families [122,130,131]. For this reason, in 2015, Choi et al. investigated the possibility of these mutations being inherited from a common ancestor. Indeed, it was proven so that all of the tested patients with these specific mutations shared a common haplotype, thus suggesting that they have been inherited from a common ancestor and behave as founder mutations [131]. Identification and characterization of the mutations will demonstrate the regions of GNRHR that are important for ligand binding, signaling, proper protein folding, and correct localization.

2.6. GnRHR Ligands: Agonists, Antagonists, and Pharmacoperones

The GNRHR is a key regulator of reproductive processes, and a wide range of ligands, including agonists, antagonists, and pharmacoperones, can control its function [23,132,133,134]. These compounds have various effects on the receptor, controlling the secretion of LH and FSH, all of which are important in the male and female reproduction process.

2.7. GnRHR Agonists

2.7.1. Gonadotropin-Releasing Hormone (GnRH)

The endogenous GnRH, when bound to the GNRHR, functions as an agonist. GnRH is synthesized in the hypothalamus and serves as the catalyst for the pituitary gland’s pulsatile secretion of LH and FSH [45]. Its pulsatile secretion behavior is critical for reproductive process regulation.

2.7.2. GnRH Analogues (GnRHa)

Synthetic GnRH analogues are used to treat infertility, endometriosis, uterine fibroids, precocious puberty, hypogonadotropic hypogonadism, and hormone-sensitive cancers of the breast in women and prostate in men [135,136,137,138,139,140,141,142]. In HH treatment, GnRHR analogues bind to GnRHR on the pituitary gland, leading to the initial release of LH and FSH. Upon administration of a GnRHR analogue, there is an initial “flare” effect, in which there is a brief surge of LH and FSH production. This flare can temporarily worsen symptoms in some individuals with HH. After the initial flare, continuous exposure to GnRHR analogue leads to desensitization of the pituitary gland. The pituitary becomes less responsive to GnRH, which results in a decrease in LH and FSH production. By initially stimulating the release of LH and FSH and subsequently downregulating their production, GnRHR analogues can help normalize sex hormone (estrogen and testosterone) levels over time. This process is critical for reproductive regulation due to the decrease in estrogen and testosterone levels in HH patients [135]. Examples of GnRHa used in therapy are the leuprorelin, goserelin, nafarelin, triptorelin, buserelin, and histrelin. In addition, deslorelin is used in veterinary medicine for a variety of purposes such as promoting ovulation and the treatment of high-risk pregnancies in animals. Gonadorelin is used in both humans and animals. In contrast to other GnRH analogues, which are used to inhibit LH and FSH secretion, deslorelin is associated with stimulation of LH secretion (Table 2).

2.8. GnRHR Antagonists

GnRH antagonists, contrary to GnRHa, directly block the GnRH receptor, and thus the action of GnRH, without the initial activation seen with agonists. The resulting LH suppression causes rapid reduction of the production of testosterone in the testes in men, and a reduction of estradiol and progesterone production from the ovaries in women. GnRH antagonists are capable of preventing gonadal sex hormone production and suppressing sex hormone levels [143]. In controlled ovarian stimulation protocols for in vitro fertilisation (IVF), this rapid blockade prevents premature LH surges [144]. GnRH antagonists are used in assisted reproductive technologies for fertility treatment as well as the treatment of conditions such as precocious puberty, endometriosis, uterine fibroids, and prostate cancer [137,138,139,140,145]. Many GnRH antagonists, like cetrorelix, degarelix, abarelix, and ganirelix, have a structure analogous to natural GnRH but have an antagonistic activity, whereas others, like elagolix, linzagolix, and relugolix, are non-peptide compounds (Table 3).

2.9. GnRHR Pharmacoperones

Pharmacological chaperones or Pharmacoperones are small molecules that bind to a target protein and correct or improve its folding, trafficking, or stability. These molecules have the ability to restore proper protein function, making them a potential therapy for genetic disorders caused by misfolded or impaired proteins [134]. In the case of GnRHR, specific genetic mutations can cause its misfolding and retention in the endoplasmic reticulum, preventing it from reaching the cell membrane. Pharmacoperones can therefore successfully rescue mutant GNRHRs that would otherwise be non-functional, helping to properly fold the receptor and thus, traffic it to the cell membrane, while restoring its ability to respond to GnRH [146,147]. All described GnRHR pharmacoperones act as receptor antagonists. The first study was conducted using four different pharmacoperones; IN3, Q89, A177775, and TAK-013, and tested their ability to restore GnRHR function in the COS-7 cell line that expressed the mutant GnRHR. All four pharmacoperones were successful in restoring cell-surface expression and stimulating constitutive activity [148]. The properties of pharmacoperone IN3 were further studied in transgenic mice with a hypogonadotropic hypogonadism phenotype due to the GnRHR p.Glu90Lys mutation. After a 30-day treatment with IN3, male mice showed elevated sperm concentration, positive changes in sperm morphology, and increased expression of steroidogenic enzymes [148]. This approach holds promise for the treatment of genetic disorders caused by mutations in GNRHR, such as congenital hypogonadotropic hypogonadism (CHH).

2.10. Clinical Implications of Mutated GnRHR

The main feature in patients carrying mutations in GnRHR is normosmic hypogonadotropic hypogonadism. These patients exhibit a variety of clinical symptoms characterized by different phenotypic diagnosis and/or different evaluation at age of diagnosis. Spontaneous pulsality of gonadotropins is not normal in patients with GnRHR mutations, showing reduced intensity but normal frequency and absence of pulsality of LH and FSH secretion [89,104,110,119,149,150].
In HH, the HPG axis is disrupted due to dysfunction at the hypothalamic and/or pituitary level. This disorder leads to a deficiency of the key hormones GnRH, LH, and FSH, resulting in a cascade of effects. The hypothalamus fails to produce and release GnRH or does so inadequately. This means that there is insufficient signal to the pituitary gland to stimulate the release of LH and FSH. Regardless of the specific point of dysfunction, the end result is low levels of LH and FSH in the bloodstream. With low levels of LH and FSH, the gonads receive inadequate stimulation. In males, this result in decreased testosterone production, while in females, there is a lack of proper ovarian stimulation for the production of estradiol and progesterone [151,152,153,154]. As a result, individuals with HH have low levels of sex hormones, leading to various clinical features such as delayed or absent puberty, infertility, and absence of secondary sexual characteristics.
More specifically, in childhood and adolescence, HH is characterized by delayed or completely absent of puberty. Girls with HH often do not show the expected signs of puberty at the typical age, such as breast development and minimal body hair growth, while boys show absence of facial and body hair growth, impaired testicular development, and limited muscle growth. In addition, delayed growth and development in children and adolescents, defined by delayed bone age compared to chronological age, can result in shorter stature and slower growth rates compared to their peers. As a consequence of insufficient sex hormone production, individuals with HH in this age group are typically infertile. This is due to the gonads’ inability to produce mature eggs or sperm necessary for reproduction. Adolescents with HH may experience several symptoms related to low levels of sex hormones. In boys, this may include low libido, erectile dysfunction, and fatigue. Girls may experience menstrual irregularities, including primary or secondary amenorrhea (absence of menstruation), as well as symptoms such as mood changes and fatigue. The uterus is the size of that at the prepubertal stage and the ovaries are small or absent due to lack of follicular stimulation. Furthermore, the delayed or absent puberty and the absence of typical secondary sexual characteristics can have a significant psychosocial impact on adolescents. This may lead to feelings of self-consciousness, reduced self-esteem, and emotional challenges [104,114,117,119,125]. In adulthood, HH presents with a range of clinical characteristics that reflect the deficiency of sex hormones, particularly testosterone in males and estradiol in females. These clinical characteristics can vary depending on the underlying cause and the individual’s specific case. In males, HH can result in the loss of secondary sexual characteristics, such as decreased facial and body hair growth, reduced muscle mass, gynecomastia, microphallus, and a decrease in the size of the testes [89,110,113,114,119,149]. In females, there may be a loss of breast development, changes in body fat distribution, and the absence of typical female secondary sexual characteristics. Infertility is a significant concern for individuals with HH in adulthood. The lack of adequate sex hormone production can lead to the inability to conceive naturally. Both men and women with HH may experience infertility, and assisted reproductive technologies may be necessary to achieve pregnancy [96,104,110,111,150,155]. However, hormonal treatment with human chorionic gonadotropin resulted in production of normal sperm counts leading to successful conception and pregnancy [89,149,150,156]. Sexual dysfunction is a common feature of HH in both men and women. In males, this may manifest as erectile dysfunction and reduced libido. In females, it can lead to decreased sexual desire and vaginal dryness. Low sex hormone levels in HH can lead to symptoms of fatigue, reduced energy levels, and a general sense of sickness. This can affect an individual’s quality of life and overall well-being. Men with HH may experience symptoms such as hot flashes (similar to those seen in menopause), changes in body composition, and a decrease in strength and stamina. The lack of testosterone can result in muscle weakness and an increased risk of osteoporosis. Moreover, low testosterone levels in men with HH can be associated with metabolic effects, including increased body fat, insulin resistance, and potentially an increased risk of cardiovascular disease. Women with HH may have irregular menstrual cycles, amenorrhea (absence of menstruation), and symptoms like hot flashes, night sweats, and changes in bone density [157,158,159]. Although most patients with mutations in GnRHR are thought to have hypogonadism early at birth, this is diagnosed later when clinical symptoms are more prominent [160]. The underlying causes of CHH can vary and may include genetic mutations, congenital abnormalities, or disruptions in the hypothalamic-pituitary-gonadal axis. It is important to note that early diagnosis and management of HH in childhood and adolescence are crucial to address these clinical characteristics effectively. Treatment typically involves hormone replacement therapy to induce and support the development of secondary sexual characteristics and normal growth [159,161,162]. Similarly, early diagnosis and management of HH in adulthood are important to address the clinical characteristics effectively. Treatment often involves hormone replacement therapy (e.g., testosterone replacement in men or estradiol/progesterone replacement in women) to correct hormonal imbalances and alleviate associated symptoms. Endocrinologists and specialists in hormonal disorders play a key role in the diagnosis and management of HH in children, adolescents, and adults.

3. Conclusions

In summary, GnRH and its receptor orchestrate complex signaling pathways, influenced by tissue-specific distribution. These pathways involve heterotrimeric G proteins, ultimately leading to gonadotropin production. Various ligands, including agonists, antagonists, and pharmacoperones, modulate the receptor’s function. Genomic and protein structures reveal critical regions and mutations associated with reproductive disorders. GnRH receptor expression extends to diverse tissues. Clinical implications encompass hypogonadotropic hypogonadism with symptoms affecting both genders. Future perspectives include tailored therapies for GnRHR mutations and advancing precision medicine in reproductive disorders. Current research may reveal additional roles for GNRHR in non-reproductive tissues, expanding our understanding of their broader physiological impact and potential therapeutic applications beyond reproduction.

Author Contributions

Conceptualization, P.F., V.N., L.A.P. and N.S.; writing—original draft preparation, P.F., V.N.; writing—review and editing, P.F., V.N., I.P., L.A.P. and N.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the A.G Leventis Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The department of Molecular Genetics, Function and Therapy of the Cyprus Institute of Neurology and Genetics would like to thank the European Reference Network on Rare Endocrine Conditions: Project ID N0 739543 (https://endo-ern.eu/about/reference-centres/ (accessed on 2 October 2023)). We would like to thank Nicolas C. Nicolaides for his contribution to the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maggi, R.; Cariboni, A.M.; Marelli, M.M.; Moretti, R.M.; Andre, V.; Marzagalli, M.; Limonta, P. GnRH and GnRH receptors in the pathophysiology of the human female reproductive system. Hum. Reprod. Update 2016, 22, 358–381. [Google Scholar] [CrossRef] [PubMed]
  2. Schally, A.V.; Arimura, A.; Baba, Y.; Nair, R.M.; Matsuo, H.; Redding, T.W.; Debeljuk, L. Isolation and properties of the FSH and LH-releasing hormone. Biochem. Biophys. Res. Commun. 1971, 43, 393–399. [Google Scholar] [CrossRef] [PubMed]
  3. Plant, T.M.; Zeleznik, A.J. (Eds.) Knobil and Neill’s Physiology of Reproduction, 4th ed.; Academic Press: New York, NY, USA, 2014. [Google Scholar]
  4. Perrett, R.M.; McArdle, C.A. Molecular mechanisms of gonadotropin-releasing hormone signaling: Integrating cyclic nucleotides into the network. Front. Endocrinol. 2013, 4, 180. [Google Scholar] [CrossRef]
  5. Bliss, S.P.; Navratil, A.M.; Xie, J.; Roberson, M.S. GnRH signaling, the gonadotrope and endocrine control of fertility. Front. Neuroendocrinol. 2010, 31, 322–340. [Google Scholar] [CrossRef] [PubMed]
  6. Coss, D. Regulation of reproduction via tight control of gonadotropin hormone levels. Mol. Cell Endocrinol. 2018, 463, 116–130. [Google Scholar] [CrossRef] [PubMed]
  7. Naor, Z. Signaling by G-protein-coupled receptor (GPCR): Studies on the GnRH receptor. Front. Neuroendocrinol. 2009, 30, 10–29. [Google Scholar] [CrossRef]
  8. Kuiri-Hanninen, T.; Sankilampi, U.; Dunkel, L. Activation of the hypothalamic-pituitary-gonadal axis in infancy: Minipuberty. Horm. Res. Paediatr. 2014, 82, 73–80. [Google Scholar] [CrossRef]
  9. Young, J.; Xu, C.; Papadakis, G.E.; Acierno, J.S.; Maione, L.; Hietamaki, J.; Raivio, T.; Pitteloud, N. Clinical Management of Congenital Hypogonadotropic Hypogonadism. Endocr. Rev. 2019, 40, 669–710. [Google Scholar] [CrossRef]
  10. Brioude, F.; Bouligand, J.; Trabado, S.; Francou, B.; Salenave, S.; Kamenicky, P.; Brailly-Tabard, S.; Chanson, P.; Guiochon-Mantel, A.; Young, J. Non-syndromic congenital hypogonadotropic hypogonadism: Clinical presentation and genotype-phenotype relationships. Eur. J. Endocrinol. 2010, 162, 835–851. [Google Scholar] [CrossRef]
  11. Morgan, K.; Conklin, D.; Pawson, A.J.; Sellar, R.; Ott, T.R.; Millar, R.P. A transcriptionally active human type II gonadotropin-releasing hormone receptor gene homolog overlaps two genes in the antisense orientation on chromosome 1q.12. Endocrinology 2003, 144, 423–436. [Google Scholar] [CrossRef]
  12. Neill, J.D. Mammalian gonadotropin-releasing hormone (GnRH) receptor subtypes. Arch. Physiol. Biochem. 2002, 110, 129–136. [Google Scholar] [CrossRef] [PubMed]
  13. Neill, J.D. GnRH and GnRH receptor genes in the human genome. Endocrinology 2002, 143, 737–743. [Google Scholar] [CrossRef] [PubMed]
  14. Stojilkovic, S.S.; Reinhart, J.; Catt, K.J. Gonadotropin-releasing hormone receptors: Structure and signal transduction pathways. Endocr. Rev. 1994, 15, 462–499. [Google Scholar] [CrossRef]
  15. Limonta, P.; Montagnani Marelli, M.; Mai, S.; Motta, M.; Martini, L.; Moretti, R.M. GnRH receptors in cancer: From cell biology to novel targeted therapeutic strategies. Endocr. Rev. 2012, 33, 784–811. [Google Scholar] [CrossRef] [PubMed]
  16. Grundker, C.; Emons, G. The Role of Gonadotropin-Releasing Hormone in Cancer Cell Proliferation and Metastasis. Front. Endocrinol. 2017, 8, 187. [Google Scholar] [CrossRef] [PubMed]
  17. Aguilar-Rojas, A.; Huerta-Reyes, M. Human gonadotropin-releasing hormone receptor-activated cellular functions and signaling pathways in extra-pituitary tissues and cancer cells (Review). Oncol. Rep. 2009, 22, 981–990. [Google Scholar] [CrossRef]
  18. Chen, Y.M.; Qi, Q.R.; Xie, Q.Z.; Yang, Y.F.; Xia, Y.; Zhou, X.D. Effect of Progestin-primed Ovarian Stimulation Protocol on Outcomes of Aged Infertile Women Who Failed to Get Pregnant in the First IVF/ ICSI Cycle: A Self-controlled Study. Curr. Med. Sci. 2018, 38, 513–518. [Google Scholar] [CrossRef]
  19. Ali, M.; Chaudhry, Z.T.; Al-Hendy, A. Successes and failures of uterine leiomyoma drug discovery. Expert Opin. Drug Discov. 2018, 13, 169–177. [Google Scholar] [CrossRef]
  20. Perricos, A.; Wenzl, R. Efficacy of elagolix in the treatment of endometriosis. Expert Opin. Pharmacother. 2017, 18, 1391–1397. [Google Scholar] [CrossRef]
  21. Schally, A.V.; Block, N.L.; Rick, F.G. Discovery of LHRH and development of LHRH analogs for prostate cancer treatment. Prostate 2017, 77, 1036–1054. [Google Scholar] [CrossRef]
  22. Finch, A.R.; Caunt, C.J.; Armstrong, S.P.; McArdle, C.A. Agonist-induced internalization and downregulation of gonadotropin-releasing hormone receptors. Am. J. Physiol. Cell Physiol. 2009, 297, C591–C600. [Google Scholar] [CrossRef] [PubMed]
  23. Engel, J.B.; Schally, A.V. Drug Insight: Clinical use of agonists and antagonists of luteinizing-hormone-releasing hormone. Nat. Clin. Pract. Endocrinol. Metab. 2007, 3, 157–167. [Google Scholar] [CrossRef]
  24. Fan, N.C.; Peng, C.; Krisinger, J.; Leung, P.C. The human gonadotropin-releasing hormone receptor gene: Complete structure including multiple promoters, transcription initiation sites, and polyadenylation signals. Mol. Cell Endocrinol. 1995, 107, R1–R8. [Google Scholar] [CrossRef] [PubMed]
  25. Kakar, S.S. Molecular structure of the human gonadotropin-releasing hormone receptor gene. Eur. J. Endocrinol. 1997, 137, 183–192. [Google Scholar] [CrossRef] [PubMed]
  26. Albarracin, C.T.; Kaiser, U.B.; Chin, W.W. Isolation and characterization of the 5’-flanking region of the mouse gonadotropin-releasing hormone receptor gene. Endocrinology 1994, 135, 2300–2306. [Google Scholar] [CrossRef]
  27. Leung, P.C.; Squire, J.; Peng, C.; Fan, N.; Hayden, M.R.; Olofsson, J.I. Mapping of the gonadotropin-releasing hormone (GnRH) receptor gene to human chromosome 4q21.2 by fluorescence in situ hybridization. Mamm. Genome 1995, 6, 309–310. [Google Scholar] [CrossRef]
  28. Grosse, R.; Schoneberg, T.; Schultz, G.; Gudermann, T. Inhibition of gonadotropin-releasing hormone receptor signaling by expression of a splice variant of the human receptor. Mol. Endocrinol. 1997, 11, 1305–1318. [Google Scholar] [CrossRef]
  29. Trzaskowski, B.; Latek, D.; Yuan, S.; Ghoshdastider, U.; Debinski, A.; Filipek, S. Action of molecular switches in GPCRs--theoretical and experimental studies. Curr. Med. Chem. 2012, 19, 1090–1109. [Google Scholar] [CrossRef]
  30. Davidson, J.S.; Assefa, D.; Pawson, A.; Davies, P.; Hapgood, J.; Becker, I.; Flanagan, C.; Roeske, R.; Millar, R. Irreversible activation of the gonadotropin-releasing hormone receptor by photoaffinity cross-linking: Localization of attachment site to Cys residue in N-terminal segment. Biochemistry 1997, 36, 12881–12889. [Google Scholar] [CrossRef]
  31. Hoffmann, S.H.; ter Laak, T.; Kuhne, R.; Reilander, H.; Beckers, T. Residues within transmembrane helices 2 and 5 of the human gonadotropin-releasing hormone receptor contribute to agonist and antagonist binding. Mol. Endocrinol. 2000, 14, 1099–1115. [Google Scholar] [CrossRef]
  32. Zhou, W.; Rodic, V.; Kitanovic, S.; Flanagan, C.A.; Chi, L.; Weinstein, H.; Maayani, S.; Millar, R.P.; Sealfon, S.C. A locus of the gonadotropin-releasing hormone receptor that differentiates agonist and antagonist binding sites. J. Biol. Chem. 1995, 270, 18853–18857. [Google Scholar] [CrossRef] [PubMed]
  33. Davidson, J.S.; McArdle, C.A.; Davies, P.; Elario, R.; Flanagan, C.A.; Millar, R.P. Asn102 of the gonadotropin-releasing hormone receptor is a critical determinant of potency for agonists containing C-terminal glycinamide. J. Biol. Chem. 1996, 271, 15510–15514. [Google Scholar] [CrossRef]
  34. Flanagan, C.A.; Rodic, V.; Konvicka, K.; Yuen, T.; Chi, L.; Rivier, J.E.; Millar, R.P.; Weinstein, H.; Sealfon, S.C. Multiple interactions of the Asp(2.61(98)) side chain of the gonadotropin-releasing hormone receptor contribute differentially to ligand interaction. Biochemistry 2000, 39, 8133–8141. [Google Scholar] [CrossRef] [PubMed]
  35. Fromme, B.J.; Katz, A.A.; Roeske, R.W.; Millar, R.P.; Flanagan, C.A. Role of aspartate7.32(302) of the human gonadotropin-releasing hormone receptor in stabilizing a high-affinity ligand conformation. Mol. Pharmacol. 2001, 60, 1280–1287. [Google Scholar] [CrossRef]
  36. Strader, C.D.; Fong, T.M.; Tota, M.R.; Underwood, D.; Dixon, R.A. Structure and function of G protein-coupled receptors. Annu. Rev. Biochem. 1994, 63, 101–132. [Google Scholar] [CrossRef] [PubMed]
  37. Palczewski, K.; Benovic, J.L. G-protein-coupled receptor kinases. Trends Biochem. Sci. 1991, 16, 387–391. [Google Scholar] [CrossRef] [PubMed]
  38. Davidson, J.S.; Wakefield, I.K.; Millar, R.P. Absence of rapid desensitization of the mouse gonadotropin-releasing hormone receptor. Biochem. J. 1994, 300 (Pt 2), 299–302. [Google Scholar] [CrossRef]
  39. McArdle, C.A.; Forrest-Owen, W.; Willars, G.; Davidson, J.; Poch, A.; Kratzmeier, M. Desensitization of gonadotropin-releasing hormone action in the gonadotrope-derived alpha T3-1 cell line. Endocrinology 1995, 136, 4864–4871. [Google Scholar] [CrossRef]
  40. Probst, W.C.; Snyder, L.A.; Schuster, D.I.; Brosius, J.; Sealfon, S.C. Sequence alignment of the G-protein coupled receptor superfamily. DNA Cell Biol. 1992, 11, 1–20. [Google Scholar] [CrossRef]
  41. Baldwin, J.M. Structure and function of receptors coupled to G proteins. Curr. Opin. Cell Biol. 1994, 6, 180–190. [Google Scholar] [CrossRef]
  42. Davidson, J.S.; Flanagan, C.A.; Becker, I.I.; Illing, N.; Sealfon, S.C.; Millar, R.P. Molecular function of the gonadotropin-releasing hormone receptor: Insights from site-directed mutagenesis. Mol. Cell Endocrinol. 1994, 100, 9–14. [Google Scholar] [CrossRef] [PubMed]
  43. Arora, K.K.; Sakai, A.; Catt, K.J. Effects of second intracellular loop mutations on signal transduction and internalization of the gonadotropin-releasing hormone receptor. J. Biol. Chem. 1995, 270, 22820–22826. [Google Scholar] [CrossRef] [PubMed]
  44. Ballesteros, J.; Kitanovic, S.; Guarnieri, F.; Davies, P.; Fromme, B.J.; Konvicka, K.; Chi, L.; Millar, R.P.; Davidson, J.S.; Weinstein, H.; et al. Functional microdomains in G-protein-coupled receptors. The conserved arginine-cage motif in the gonadotropin-releasing hormone receptor. J. Biol. Chem. 1998, 273, 10445–10453. [Google Scholar] [CrossRef] [PubMed]
  45. Kakar, S.S.; Jennes, L. Expression of gonadotropin-releasing hormone and gonadotropin-releasing hormone receptor mRNAs in various non-reproductive human tissues. Cancer Lett. 1995, 98, 57–62. [Google Scholar] [CrossRef]
  46. Liu, F.; Usui, I.; Evans, L.G.; Austin, D.A.; Mellon, P.L.; Olefsky, J.M.; Webster, N.J. Involvement of both G(q/11) and G(s) proteins in gonadotropin-releasing hormone receptor-mediated signaling in L beta T2 cells. J. Biol. Chem. 2002, 277, 32099–32108. [Google Scholar] [CrossRef]
  47. Lambert, N.A. Dissociation of heterotrimeric g proteins in cells. Sci. Signal. 2008, 1, re5. [Google Scholar] [CrossRef]
  48. Sealfon, S.C.; Weinstein, H.; Millar, R.P. Molecular mechanisms of ligand interaction with the gonadotropin-releasing hormone receptor. Endocr. Rev. 1997, 18, 180–205. [Google Scholar] [CrossRef]
  49. Hamm, H.E. How activated receptors couple to G proteins. Proc. Natl. Acad. Sci. USA 2001, 98, 4819–4821. [Google Scholar] [CrossRef]
  50. Oldham, W.M.; Hamm, H.E. Heterotrimeric G protein activation by G-protein-coupled receptors. Nat. Rev. Mol. Cell Biol. 2008, 9, 60–71. [Google Scholar] [CrossRef]
  51. Simon, M.I.; Strathmann, M.P.; Gautam, N. Diversity of G proteins in signal transduction. Science 1991, 252, 802–808. [Google Scholar] [CrossRef]
  52. Lerea, C.L.; Somers, D.E.; Hurley, J.B.; Klock, I.B.; Bunt-Milam, A.H. Identification of specific transducin alpha subunits in retinal rod and cone photoreceptors. Science 1986, 234, 77–80. [Google Scholar] [CrossRef]
  53. Grosse, R.; Schmid, A.; Schoneberg, T.; Herrlich, A.; Muhn, P.; Schultz, G.; Gudermann, T. Gonadotropin-releasing hormone receptor initiates multiple signaling pathways by exclusively coupling to G(q/11) proteins. J. Biol. Chem. 2000, 275, 9193–9200. [Google Scholar] [CrossRef]
  54. Gilman, A.G. G proteins: Transducers of receptor-generated signals. Annu. Rev. Biochem. 1987, 56, 615–649. [Google Scholar] [CrossRef]
  55. Ford, C.E.; Skiba, N.P.; Bae, H.; Daaka, Y.; Reuveny, E.; Shekter, L.R.; Rosal, R.; Weng, G.; Yang, C.S.; Iyengar, R.; et al. Molecular basis for interactions of G protein betagamma subunits with effectors. Science 1998, 280, 1271–1274. [Google Scholar] [CrossRef]
  56. Liu, F.; Austin, D.A.; Mellon, P.L.; Olefsky, J.M.; Webster, N.J. GnRH activates ERK1/2 leading to the induction of c-fos and LHbeta protein expression in LbetaT2 cells. Mol. Endocrinol. 2002, 16, 419–434. [Google Scholar] [CrossRef]
  57. Berridge, M.J.; Irvine, R.F. Inositol trisphosphate, a novel second messenger in cellular signal transduction. Nature 1984, 312, 315–321. [Google Scholar] [CrossRef]
  58. Limor, R.; Ayalon, D.; Capponi, A.M.; Childs, G.V.; Naor, Z. Cytosolic free calcium levels in cultured pituitary cells separated by centrifugal elutriation: Effect of gonadotropin-releasing hormone. Endocrinology 1987, 120, 497–503. [Google Scholar] [CrossRef]
  59. Naor, Z.; Harris, D.; Shacham, S. Mechanism of GnRH receptor signaling: Combinatorial cross-talk of Ca2+ and protein kinase C. Front. Neuroendocrinol. 1998, 19, 1–19. [Google Scholar] [CrossRef] [PubMed]
  60. Widmann, C.; Gibson, S.; Jarpe, M.B.; Johnson, G.L. Mitogen-activated protein kinase: Conservation of a three-kinase module from yeast to human. Physiol. Rev. 1999, 79, 143–180. [Google Scholar] [CrossRef] [PubMed]
  61. Kim, Y.; Han, J.M.; Park, J.B.; Lee, S.D.; Oh, Y.S.; Chung, C.; Lee, T.G.; Kim, J.H.; Park, S.K.; Yoo, J.S.; et al. Phosphorylation and activation of phospholipase D1 by protein kinase C in vivo: Determination of multiple phosphorylation sites. Biochemistry 1999, 38, 10344–10351. [Google Scholar] [CrossRef] [PubMed]
  62. Mitchell, R.; Robertson, D.N.; Holland, P.J.; Collins, D.; Lutz, E.M.; Johnson, M.S. ADP-ribosylation factor-dependent phospholipase D activation by the M3 muscarinic receptor. J. Biol. Chem. 2003, 278, 33818–33830. [Google Scholar] [CrossRef] [PubMed]
  63. Zheng, L.; Stojilkovic, S.S.; Hunyady, L.; Krsmanovic, L.Z.; Catt, K.J. Sequential activation of phospholipase-C and -D in agonist-stimulated gonadotrophs. Endocrinology 1994, 134, 1446–1454. [Google Scholar] [CrossRef] [PubMed]
  64. Seger, R.; Krebs, E.G. The MAPK signaling cascade. FASEB J. 1995, 9, 726–735. [Google Scholar] [CrossRef] [PubMed]
  65. Naor, Z.; Benard, O.; Seger, R. Activation of MAPK cascades by G-protein-coupled receptors: The case of gonadotropin-releasing hormone receptor. Trends Endocrinol. Metab. 2000, 11, 91–99. [Google Scholar] [CrossRef] [PubMed]
  66. Kraus, S.; Naor, Z.; Seger, R. Intracellular signaling pathways mediated by the gonadotropin-releasing hormone (GnRH) receptor. Arch. Med. Res. 2001, 32, 499–509. [Google Scholar] [CrossRef]
  67. Maurer, R.A.; Kim, K.E.; Schoderbek, W.E.; Roberson, M.S.; Glenn, D.J. Regulation of glycoprotein hormone alpha-subunit gene expression. Recent Prog. Horm. Res. 1999, 54, 455–484; discussion 485. [Google Scholar]
  68. Ben-Menahem, D.; Shraga-Levine, Z.; Limor, R.; Naor, Z. Arachidonic acid and lipoxygenase products stimulate gonadotropin alpha-subunit mRNA levels in pituitary alpha T3-1 cell line: Role in gonadotropin releasing hormone action. Biochemistry 1994, 33, 12795–12799. [Google Scholar] [CrossRef]
  69. Shraga-Levine, Z.; Ben-Menahem, D.; Naor, Z. Arachidonic acid and lipoxygenase products stimulate protein kinase C beta mRNA levels in pituitary alpha T3-1 cell line: Role in gonadotropin-releasing hormone action. Biochem. J. 1996, 316 (Pt 2), 667–670. [Google Scholar] [CrossRef]
  70. Haisenleder, D.J.; Dalkin, A.C.; Ortolano, G.A.; Marshall, J.C.; Shupnik, M.A. A pulsatile gonadotropin-releasing hormone stimulus is required to increase transcription of the gonadotropin subunit genes: Evidence for differential regulation of transcription by pulse frequency in vivo. Endocrinology 1991, 128, 509–517. [Google Scholar] [CrossRef]
  71. Chi, H.; Horie, H.; Hikawa, N.; Takenaka, T. Isolation and age-related characterization of mouse Schwann cells from dorsal root ganglion explants in type I collagen gels. J. Neurosci. Res. 1993, 35, 183–187. [Google Scholar] [CrossRef]
  72. Kottler, M.L.; Bergametti, F.; Carre, M.C.; Morice, S.; Decoret, E.; Lagarde, J.P.; Starzec, A.; Counis, R. Tissue-specific pattern of variant transcripts of the human gonadotropin-releasing hormone receptor gene. Eur. J. Endocrinol. 1999, 140, 561–569. [Google Scholar] [CrossRef] [PubMed]
  73. La Rosa, S.; Celato, N.; Uccella, S.; Capella, C. Detection of gonadotropin-releasing hormone receptor in normal human pituitary cells and pituitary adenomas using immunohistochemistry. Virchows Arch. 2000, 437, 264–269. [Google Scholar] [CrossRef] [PubMed]
  74. Sanno, N.; Jin, L.; Qian, X.; Osamura, R.Y.; Scheithauer, B.W.; Kovacs, K.; Lloyd, R.V. Gonadotropin-releasing hormone and gonadotropin-releasing hormone receptor messenger ribonucleic acids expression in nontumorous and neoplastic pituitaries. J. Clin. Endocrinol. Metab. 1997, 82, 1974–1982. [Google Scholar] [CrossRef] [PubMed]
  75. Emons, G.; Pahwa, G.S.; Brack, C.; Sturm, R.; Oberheuser, F.; Knuppen, R. Gonadotropin releasing hormone binding sites in human epithelial ovarian carcinomata. Eur. J. Cancer Clin. Oncol. 1989, 25, 215–221. [Google Scholar] [CrossRef] [PubMed]
  76. Emons, G.; Ortmann, O.; Becker, M.; Irmer, G.; Springer, B.; Laun, R.; Holzel, F.; Schulz, K.D.; Schally, A.V. High affinity binding and direct antiproliferative effects of LHRH analogues in human ovarian cancer cell lines. Cancer Res. 1993, 53, 5439–5446. [Google Scholar] [PubMed]
  77. Kakar, S.S.; Grizzle, W.E.; Neill, J.D. The nucleotide sequences of human GnRH receptors in breast and ovarian tumors are identical with that found in pituitary. Mol. Cell Endocrinol. 1994, 106, 145–149. [Google Scholar] [CrossRef]
  78. Brus, L.; Lambalk, C.B.; de Koning, J.; Helder, M.N.; Janssens, R.M.; Schoemaker, J. Specific gonadotrophin-releasing hormone analogue binding predominantly in human luteinized follicular aspirates and not in human pre-ovulatory follicles. Hum. Reprod 1997, 12, 769–773. [Google Scholar] [CrossRef]
  79. Chatzaki, E.; Bax, C.M.; Eidne, K.A.; Anderson, L.; Grudzinskas, J.G.; Gallagher, C.J. The expression of gonadotropin-releasing hormone and its receptor in endometrial cancer, and its relevance as an autocrine growth factor. Cancer Res. 1996, 56, 2059–2065. [Google Scholar]
  80. Borroni, R.; Di Blasio, A.M.; Gaffuri, B.; Santorsola, R.; Busacca, M.; Vigano, P.; Vignali, M. Expression of GnRH receptor gene in human ectopic endometrial cells and inhibition of their proliferation by leuprolide acetate. Mol. Cell Endocrinol. 2000, 159, 37–43. [Google Scholar] [CrossRef]
  81. Lin, L.S.; Roberts, V.J.; Yen, S.S. Expression of human gonadotropin-releasing hormone receptor gene in the placenta and its functional relationship to human chorionic gonadotropin secretion. J. Clin. Endocrinol. Metab. 1995, 80, 580–585. [Google Scholar] [CrossRef]
  82. Bahk, J.Y.; Hyun, J.S.; Lee, H.; Kim, M.O.; Cho, G.J.; Lee, B.H.; Choi, W.S. Expression of gonadotropin-releasing hormone (GnRH) and GnRH receptor mRNA in prostate cancer cells and effect of GnRH on the proliferation of prostate cancer cells. Urol. Res. 1998, 26, 259–264. [Google Scholar] [CrossRef] [PubMed]
  83. Tieva, A.; Stattin, P.; Wikstrom, P.; Bergh, A.; Damber, J.E. Gonadotropin-releasing hormone receptor expression in the human prostate. Prostate 2001, 47, 276–284. [Google Scholar] [CrossRef]
  84. Eidne, K.A.; Flanagan, C.A.; Millar, R.P. Gonadotropin-releasing hormone binding sites in human breast carcinoma. Science 1985, 229, 989–991. [Google Scholar] [CrossRef] [PubMed]
  85. Kottler, M.L.; Starzec, A.; Carre, M.C.; Lagarde, J.P.; Martin, A.; Counis, R. The genes for gonadotropin-releasing hormone and its receptor are expressed in human breast with fibrocystic disease and cancer. Int. J. Cancer 1997, 71, 595–599. [Google Scholar] [CrossRef]
  86. Moriya, T.; Suzuki, T.; Pilichowska, M.; Ariga, N.; Kimura, N.; Ouchi, N.; Nagura, H.; Sasano, H. Immunohistochemical expression of gonadotropin releasing hormone receptor in human breast carcinoma. Pathol. Int. 2001, 51, 333–337. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, H.F.; Jeung, E.B.; Stephenson, M.; Leung, P.C. Human peripheral blood mononuclear cells express gonadotropin-releasing hormone (GnRH), GnRH receptor, and interleukin-2 receptor gamma-chain messenger ribonucleic acids that are regulated by GnRH in vitro. J. Clin. Endocrinol. Metab. 1999, 84, 743–750. [Google Scholar] [CrossRef]
  88. Moretti, R.M.; Montagnani Marelli, M.; Van Groeninghen, J.C.; Limonta, P. Locally expressed LHRH receptors mediate the oncostatic and antimetastatic activity of LHRH agonists on melanoma cells. J. Clin. Endocrinol. Metab. 2002, 87, 3791–3797. [Google Scholar] [CrossRef]
  89. de Roux, N.; Young, J.; Misrahi, M.; Genet, R.; Chanson, P.; Schaison, G.; Milgrom, E. A family with hypogonadotropic hypogonadism and mutations in the gonadotropin-releasing hormone receptor. N. Engl. J. Med. 1997, 337, 1597–1602. [Google Scholar] [CrossRef]
  90. Goncalves, C.I.; Aragues, J.M.; Bastos, M.; Barros, L.; Vicente, N.; Carvalho, D.; Lemos, M.C. GNRHR biallelic and digenic mutations in patients with normosmic congenital hypogonadotropic hypogonadism. Endocr. Connect 2017, 6, 360–366. [Google Scholar] [CrossRef]
  91. Pitteloud, N.; Quinton, R.; Pearce, S.; Raivio, T.; Acierno, J.; Dwyer, A.; Plummer, L.; Hughes, V.; Seminara, S.; Cheng, Y.Z.; et al. Digenic mutations account for variable phenotypes in idiopathic hypogonadotropic hypogonadism. J. Clin. Investig. 2007, 117, 457–463. [Google Scholar] [CrossRef]
  92. Mendez, J.P.; Zenteno, J.C.; Coronel, A.; Soriano-Ursua, M.A.; Valencia-Villalvazo, E.Y.; Soderlund, D.; Coral-Vazquez, R.M.; Canto, P. Triallelic digenic mutation in the prokineticin 2 and GNRH receptor genes in two brothers with normosmic congenital hypogonadotropic hypogonadism. Endocr. Res. 2015, 40, 166–171. [Google Scholar] [CrossRef] [PubMed]
  93. Neocleous, V.; Fanis, P.; Toumba, M.; Tanteles, G.A.; Schiza, M.; Cinarli, F.; Nicolaides, N.C.; Oulas, A.; Spyrou, G.M.; Mantzoros, C.S.; et al. GnRH Deficient Patients With Congenital Hypogonadotropic Hypogonadism: Novel Genetic Findings in ANOS1, RNF216, WDR11, FGFR1, CHD7, and POLR3A Genes in a Case Series and Review of the Literature. Front. Endocrinol. 2020, 11, 626. [Google Scholar] [CrossRef] [PubMed]
  94. Bianco, S.D.; Kaiser, U.B. The genetic and molecular basis of idiopathic hypogonadotropic hypogonadism. Nat. Rev. Endocrinol. 2009, 5, 569–576. [Google Scholar] [CrossRef] [PubMed]
  95. Gurbuz, F.; Kotan, L.D.; Mengen, E.; Siklar, Z.; Berberoglu, M.; Dokmetas, S.; Kilicli, M.F.; Guven, A.; Kirel, B.; Saka, N.; et al. Distribution of gene mutations associated with familial normosmic idiopathic hypogonadotropic hypogonadism. J. Clin. Res. Pediatr. Endocrinol. 2012, 4, 121–126. [Google Scholar] [CrossRef]
  96. Kottler, M.L.; Chauvin, S.; Lahlou, N.; Harris, C.E.; Johnston, C.J.; Lagarde, J.P.; Bouchard, P.; Farid, N.R.; Counis, R. A new compound heterozygous mutation of the gonadotropin-releasing hormone receptor (L314X, Q106R) in a woman with complete hypogonadotropic hypogonadism: Chronic estrogen administration amplifies the gonadotropin defect. J. Clin. Endocrinol. Metab. 2000, 85, 3002–3008. [Google Scholar] [CrossRef]
  97. Meysing, A.U.; Kanasaki, H.; Bedecarrats, G.Y.; Acierno, J.S., Jr.; Conn, P.M.; Martin, K.A.; Seminara, S.B.; Hall, J.E.; Crowley, W.F., Jr.; Kaiser, U.B. GNRHR mutations in a woman with idiopathic hypogonadotropic hypogonadism highlight the differential sensitivity of luteinizing hormone and follicle-stimulating hormone to gonadotropin-releasing hormone. J. Clin. Endocrinol. Metab. 2004, 89, 3189–3198. [Google Scholar] [CrossRef]
  98. Tello, J.A.; Newton, C.L.; Bouligand, J.; Guiochon-Mantel, A.; Millar, R.P.; Young, J. Congenital hypogonadotropic hypogonadism due to GnRH receptor mutations in three brothers reveal sites affecting conformation and coupling. PLoS ONE 2012, 7, e38456. [Google Scholar] [CrossRef]
  99. Vaaralahti, K.; Wehkalampi, K.; Tommiska, J.; Laitinen, E.M.; Dunkel, L.; Raivio, T. The role of gene defects underlying isolated hypogonadotropic hypogonadism in patients with constitutional delay of growth and puberty. Fertil. Steril. 2011, 95, 2756–2758. [Google Scholar] [CrossRef]
  100. Francou, B.; Bouligand, J.; Voican, A.; Amazit, L.; Trabado, S.; Fagart, J.; Meduri, G.; Brailly-Tabard, S.; Chanson, P.; Lecomte, P.; et al. Normosmic congenital hypogonadotropic hypogonadism due to TAC3/TACR3 mutations: Characterization of neuroendocrine phenotypes and novel mutations. PLoS ONE 2011, 6, e25614. [Google Scholar] [CrossRef]
  101. Layman, L.C.; Cohen, D.P.; Jin, M.; Xie, J.; Li, Z.; Reindollar, R.H.; Bolbolan, S.; Bick, D.P.; Sherins, R.R.; Duck, L.W.; et al. Mutations in gonadotropin-releasing hormone receptor gene cause hypogonadotropic hypogonadism. Nat. Genet. 1998, 18, 14–15. [Google Scholar] [CrossRef]
  102. Beneduzzi, D.; Trarbach, E.B.; Min, L.; Jorge, A.A.; Garmes, H.M.; Renk, A.C.; Fichna, M.; Fichna, P.; Arantes, K.A.; Costa, E.M.; et al. Role of gonadotropin-releasing hormone receptor mutations in patients with a wide spectrum of pubertal delay. Fertil. Steril. 2014, 102, 838–846.e832. [Google Scholar] [CrossRef] [PubMed]
  103. Gianetti, E.; Hall, J.E.; Au, M.G.; Kaiser, U.B.; Quinton, R.; Stewart, J.A.; Metzger, D.L.; Pitteloud, N.; Mericq, V.; Merino, P.M.; et al. When genetic load does not correlate with phenotypic spectrum: Lessons from the GnRH receptor (GNRHR). J. Clin. Endocrinol. Metab. 2012, 97, E1798–E1807. [Google Scholar] [CrossRef] [PubMed]
  104. Beranova, M.; Oliveira, L.M.; Bedecarrats, G.Y.; Schipani, E.; Vallejo, M.; Ammini, A.C.; Quintos, J.B.; Hall, J.E.; Martin, K.A.; Hayes, F.J.; et al. Prevalence, phenotypic spectrum, and modes of inheritance of gonadotropin-releasing hormone receptor mutations in idiopathic hypogonadotropic hypogonadism. J. Clin. Endocrinol. Metab. 2001, 86, 1580–1588. [Google Scholar] [CrossRef] [PubMed]
  105. Zernov, N.; Skoblov, M.; Baranova, A.; Boyarsky, K. Mutations in gonadotropin-releasing hormone signaling pathway in two nIHH patients with successful pregnancy outcomes. Reprod. Biol. Endocrinol. 2016, 14, 48. [Google Scholar] [CrossRef]
  106. Nair, S.; Jadhav, S.; Lila, A.; Jagtap, V.; Bukan, A.; Pandit, R.; Ekbote, A.; Dharmalingam, M.; Kumar, P.; Kalra, P.; et al. Spectrum of phenotype and genotype of congenital isolated hypogonadotropic hypogonadism in Asian Indians. Clin. Endocrinol. 2016, 85, 100–109. [Google Scholar] [CrossRef]
  107. Cassatella, D.; Howard, S.R.; Acierno, J.S.; Xu, C.; Papadakis, G.E.; Santoni, F.A.; Dwyer, A.A.; Santini, S.; Sykiotis, G.P.; Chambion, C.; et al. Congenital hypogonadotropic hypogonadism and constitutional delay of growth and puberty have distinct genetic architectures. Eur. J. Endocrinol. 2018, 178, 377–388. [Google Scholar] [CrossRef]
  108. Sarfati, J.; Guiochon-Mantel, A.; Rondard, P.; Arnulf, I.; Garcia-Pinero, A.; Wolczynski, S.; Brailly-Tabard, S.; Bidet, M.; Ramos-Arroyo, M.; Mathieu, M.; et al. A comparative phenotypic study of kallmann syndrome patients carrying monoallelic and biallelic mutations in the prokineticin 2 or prokineticin receptor 2 genes. J. Clin. Endocrinol. Metab. 2010, 95, 659–669. [Google Scholar] [CrossRef]
  109. Quaynor, S.D.; Bosley, M.E.; Duckworth, C.G.; Porter, K.R.; Kim, S.H.; Kim, H.G.; Chorich, L.P.; Sullivan, M.E.; Choi, J.H.; Cameron, R.S.; et al. Targeted next generation sequencing approach identifies eighteen new candidate genes in normosmic hypogonadotropic hypogonadism and Kallmann syndrome. Mol. Cell Endocrinol. 2016, 437, 86–96. [Google Scholar] [CrossRef]
  110. de Roux, N.; Young, J.; Brailly-Tabard, S.; Misrahi, M.; Milgrom, E.; Schaison, G. The same molecular defects of the gonadotropin-releasing hormone receptor determine a variable degree of hypogonadism in affected kindred. J. Clin. Endocrinol. Metab. 1999, 84, 567–572. [Google Scholar] [CrossRef]
  111. Silveira, L.F.; Stewart, P.M.; Thomas, M.; Clark, D.A.; Bouloux, P.M.; MacColl, G.S. Novel homozygous splice acceptor site GnRH receptor (GnRHR) mutation: Human GnRHR “knockout”. J. Clin. Endocrinol. Metab. 2002, 87, 2973–2977. [Google Scholar] [CrossRef]
  112. Fan, Y.; Zhang, X.; Wang, L.; Wang, R.; Huang, Z.; Sun, Y.; Yao, R.; Huang, X.; Ye, J.; Han, L.; et al. Diagnostic Application of Targeted Next-Generation Sequencing of 80 Genes Associated with Disorders of Sexual Development. Sci. Rep. 2017, 7, 44536. [Google Scholar] [CrossRef] [PubMed]
  113. Karges, B.; Karges, W.; Mine, M.; Ludwig, L.; Kuhne, R.; Milgrom, E.; de Roux, N. Mutation Ala(171)Thr stabilizes the gonadotropin-releasing hormone receptor in its inactive conformation, causing familial hypogonadotropic hypogonadism. J. Clin. Endocrinol. Metab. 2003, 88, 1873–1879. [Google Scholar] [CrossRef] [PubMed]
  114. Pralong, F.P.; Gomez, F.; Castillo, E.; Cotecchia, S.; Abuin, L.; Aubert, M.L.; Portmann, L.; Gaillard, R.C. Complete hypogonadotropic hypogonadism associated with a novel inactivating mutation of the gonadotropin-releasing hormone receptor. J. Clin. Endocrinol. Metab. 1999, 84, 3811–3816. [Google Scholar] [CrossRef] [PubMed]
  115. Marcos, S.; Sarfati, J.; Leroy, C.; Fouveaut, C.; Parent, P.; Metz, C.; Wolczynski, S.; Gerard, M.; Bieth, E.; Kurtz, F.; et al. The prevalence of CHD7 missense versus truncating mutations is higher in patients with Kallmann syndrome than in typical CHARGE patients. J. Clin. Endocrinol. Metab. 2014, 99, E2138–E2143. [Google Scholar] [CrossRef] [PubMed]
  116. Vagenakis, G.A.; Sgourou, A.; Papachatzopoulou, A.; Kourounis, G.; Papavassiliou, A.G.; Georgopoulos, N.A. The gonadotropin-releasing hormone (GnRH)-1 gene, the GnRH receptor gene, and their promoters in patients with idiopathic hypogonadotropic hypogonadism with or without resistance to GnRH action. Fertil. Steril. 2005, 84, 1762–1765. [Google Scholar] [CrossRef]
  117. Costa, E.M.; Bedecarrats, G.Y.; Mendonca, B.B.; Arnhold, I.J.; Kaiser, U.B.; Latronico, A.C. Two novel mutations in the gonadotropin-releasing hormone receptor gene in Brazilian patients with hypogonadotropic hypogonadism and normal olfaction. J. Clin. Endocrinol. Metab. 2001, 86, 2680–2686. [Google Scholar] [CrossRef]
  118. Topaloglu, A.K.; Lu, Z.L.; Farooqi, I.S.; Mungan, N.O.; Yuksel, B.; O’Rahilly, S.; Millar, R.P. Molecular genetic analysis of normosmic hypogonadotropic hypogonadism in a Turkish population: Identification and detailed functional characterization of a novel mutation in the gonadotropin-releasing hormone receptor gene. Neuroendocrinology 2006, 84, 301–308. [Google Scholar] [CrossRef] [PubMed]
  119. Caron, P.; Chauvin, S.; Christin-Maitre, S.; Bennet, A.; Lahlou, N.; Counis, R.; Bouchard, P.; Kottler, M.L. Resistance of hypogonadic patients with mutated GnRH receptor genes to pulsatile GnRH administration. J. Clin. Endocrinol. Metab. 1999, 84, 990–996. [Google Scholar] [CrossRef]
  120. Zhang, R.; Linpeng, S.; Li, Z.; Cao, Y.; Tan, H.; Liang, D.; Wu, L. Deficiency in GnRH receptor trafficking due to a novel homozygous mutation causes idiopathic hypogonadotropic hypogonadism in three prepubertal siblings. Gene 2018, 669, 42–46. [Google Scholar] [CrossRef]
  121. Antelli, A.; Baldazzi, L.; Balsamo, A.; Pirazzoli, P.; Nicoletti, A.; Gennari, M.; Cicognani, A. Two novel GnRHR gene mutations in two siblings with hypogonadotropic hypogonadism. Eur. J. Endocrinol. 2006, 155, 201–205. [Google Scholar] [CrossRef]
  122. Cioppi, F.; Riera-Escamilla, A.; Manilall, A.; Guarducci, E.; Todisco, T.; Corona, G.; Colombo, F.; Bonomi, M.; Flanagan, C.A.; Krausz, C. Genetics of ncHH: From a peculiar inheritance of a novel GNRHR mutation to a comprehensive review of the literature. Andrology 2019, 7, 88–101. [Google Scholar] [CrossRef]
  123. Francou, B.; Paul, C.; Amazit, L.; Cartes, A.; Bouvattier, C.; Albarel, F.; Maiter, D.; Chanson, P.; Trabado, S.; Brailly-Tabard, S.; et al. Prevalence of KISS1 Receptor mutations in a series of 603 patients with normosmic congenital hypogonadotrophic hypogonadism and characterization of novel mutations: A single-centre study. Hum. Reprod. 2016, 31, 1363–1374. [Google Scholar] [CrossRef] [PubMed]
  124. Quaynor, S.D.; Kim, H.G.; Cappello, E.M.; Williams, T.; Chorich, L.P.; Bick, D.P.; Sherins, R.J.; Layman, L.C. The prevalence of digenic mutations in patients with normosmic hypogonadotropic hypogonadism and Kallmann syndrome. Fertil. Steril. 2011, 96, 1424–1430.e1426. [Google Scholar] [CrossRef] [PubMed]
  125. Soderlund, D.; Canto, P.; de la Chesnaye, E.; Ulloa-Aguirre, A.; Mendez, J.P. A novel homozygous mutation in the second transmembrane domain of the gonadotrophin releasing hormone receptor gene. Clin. Endocrinol. 2001, 54, 493–498. [Google Scholar] [CrossRef]
  126. Raivio, T.; Sidis, Y.; Plummer, L.; Chen, H.; Ma, J.; Mukherjee, A.; Jacobson-Dickman, E.; Quinton, R.; Van Vliet, G.; Lavoie, H.; et al. Impaired fibroblast growth factor receptor 1 signaling as a cause of normosmic idiopathic hypogonadotropic hypogonadism. J. Clin. Endocrinol. Metab. 2009, 94, 4380–4390. [Google Scholar] [CrossRef]
  127. Hussain, H.M.J.; Murtaza, G.; Jiang, X.; Khan, R.; Khan, M.; Kakakhel, M.B.S.; Khan, T.; Wahab, F.; Zhang, H.; Zhang, Y.; et al. Whole Exome Sequencing Revealed a Novel Nonsense Variant in the GNRHR Gene Causing Normosmic Hypogonadotropic Hypogonadism in a Pakistani Family. Horm. Res. Paediatr. 2019, 91, 9–16. [Google Scholar] [CrossRef] [PubMed]
  128. Shaw, N.D.; Seminara, S.B.; Welt, C.K.; Au, M.G.; Plummer, L.; Hughes, V.A.; Dwyer, A.A.; Martin, K.A.; Quinton, R.; Mericq, V.; et al. Expanding the phenotype and genotype of female GnRH deficiency. J. Clin. Endocrinol. Metab. 2011, 96, E566–E576. [Google Scholar] [CrossRef]
  129. Basaran, Y.; Bolu, E.; Unal, H.U.; Sagkan, R.I.; Taslipinar, A.; Ozgurtas, T.; Musabak, U. Multiplex ligation dependent probe amplification analysis of KAL1, GNRH1, GNRHR, PROK2 and PROKR2 in male patients with idiopathic hypogonadotropic hypogonadism. Endokrynol. Pol. 2013, 64, 285–292. [Google Scholar] [CrossRef]
  130. Kim, H.G.; Pedersen-White, J.; Bhagavath, B.; Layman, L.C. Genotype and phenotype of patients with gonadotropin-releasing hormone receptor mutations. Front. Horm. Res. 2010, 39, 94–110. [Google Scholar] [CrossRef]
  131. Choi, J.H.; Balasubramanian, R.; Lee, P.H.; Shaw, N.D.; Hall, J.E.; Plummer, L.; Buck, C.L.; Kottler, M.L.; Jarzabek, K.; Wolczynski, S.; et al. Expanding the Spectrum of Founder Mutations Causing Isolated Gonadotropin-Releasing Hormone Deficiency. J. Clin. Endocrinol. Metab. 2015, 100, E1378–E1385. [Google Scholar] [CrossRef]
  132. Ortmann, O.; Weiss, J.M.; Diedrich, K. Gonadotrophin-releasing hormone (GnRH) and GnRH agonists: Mechanisms of action. Reprod. Biomed. Online 2002, 5 (Suppl. S1), 1–7. [Google Scholar] [CrossRef] [PubMed]
  133. Ortmann, O.; Diedrich, K. Pituitary and extrapituitary actions of gonadotrophin-releasing hormone and its analogues. Hum. Reprod. 1999, 14 (Suppl. S1), 194–206. [Google Scholar] [CrossRef] [PubMed]
  134. Conn, P.M.; Ulloa-Aguirre, A. Pharmacological chaperones for misfolded gonadotropin-releasing hormone receptors. Adv. Pharmacol. 2011, 62, 109–141. [Google Scholar] [CrossRef] [PubMed]
  135. Kumar, P.; Sharma, A. Gonadotropin-releasing hormone analogs: Understanding advantages and limitations. J. Hum. Reprod. Sci. 2014, 7, 170–174. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, J.H.; Choi, Y.S. The role of gonadotropin-releasing hormone agonists in female fertility preservation. Clin. Exp. Reprod. Med. 2021, 48, 11–26. [Google Scholar] [CrossRef] [PubMed]
  137. Resta, C.; Moustogiannis, A.; Chatzinikita, E.; Malligiannis Ntalianis, D.; Malligiannis Ntalianis, K.; Philippou, A.; Koutsilieris, M.; Vlahos, N. Gonadotropin-Releasing Hormone (GnRH)/GnRH Receptors and Their Role in the Treatment of Endometriosis. Cureus 2023, 15, e38136. [Google Scholar] [CrossRef] [PubMed]
  138. Golan, A. GnRH analogues in the treatment of uterine fibroids. Hum. Reprod. 1996, 11 (Suppl. S3), 33–41. [Google Scholar] [CrossRef]
  139. Eugster, E.A. Treatment of Central Precocious Puberty. J. Endocr. Soc. 2019, 3, 965–972. [Google Scholar] [CrossRef]
  140. Huerta-Reyes, M.; Maya-Nunez, G.; Perez-Solis, M.A.; Lopez-Munoz, E.; Guillen, N.; Olivo-Marin, J.C.; Aguilar-Rojas, A. Treatment of Breast Cancer With Gonadotropin-Releasing Hormone Analogs. Front. Oncol. 2019, 9, 943. [Google Scholar] [CrossRef]
  141. Choi, S.; Lee, A.K. Efficacy and safety of gonadotropin-releasing hormone agonists used in the treatment of prostate cancer. Drug Healthc. Patient Saf. 2011, 3, 107–119. [Google Scholar] [CrossRef]
  142. Surrey, E.S. GnRH agonists in the treatment of symptomatic endometriosis: A review. FS Rep. 2023, 4, 40–45. [Google Scholar] [CrossRef]
  143. Van Poppel, H.; Klotz, L. Gonadotropin-releasing hormone: An update review of the antagonists versus agonists. Int. J. Urol. 2012, 19, 594–601. [Google Scholar] [CrossRef]
  144. Weiss, J.M.; Diedrich, K.; Ludwig, M. Gonadotropin-releasing hormone antagonists: Pharmacology and clinical use in women. Treat. Endocrinol. 2002, 1, 281–291. [Google Scholar] [CrossRef]
  145. Weiss, J.M.; Ludwig, M.; Ortmann, O.; Diedrich, K. GnRH antagonists in the treatment of infertility. Ann. Med. 2003, 35, 512–522. [Google Scholar] [CrossRef]
  146. Janovick, J.A.; Patny, A.; Mosley, R.; Goulet, M.T.; Altman, M.D.; Rush, T.S., 3rd; Cornea, A.; Conn, P.M. Molecular mechanism of action of pharmacoperone rescue of misrouted GPCR mutants: The GnRH receptor. Mol. Endocrinol. 2009, 23, 157–168. [Google Scholar] [CrossRef] [PubMed]
  147. Smith, E.; Janovick, J.A.; Bannister, T.D.; Shumate, J.; Ganapathy, V.; Scampavia, L.; Spicer, T.P. Rescue of mutant gonadotropin-releasing hormone receptor function independent of cognate receptor activity. Sci. Rep. 2020, 10, 10579. [Google Scholar] [CrossRef] [PubMed]
  148. Janovick, J.A.; Pogozheva, I.D.; Mosberg, H.I.; Cornea, A.; Conn, P.M. Rescue of misrouted GnRHR mutants reveals its constitutive activity. Mol. Endocrinol. 2012, 26, 1179–1188. [Google Scholar] [CrossRef] [PubMed]
  149. Pitteloud, N.; Boepple, P.A.; DeCruz, S.; Valkenburgh, S.B.; Crowley, W.F., Jr.; Hayes, F.J. The fertile eunuch variant of idiopathic hypogonadotropic hypogonadism: Spontaneous reversal associated with a homozygous mutation in the gonadotropin-releasing hormone receptor. J. Clin. Endocrinol. Metab. 2001, 86, 2470–2475. [Google Scholar] [CrossRef] [PubMed]
  150. Seminara, S.B.; Beranova, M.; Oliveira, L.M.; Martin, K.A.; Crowley, W.F., Jr.; Hall, J.E. Successful use of pulsatile gonadotropin-releasing hormone (GnRH) for ovulation induction and pregnancy in a patient with GnRH receptor mutations. J. Clin. Endocrinol. Metab. 2000, 85, 556–562. [Google Scholar] [CrossRef] [PubMed]
  151. Finkelstein, J.S.; Whitcomb, R.W.; O’Dea, L.S.; Longcope, C.; Schoenfeld, D.A.; Crowley, W.F., Jr. Sex steroid control of gonadotropin secretion in the human male. I. Effects of testosterone administration in normal and gonadotropin-releasing hormone-deficient men. J. Clin. Endocrinol. Metab. 1991, 73, 609–620. [Google Scholar] [CrossRef] [PubMed]
  152. Schwarting, G.A.; Wierman, M.E.; Tobet, S.A. Gonadotropin-releasing hormone neuronal migration. Semin. Reprod. Med. 2007, 25, 305–312. [Google Scholar] [CrossRef] [PubMed]
  153. Ferris, H.A.; Shupnik, M.A. Mechanisms for pulsatile regulation of the gonadotropin subunit genes by GNRH1. Biol. Reprod. 2006, 74, 993–998. [Google Scholar] [CrossRef] [PubMed]
  154. Matsumoto, A.M.; Karpas, A.E.; Bremner, W.J. Chronic human chorionic gonadotropin administration in normal men: Evidence that follicle-stimulating hormone is necessary for the maintenance of quantitatively normal spermatogenesis in man. J. Clin. Endocrinol. Metab. 1986, 62, 1184–1192. [Google Scholar] [CrossRef]
  155. Layman, L.C.; Cohen, D.P.; Xie, J.; Smith, G.D. Clinical phenotype and infertility treatment in a male with hypogonadotropic hypogonadism due to mutations Ala129Asp/Arg262Gln of the gonadotropin-releasing hormone receptor. Fertil. Steril. 2002, 78, 1317–1320. [Google Scholar] [CrossRef] [PubMed]
  156. Dewailly, D.; Boucher, A.; Decanter, C.; Lagarde, J.P.; Counis, R.; Kottler, M.L. Spontaneous pregnancy in a patient who was homozygous for the Q106R mutation in the gonadotropin-releasing hormone receptor gene. Fertil. Steril. 2002, 77, 1288–1291. [Google Scholar] [CrossRef] [PubMed]
  157. Layman, L.C. Hypogonadotropic hypogonadism. Endocrinol. Metab. Clin. North. Am. 2007, 36, 283–296. [Google Scholar] [CrossRef]
  158. Bouligand, J.; Ghervan, C.; Tello, J.A.; Brailly-Tabard, S.; Salenave, S.; Chanson, P.; Lombes, M.; Millar, R.P.; Guiochon-Mantel, A.; Young, J. Isolated familial hypogonadotropic hypogonadism and a GNRH1 mutation. N. Engl. J. Med. 2009, 360, 2742–2748. [Google Scholar] [CrossRef]
  159. Fraietta, R.; Zylberstejn, D.S.; Esteves, S.C. Hypogonadotropic hypogonadism revisited. Clinics 2013, 68 (Suppl. S1), 81–88. [Google Scholar] [CrossRef]
  160. Karges, B.; Karges, W.; de Roux, N. Clinical and molecular genetics of the human GnRH receptor. Hum. Reprod. Update 2003, 9, 523–530. [Google Scholar] [CrossRef]
  161. Han, T.S.; Bouloux, P.M. What is the optimal therapy for young males with hypogonadotropic hypogonadism? Clin. Endocrinol. 2010, 72, 731–737. [Google Scholar] [CrossRef]
  162. Burris, A.S.; Rodbard, H.W.; Winters, S.J.; Sherins, R.J. Gonadotropin therapy in men with isolated hypogonadotropic hypogonadism: The response to human chorionic gonadotropin is predicted by initial testicular size. J. Clin. Endocrinol. Metab. 1988, 66, 1144–1151. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Gene and protein structure of GnRHR. (A) Schematic representation of the GNRHR gene and protein. Exon/Intron organization on the gene and the amino-terminal (NH2) tail, transmembrane (TM), intracellular (IL), extracellular (EL), and carboxy-terminal (COOH) domains on protein are indicated. The three exons of gene are indicated with numbers 1–3. (B) The GnRHR structural organization in the pituitary gonadotrope cell membrane. Ligand-binding amino acid residues are indicated with orange color, G protein coupling amino acid residues are indicate with purple color, the highly conserved Asp-Arg residues are indicated with green color, and the Ser140 which is different compare to other GPCRs, is indicated with blue color. The seven transmembrane domains of the protein are indicated with numbers 1–7.
Figure 1. Gene and protein structure of GnRHR. (A) Schematic representation of the GNRHR gene and protein. Exon/Intron organization on the gene and the amino-terminal (NH2) tail, transmembrane (TM), intracellular (IL), extracellular (EL), and carboxy-terminal (COOH) domains on protein are indicated. The three exons of gene are indicated with numbers 1–3. (B) The GnRHR structural organization in the pituitary gonadotrope cell membrane. Ligand-binding amino acid residues are indicated with orange color, G protein coupling amino acid residues are indicate with purple color, the highly conserved Asp-Arg residues are indicated with green color, and the Ser140 which is different compare to other GPCRs, is indicated with blue color. The seven transmembrane domains of the protein are indicated with numbers 1–7.
Ijms 24 15965 g001
Figure 2. Signal transduction pathways of GnRHR. Schematic illustration of GnRHR activation by GnRH ligand binding to the pituitary gonadotrope cell leading to the expression of the gonadotropin genes, LH/FSH. When GnRH binds to its receptor (GnRHR), structural changes occur in both the receptor and the heterotrimeric GTP-binding proteins (G proteins), specifically the Gαq/11 subunit. This leads to a shift in the affinity of G protein for GTP over GDP, causing the Gα subunit to detach from the Gβγ dimer. The Gαq/11 subunit primarily triggers the phospholipase Cβ (PLCβ) pathway, leading to the hydrolysis of phosphatidylinositol 4,5 bisphosphate (PIP2) to inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 induces Ca2+ release from the endoplasmic reticulum, whereas DAG, along with Ca2+, activates protein kinase C (PKC). In addition, GnRH binding to the GnRHR causes delayed activation of phospholipase D (PLD) that hydrolyzes the membrane phosphatidylcholine (PC), generating phosphatidylethanol (PET) and phosphatidic acid (PA) that cause DAG production, resulting in prolonged PKC activation. PKC then triggers the activation of various kinases, including Raf-1, src, and mitogen-activated protein kinases (MAPKs), which ultimately activate transcription factors such as Elk-1, Egr-1, c-Fos and c-Jun, leading to the expression of genes involved in gonadotropin production. The pathway also involves the activation of phospholipase A2 (PLA2) that hydrolyses PC to produce arachidonic acid (AA) which is converted to prostaglandins (PGs), thromboxanes (Txs), and leukotrienes (LTs) with the latter playing a role in gene activation and gonadotropin production.
Figure 2. Signal transduction pathways of GnRHR. Schematic illustration of GnRHR activation by GnRH ligand binding to the pituitary gonadotrope cell leading to the expression of the gonadotropin genes, LH/FSH. When GnRH binds to its receptor (GnRHR), structural changes occur in both the receptor and the heterotrimeric GTP-binding proteins (G proteins), specifically the Gαq/11 subunit. This leads to a shift in the affinity of G protein for GTP over GDP, causing the Gα subunit to detach from the Gβγ dimer. The Gαq/11 subunit primarily triggers the phospholipase Cβ (PLCβ) pathway, leading to the hydrolysis of phosphatidylinositol 4,5 bisphosphate (PIP2) to inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 induces Ca2+ release from the endoplasmic reticulum, whereas DAG, along with Ca2+, activates protein kinase C (PKC). In addition, GnRH binding to the GnRHR causes delayed activation of phospholipase D (PLD) that hydrolyzes the membrane phosphatidylcholine (PC), generating phosphatidylethanol (PET) and phosphatidic acid (PA) that cause DAG production, resulting in prolonged PKC activation. PKC then triggers the activation of various kinases, including Raf-1, src, and mitogen-activated protein kinases (MAPKs), which ultimately activate transcription factors such as Elk-1, Egr-1, c-Fos and c-Jun, leading to the expression of genes involved in gonadotropin production. The pathway also involves the activation of phospholipase A2 (PLA2) that hydrolyses PC to produce arachidonic acid (AA) which is converted to prostaglandins (PGs), thromboxanes (Txs), and leukotrienes (LTs) with the latter playing a role in gene activation and gonadotropin production.
Ijms 24 15965 g002
Figure 3. Disease-associated GnRHR mutations. Schematic illustration of the GnRHR protein with indication of the position of each mutation in the amino-terminal (NH2) tail, transmembrane (TM), intracellular (IL), extracellular (EL), and carboxy-terminal (COOH) domains.
Figure 3. Disease-associated GnRHR mutations. Schematic illustration of the GnRHR protein with indication of the position of each mutation in the amino-terminal (NH2) tail, transmembrane (TM), intracellular (IL), extracellular (EL), and carboxy-terminal (COOH) domains.
Ijms 24 15965 g003
Table 1. Disease-associated mutations found in GnRHR.
Table 1. Disease-associated mutations found in GnRHR.
cDNA (NM_000406.3)Protein (NP_000397.1)Molecular ConsequenceRegion
Affected
PhenotypeClassificationReference
c.2T>Cp.Met1ThrMissenseNH2 tailHypogonadotropic hypogonadismPathogenic[103]
c.30T>Ap.Asn10LysMissenseNH2 tailHypogonadotropic hypogonadismLikely Pathogenic[117]
c.30_31delinsAAp.Asn10_Gln11delinsLysLysMissenseNH2 tailHypogonadotropic hypogonadismPathogenic[117]
c.31C>Ap.Gln11LysMissenseNH2 tailHypogonadotropic hypogonadism.Likely Pathogenic[97]
c.32delAp.Gln11fsX23FrameshiftNH2 tailHypogonadotropic hypogonadismPathogenic[103]
c.35delAp.Asn12Ilefs*12FrameshiftNH2 tailHypogonadotropic hypogonadismPathogenic[128]
c.53A>Gp.Asn18SerMissenseNH2 tailHypogonadotropic hypogonadismLikely Pathogenic[115]
c.94A>Gp.Thr32AlaMissenseNH2 tailHypogonadotropic hypogonadismPathogenic[106]
c.95 C>Tp.Thr32IleMissenseNH2 tailHypogonadotropic hypogonadismPathogenic/Likely Pathogenic[103]
c.110T>Gp.Ile37SerMissenseNH2 tailHypogonadotropic hypogonadismPathogenic[115]
c.112C>Tp.Arg38*nonsenseNH2 tailHypogonadotropic hypogonadismPathogenic[127]
c.113_114insGp.Arg38Argfs*15FrameshiftNH2 tailhypogonadotropic hypogonadismPathogenic[115]
c.247C>Tp.Leu83ValMissenseTM2Hypogonadotropic hypogonadismPathogenic[126]
c.266T>Ap.Leu89*nonsenseTM2Hypogonadotropic hypogonadismPathogenic[107]
c.268G>Ap.Glu90LysMissenseTM2Hypogonadotropic hypogonadismPathogenic/Likely Pathogenic[125]
c.270G>Cp.Glu90AspMissenseTM2Hypogonadotropic hypogonadismLikely Pathogenic[115]
c.275T>Cp.Leu92ProMissenseTM2Hypogonadotropic hypogonadismLikely Pathogenic[124]
c.281T>Cp.Val94AlaMissenseTM2Hypogonadotropic hypogonadismLikely Pathogenic[123]
c.286C>Tp.Pro96SerMissenseTM2Hypogonadotropic hypogonadismLikely Pathogenic[103]
c.296G>Ap.Gly99GluMissenseTM2Hypogonadotropic hypogonadismLikely Pathogenic[122]
c.311C>Tp.Thr104IleMissenseEL1Hypogonadotropic hypogonadismLikely Pathogenic[121]
c.317A>Gp.Gln106ArgMissenseEL1Hypogonadotropic hypogonadismPathogenic/Likely Pathogenic[89]
c.323 A>Gp.Tyr108CysMissenseEL1Hypogonadotropic hypogonadismLikely Pathogenic[121]
c.350T>Cp.Leu117ProMissenseTM3Delayed PubertyLikely Pathogenic[103]
c.350T>Gp.Leu117ArgMissenseTM3Hypogonadotropic hypogonadismPathogenic/Likely Pathogenic[95]
c.364C>Tp.Leu122PheMissenseTM3Hypogonadotropic hypogonadismPathogenic[120]
c.386C>Ap.Ala129AspMissenseTM3Hypogonadotropic hypogonadismPathogenic[119]
c.392T>Cp.Met131ThrMissenseTM3Hypogonadotropic hypogonadismPathogenic[95]
c.401T>Gp.Val134GlyMissenseTM3Hypogonadotropic hypogonadismPathogenic[102]
c.415C>Tp.Arg139CysMissenseIL2Hypogonadotropic hypogonadismPathogenic[118]
c.416G>Ap.Arg139HisMissenseIL2Hypogonadotropic hypogonadismPathogenic[117]
c.436C>Tp.Pro146SerMissenseIL2Hypogonadotropic hypogonadismLikely Pathogenic[116]
c.487G>Tp.Ala163SerMissenseTM4Kallman syndromeLikely Pathogenic[115]
c.497T>Cp.Leu166ProMissenseTM4Hypogonadotropic hypogonadismLikely Pathogenic[103]
c.504T>Ap.Ser168ArgMissenseTM4Hypogonadotropic hypogonadismPathogenic[114]
c.511G>Ap.Ala171ThrMissenseTM4Hypogonadotropic hypogonadismPathogenic[113]
c.521A>Gp.Gln174ArgMissenseTM4DSDLikely Pathogenic[112]
c.523-1G>A-Splice acceptor-Hypogonadotropic hypogonadismPathogenic[111]
c.599G>Ap.Cys200TyrMissenseEL2Hypogonadotropic hypogonadismPathogenic[104]
c.651C>Ap.Ser217ArgMissenseTM5Hypogonadotropic hypogonadismPathogenic[110]
c.662T>Ap.Ile221AsnMissenseTM5Hypogonadotropic hypogonadismHypogonadotropic hypogonadism[109]
c.719G>Ap.Arg240GlnMissenseIL3Kallmann syndromeLikely Pathogenic[108]
c.784C>Tp.Arg262TrpMissenseIL3Hypogonadotropic hypogonadismLikely Pathogenic[107]
c.785G>Ap.Arg262GlnMissenseIL3Hypogonadotropic hypogonadismPathogenic/Likely Pathogenic[89]
c.797T>Gp.Leu266ArgMissenseIL3Hypogonadotropic hypogonadismPathogenic/Likely Pathogenic[104]
c.806C>Tp.Thr269MetMissenseIL3Hypogonadotropic hypogonadismPathogenic[105,106]
c.836G>Ap.Cys279TyrMissenseTM6Hypogonadotropic hypogonadismPathogenic[104]
c.842C>Tp.Thr281IleMissenseTM6Hypogonadotropic hypogonadismPathogenic[103]
c.845C>Gp.Pro282ArgMissenseTM6Hypogonadotropic hypogonadismLikely Pathogenic[98]
c.847T>Cp.Tyr283HisMissenseTM6Hypogonadotropic hypogonadismPathogenic[102]
c.851A>Gp.Tyr284CysMissenseTM6Hypogonadotropic hypogonadismLikely Pathogenic[101]
c.869A>Tp.Tyr290PheMissenseTM6Hypogonadotropic hypogonadismLikely Pathogenic[100]
c.924_926delCTTp.Phe309delIn frame deletionTM7Delayed PubertyLikely Pathogenic[99]
c.937_947delp.Phe313Metfs*3FrameshiftTM7hypogonadotropic hypogonadismPathogenic[90]
c.941T>Ap.Leu314*nonsenseTM7Hypogonadotropic hypogonadismPathogenic[96]
c.959C>Tp.Pro320LeuMissenseTM7Hypogonadotropic hypogonadismPathogenic[97]
c.968A>Gp.Tyr323CysMissenseTM7Hypogonadotropic HypogonadismPathogenic[98]
c.987A>Gp.X329WextX22FrameshiftCOOH tailHypogonadotropic hypogonadismLikely Pathogenic[95]
Dup of Exon 1-Exon Duplication-Hypogonadotropic hypogonadismPathogenic[129]
Del of Exon 2-Exon Deletion-Hypogonadotropic hypogonadismPathogenic[129]
Table 2. GnRHR agonists with clinical or veterinary applications.
Table 2. GnRHR agonists with clinical or veterinary applications.
NameBrand NamePubChem CIDMedical Applications
GnRH- Natural ligand of GnRHR
BuserelinSuprefact50225Breast cancer, Endometrial hyperplasia, Endometriosis, Female infertility, Prostate cancer, Uterine fibroids
GoserelinZoladex5311128Breast cancer, Endometriosis, Female infertility, Prostate cancer, Uterine fibroids, Uterine hemorrhage
HistrelinVantas, Supprelin25077993Precocious puberty, Prostate cancer
LeuprorelinLupron657181Breast cancer, Endometriosis, Menorrhagia, Precocious puberty, Prostate cancer, Uterine fibroids
NafarelinSynarel25077405Precocious puberty, Endometriosis
TriptorelinDecapeptyl25074470Breast cancer, Endometriosis, Female infertility, Precocious puberty, Prostate cancer, Uterine fibroids
GonadorelinFactrel638793Cryptorchidism, Delayed puberty, Hypogonadotropic hypogonadism, Veterinary medicine (assisted reproduction)
LecirelinDalmarelin66577115Veterinary medicine (assisted reproduction)
PeforelinMaprelin16197823Veterinary medicine (assisted reproduction)
Azagly-nafarelinGonazon156613532Veterinary medicine (assisted reproduction)
DeslorelinOvuplant, Suprelorin25077495Veterinary medicine (assisted reproduction)
FertirelinOvalyse188304Veterinary medicine (assisted reproduction)
Table 3. GnRHR antagonists with clinical applications.
Table 3. GnRHR antagonists with clinical applications.
NameBrand NameMolecule
Status
PubChem CIDMedical Applications
AbarelixPlenaxisPeptide16131215Prostate cancer
CetrorelixCetrotidePeptide25074887Female infertility
DegarelixFirmagonPeptide6136245Prostate cancer
GanirelixOrgalutranPeptide16130957Female infertility
LinzagolixKLH-2109, OBE-2109Non-peptide16656889Endometriosis, Uterine fibroids
RelugolixReluminaNon-peptide10348973Uterine fibroids, Prostate cancer
ElagolixOrilissaNon-peptide11250647Endometriosis, Uterine fibroids
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fanis, P.; Neocleous, V.; Papapetrou, I.; Phylactou, L.A.; Skordis, N. Gonadotropin-Releasing Hormone Receptor (GnRHR) and Hypogonadotropic Hypogonadism. Int. J. Mol. Sci. 2023, 24, 15965. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242115965

AMA Style

Fanis P, Neocleous V, Papapetrou I, Phylactou LA, Skordis N. Gonadotropin-Releasing Hormone Receptor (GnRHR) and Hypogonadotropic Hypogonadism. International Journal of Molecular Sciences. 2023; 24(21):15965. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242115965

Chicago/Turabian Style

Fanis, Pavlos, Vassos Neocleous, Irene Papapetrou, Leonidas A. Phylactou, and Nicos Skordis. 2023. "Gonadotropin-Releasing Hormone Receptor (GnRHR) and Hypogonadotropic Hypogonadism" International Journal of Molecular Sciences 24, no. 21: 15965. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242115965

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop