Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 250
Filter
1.
Endocrinology ; 163(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-34864945

ABSTRACT

Gonadotropin-releasing hormone (GnRH) regulates gonadal function via its stimulatory effects on gonadotropin production by pituitary gonadotrope cells. GnRH is released from the hypothalamus in pulses and GnRH pulse frequency differentially regulates follicle-stimulating hormone (FSH) and luteinizing hormone (LH) synthesis and secretion. The GnRH receptor (GnRHR) is a G protein-coupled receptor that canonically activates Gα q/11-dependent signaling on ligand binding. However, the receptor can also couple to Gα s and in vitro data suggest that toggling between different G proteins may contribute to GnRH pulse frequency decoding. For example, as we show here, knockdown of Gα s impairs GnRH-stimulated FSH synthesis at low- but not high-pulse frequency in a model gonadotrope-derived cell line. We next used a Cre-lox conditional knockout approach to interrogate the relative roles of Gα q/11 and Gα s proteins in gonadotrope function in mice. Gonadotrope-specific Gα q/11 knockouts exhibit hypogonadotropic hypogonadism and infertility, akin to the phenotypes seen in GnRH- or GnRHR-deficient mice. In contrast, under standard conditions, gonadotrope-specific Gα s knockouts produce gonadotropins at normal levels and are fertile. However, the LH surge amplitude is blunted in Gα s knockout females and postgonadectomy increases in FSH and LH are reduced both in males and females. These data suggest that GnRH may signal principally via Gα q/11 to stimulate gonadotropin production, but that Gα s plays important roles in gonadotrope function in vivo when GnRH secretion is enhanced.


Subject(s)
Chromogranins/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Gonadotrophs/metabolism , Gonadotropins/metabolism , Animals , Castration , Cell Line , Chromogranins/genetics , Female , Fertility/genetics , Fertility/physiology , Follicle Stimulating Hormone, beta Subunit/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/physiology , Gonadotropins/genetics , HEK293 Cells , Humans , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LHRH/genetics , Receptors, LHRH/physiology , Sexual Maturation , Signal Transduction/physiology
2.
Elife ; 102021 12 23.
Article in English | MEDLINE | ID: mdl-34939930

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is the primary neuropeptide controlling reproduction in vertebrates. GnRH stimulates follicle-stimulating hormone (FSH) and luteinizing hormone (LH) synthesis via a G-protein-coupled receptor, GnRHR, in the pituitary gland. In mammals, GnRHR lacks a C-terminal cytosolic tail (Ctail) and does not exhibit homologous desensitization. This might be an evolutionary adaptation that enables LH surge generation and ovulation. To test this idea, we fused the chicken GnRHR Ctail to the endogenous murine GnRHR in a transgenic model. The LH surge was blunted, but not blocked in these mice. In contrast, they showed reductions in FSH production, ovarian follicle development, and fertility. Addition of the Ctail altered the nature of agonist-induced calcium signaling required for normal FSH production. The loss of the GnRHR Ctail during mammalian evolution is unlikely to have conferred a selective advantage by enabling the LH surge. The adaptive significance of this specialization remains to be determined.


Subject(s)
Fertility , Luteinizing Hormone/metabolism , Receptors, LHRH/chemistry , Receptors, LHRH/physiology , Animals , Chickens , Female , Follicle Stimulating Hormone/metabolism , Mice , Mice, Transgenic , Ovarian Follicle/physiology , Receptors, G-Protein-Coupled/physiology
3.
Cell Rep ; 37(8): 110046, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818554

ABSTRACT

Membrane proteins are prone to misfolding and degradation. This is particularly true for mammalian forms of the gonadotropin-releasing hormone receptor (GnRHR). Although they function at the plasma membrane, mammalian GnRHRs accumulate within the secretory pathway. Their apparent instability is believed to have evolved through selection for attenuated GnRHR activity. Nevertheless, the molecular basis of this adaptation remains unclear. We show that adaptation coincides with a C-terminal truncation that compromises the translocon-mediated membrane integration of its seventh transmembrane domain (TM7). We also identify a series of polar residues in mammalian GnRHRs that compromise the membrane integration of TM2 and TM6. Reverting a lipid-exposed polar residue in TM6 to an ancestral hydrophobic residue restores expression with no impact on function. Evolutionary trends suggest variations in the polarity of this residue track with reproductive phenotypes. Our findings suggest that the marginal energetics of cotranslational folding can be exploited to tune membrane protein fitness.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Amino Acid Sequence/genetics , Animals , Cell Membrane/metabolism , Databases, Genetic , Evolution, Molecular , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/metabolism , Membrane Proteins/physiology , Phylogeny , Protein Domains/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Receptors, LHRH/physiology
4.
J Neuroendocrinol ; 31(9): e12709, 2019 09.
Article in English | MEDLINE | ID: mdl-30882966

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is a major regulator and activator of the hypothalamic-pituitary-gonadal axis. Many studies have demonstrated the importance of GnRH in reproduction and sexual behaviour. However, to date, only a single study shows an involvement of GnRH in maternal behaviour where a 30% reduction of GnRH neurones abolishes a mother's motivation to retrieve pups. On this basis, we aimed to investigate the effects of acute central GnRH receptor blockade in lactating rats on maternal care under non-stress and stress conditions, maternal motivation in the pup retrieval test, maternal anxiety on the elevated plus maze, and maternal aggression in the maternal defence test. We found that acute central infusion of a GnRH antagonist ([d-Phe2,6 ,Pro3 ]-luteinising hormone-releasing hormone; 0.5 ng 5 µL-1 ) impaired a mother's attack behaviour against a female intruder rat during the maternal defence test compared to vehicle controls. However, in contrast to the previous study on reduced GnRH neurones, acute central GnRH antagonism did not affect pup retrieval, nor any other parameter of maternal behaviour or maternal anxiety. Taken together, GnRH receptor activation is mandatory for protection of the offspring. These findings shed new light on GnRH as a neuropeptide acting not exclusively on the reproductive axis but, additionally, on maternal behaviour including pup retrieval and maternal aggression.


Subject(s)
Aggression/physiology , Gonadotropin-Releasing Hormone/physiology , Maternal Behavior/physiology , Receptors, LHRH/physiology , Animals , Behavior, Animal , Female , Lactation , Motivation/physiology , Rats, Wistar , Receptors, LHRH/antagonists & inhibitors
5.
J Neuroendocrinol ; 30(12): e12658, 2018 12.
Article in English | MEDLINE | ID: mdl-30415482

ABSTRACT

Congenital hypogonadotrophic hypogonadism (CHH) is a challenging inherited endocrine disorder characterised by absent or incomplete pubertal development and infertility as a result of the low action/secretion of the hypothalamic gonadotrophin-releasing hormone (GnRH). Given a growing list of gene mutations accounting for CHH, the application of massively parallel sequencing comprises an excellent molecular diagnostic approach because it enables the simultaneous evaluation of many genes. The present study proposes the use of whole exome sequencing (WES) to identify causative and modifying mutations based on a phenotype-genotype CHH analysis using an in-house exome pipeline. Based on 44 known genes related to CHH in humans, we were able to identify a novel homozygous gonadotrophin-releasing hormone receptor (GNRHR) p.Thr269Met mutant, which segregates with the CHH kindred and was predicted to be deleterious by in silico analysis. A functional study measuring intracellular inositol phosphate (IP) when stimulated with GnRH on COS-7 cells confirmed that the p.Thr269Met GnRHR mutant performed greatly diminished IP accumulation relative to the transfected wild-type GnRHR. Additionally, the proband carries three heterozygous variants in CCDC141 and one homozygous in SEMA3A gene, although their effects with respect to modifying the phenotype are uncertain. Because they do not segregate with reproductive phenotype in family members, we advocate they do not contribute to CHH oligogenicity. WES proved to be useful for CHH molecular diagnosis and reinforced its benefit with respect to identifying heterogeneous genetic disorders. Our findings expand the GnRHR mutation spectrum and phenotype-genotype correlation in CHH.


Subject(s)
Genetic Predisposition to Disease/genetics , Hypogonadism/genetics , Pedigree , Receptors, LHRH/genetics , Brazil , Cells, Cultured , Female , Humans , Inositol Phosphates/metabolism , Male , Mutation , Receptors, LHRH/physiology , Exome Sequencing
6.
Horm Behav ; 98: 77-87, 2018 02.
Article in English | MEDLINE | ID: mdl-29269179

ABSTRACT

The present study assessed the participation of membrane G-protein coupled estrogen receptor 1 (GPER-1) and gonadotropin releasing hormone 1 (GnRH-1) receptor in the display of lordosis induced by intracerebroventricular (icv) administration of G1, a GPER-1 agonist, and by unesterified 17ß-estradiol (free E2). In addition, we assessed the participation of both estrogen and progestin receptors in the lordosis behavior induced by G1 in ovariectomized (OVX), E2-benzoate (EB)-primed rats. In Experiment 1, icv injection of G1 induced lordosis behavior at 120 and 240min. In Experiment 2, icv injection of the GPER-1 antagonist G15 significantly reduced lordosis behavior induced by either G1 or free E2. In addition, Antide, a GnRH-1 receptor antagonist, significantly depressed G1 facilitation of lordosis behavior in OVX, EB-primed rats. Similarly, icv injection of Antide blocked the stimulatory effect of E2 on lordosis behavior. In Experiment 3, systemic injection of either tamoxifen or RU486 significantly reduced lordosis behavior induced by icv administration of G1 in OVX, EB-primed rats. The results suggest that GnRH release activates both estrogen and progestin receptors and that this activation is important in the chain of events leading to the display of lordosis behavior in response to activation of GPER-1 in estrogen-primed rats.


Subject(s)
Estradiol/pharmacology , Posture/physiology , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/agonists , Receptors, LHRH/physiology , Receptors, Progesterone/physiology , Sexual Behavior, Animal/drug effects , Animals , Female , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Oligopeptides/pharmacology , Rats , Rats, Sprague-Dawley , Sexual Behavior, Animal/physiology , Tamoxifen/pharmacology
7.
Reprod Domest Anim ; 52(6): 1104-1112, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28963736

ABSTRACT

The goals of this study were as follows: (Experiment 1) to examine the basic capability of canine corpora lutea (CL) to respond to GnRH by assessing expression of gonadotropin-releasing hormone receptor (GnRH-R) in luteal samples collected throughout the luteal lifespan from non-pregnant dogs, and (Experiment 2) to investigate the effects of pre-pubertal application of the GnRH agonist deslorelin acetate on luteal function following the first oestrus. Mature CL were collected during the mid-luteal phase (days 30-45) from treated and control bitches. Transcript levels of several factors were determined: estrogen receptors (ESR1/ERα, ESR2/ERß), progesterone (P4)-receptor (PGR), prolactin receptor (PRLR), PGE2-synthase (PTGES) and PGE2 receptors (PTGER2/EP2, PTGER4/EP4), vascular endothelial growth factor (VEGFA) and VEGF receptors (VEGFR1 and VEGFR2), cyclooxygenase 2 (COX2/PTGS2), steroidogenic acute regulatory protein (STAR) and 3ß-hydroxysteroid dehydrogenase (3ßHSD). Additionally, levels of Kisspeptin 1 (Kiss1) and its receptor (KISS1-R) were evaluated. Although generally low, GnRH-R expression was time dependent and was elevated during early dioestrus, with a significant decrease towards luteal regression. In deslorelin-treated and control dogs, its expression was either low or frequently below the detection limit. EP2 and VEGFR1 were higher in the treated group, which could be caused by a feedback mechanism after long-term suppression of reproductive activity. Despite large individual variations, 3ßHSD was higher in the deslorelin-treated group. This, along with unchanged STAR expression, was apparently not mirrored in increased luteal functionality, because similar P4 levels were detected in both groups. Finally, the deslorelin-mediated long-term delay of puberty does not have negative carry-over effects on subsequent ovarian functionality in bitches.


Subject(s)
Corpus Luteum/drug effects , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/physiology , Triptorelin Pamoate/analogs & derivatives , Animals , Corpus Luteum/growth & development , Dogs , Female , Kisspeptins/analysis , Receptors, Cell Surface , Receptors, Steroid , Sexual Maturation/drug effects , Triptorelin Pamoate/pharmacology
8.
PLoS One ; 11(7): e0160292, 2016.
Article in English | MEDLINE | ID: mdl-27467252

ABSTRACT

A gonadotropin-releasing hormone (GnRH)-like molecule was previously identified in a gastropod, Aplysia californica, and named ap-GnRH. In this study, we cloned the full-length cDNA of a putative ap-GnRH receptor (ap-GnRHR) and functionally authenticated this receptor as a bona fide ap-GnRHR. This receptor contains two potential translation start sites, each accompanied by a Kozak sequence, suggesting the translation of a long and a short form of the receptor is possible. The putative ap-GnRHR maintains the conserved structural motifs of GnRHR-like receptors and shares 45% sequence identity with the octopus GnRHR. The expression of the putative ap-GnRHR short form is ubiquitous in all tissues examined, whereas the long form is only expressed in parts of the central nervous system, osphradium, small hermaphroditic duct, and ovotestis. The cDNA encoding the long or the short receptor was transfected into the Drosophila S2 cell line and subject to a radioreceptor assay using 125I-labeled ap-GnRH as the radioligand. Further, the transfected cells were treated with various concentrations of ap-GnRH and measured for the accumulation of cAMP and inositol monophosphate (IP1). Radioreceptor assay revealed that only the long receptor bound specifically to the radioligand. Further, only the long receptor responded to ap-GnRH with an increased accumulation of IP1, but not cAMP. Our studies show that despite the more prevalent expression of the short receptor, only the long receptor is the functional ap-GnRHR. Importantly, this is only the second report on the authentication of a protostome GnRHR, and based on the function and the phylogenetic grouping of ap-GnRHR, we suggest that this receptor is more similar to protostome corazonin receptors than chordate GnRHRs.


Subject(s)
Biological Evolution , Receptors, LHRH/physiology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , DNA, Complementary/genetics , Gastropoda , Phylogeny , Radioligand Assay , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
9.
J Exp Zool A Ecol Genet Physiol ; 325(7): 453-66, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27455909

ABSTRACT

Gonadotropin-releasing hormone (GnRH) plays a vital role in the regulation of reproduction through interaction with a specific receptor (the GnRH receptor). In this study, the GnRH receptor gene from the cuttlefish Sepiella japonica (SjGnRHR) was identified and characterized. The cloned full-length SjGnRHR cDNA was 1,468 bp long and contained a 1,029 bp open reading frame encoding 342 amino acid residues, 8 bp of 5' untranslated regions (UTR), and 431 bp of 3' UTR. The putative protein was predicted to have a molecular weight of 38.75 kDa and an isoelectric point of 9.47. In addition, this protein was identified as belonging to the rhodopsin-type (class A) G protein-coupled receptor family. The predicted amino acid sequence contained two N-linked glycosylation sites and 18 phosphorylation sites. Multiple sequence alignment, phylogenetic tree analysis, and three-dimensional structure modeling were conducted to clarify SjGnRHR bioinformatics characteristics. In vitro SjGnRHR expression was carried out using HEK293 cells and the pEGFP-N1 plasmid, to verify the transmembrane properties of this protein. The interaction between the S. japonica GnRH receptor and its ligand was clarified using internalization analysis. SjGnRHR transcriptional quantification confirmed the wide distribution of SjGnRHR in various S. japonica mature tissues. In addition, the transcriptional profile of SjGnRHR in the female brain and ovary during gonadal development was analyzed. Results indicate that GnRHR may be involved in diverse S. japonica physiological functions, especially in the control of reproduction.


Subject(s)
Decapodiformes/metabolism , Gene Expression , Receptors, LHRH/chemistry , Amino Acid Sequence , Animals , Cloning, Molecular , Decapodiformes/physiology , Female , Humans , Male , Models, Molecular , Organ Specificity , Phylogeny , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Receptors, LHRH/physiology , Reproduction , Sequence Alignment
10.
Eur Surg Res ; 57(1-2): 22-33, 2016.
Article in English | MEDLINE | ID: mdl-27089503

ABSTRACT

BACKGROUND: Gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) are involved in the reproductive cycle and regulate the secretion of sex steroids from the gonads. In mammals, GnRH1 is secreted as a hormone from the hypothalamus, whereas both GnRH1 and GnRH2 are present as neuropeptides in a variety of tissues. This review describes the role of GnRH in the gastrointestinal tract. SUMMARY: GnRH1, GnRH2, and LH receptors in humans and rats, and GnRH receptors in rats, have been described in the gastrointestinal tract, where they affect motility, gastric and hormone secretion, and cell proliferation. GnRH analogs are clinically used in the treatment of sex hormone-dependent diseases, i.e., endometriosis and malignancies, and as pretreatments for in vitro fertilization. Severe gastrointestinal dysmotility has been shown to develop in some women after such treatment, along with a reduction in the number of enteric neurons and autoantibodies against GnRH. Consequently, a rat model of enteric neurodegeneration has been developed based on the administration of the GnRH analog buserelin. Serum IgM antibodies against GnRH1, the GnRH2 precursor progonadoliberin-2, and the GnRH receptor have also been described in patients with irritable bowel syndrome and dysmotility, as well as in patients with gastrointestinal disorders associated with diabetes mellitus, posterior laryngitis, and primary Sjögren's syndrome, although no treatments using GnRH analogs have been administered. CONCLUSION: GnRH and receptors for GnRH and LH are present in the human and rat gastrointestinal tract. Treatment with GnRH analogs may induce severe dysmotility, and a rat model of enteric neurodegeneration has been developed based on stimulation by the GnRH analog buserelin. Autoantibodies against GnRH and its receptor are found in a subgroup of patients with functional bowel disorders and dysmotility, independent of treatment with GnRH analogs.


Subject(s)
Gastrointestinal Tract/physiology , Gonadotropin-Releasing Hormone/physiology , Animals , Antibody Formation , Buserelin/pharmacology , Gastrointestinal Microbiome , Gastrointestinal Motility , Gonadotropin-Releasing Hormone/immunology , Humans , Rats , Receptors, LHRH/physiology
SELECTION OF CITATIONS
SEARCH DETAIL