Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 250
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
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
3.
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
4.
J Neurosci ; 34(46): 15297-305, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25392497

ABSTRACT

Sex differences in brain function underlie robust differences between males and females in both normal and disease states. Although alternative mechanisms exist, sexual differentiation of the male mammalian brain is initiated predominantly by testosterone secreted by the testes during the perinatal period. Despite considerable advances in understanding how testosterone and its metabolite estradiol sexually differentiate the brain, little is known about the mechanism that generates the male-specific perinatal testosterone surge. In mice, we show that a male-specific activation of GnRH neurons occurs 0-2 h following birth and that this correlates with the male-specific surge of testosterone occurring up to 5 h after birth. The necessity of GnRH signaling for the sexually differentiating effects of the perinatal testosterone surge was demonstrated by the persistence of female-like brain characteristics in adult male, GnRH receptor knock-out mice. Kisspeptin neurons have recently been identified to be potent, direct activators of GnRH neurons. We demonstrate that a population of kisspeptin neurons appears in the preoptic area of only the male between E19 and P1. The importance of kisspeptin inputs to GnRH neurons for the process of sexual differentiation was demonstrated by the lack of a normal neonatal testosterone surge, and disordered brain sexual differentiation of male mice in which the kisspeptin receptor was deleted selectively from GnRH neurons. These observations demonstrate the necessity of perinatal GnRH signaling for driving brain sexual differentiation and indicate that kisspeptin inputs to GnRH neurons are essential for this process to occur.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Neurons/physiology , Preoptic Area/metabolism , Receptors, G-Protein-Coupled/physiology , Sex Differentiation/physiology , Signal Transduction , Animals , Animals, Newborn , Female , Gonadotropin-Releasing Hormone/genetics , Male , Mice , Mice, Knockout , Neurons/metabolism , Pregnancy , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Receptors, LHRH/genetics , Receptors, LHRH/physiology , Sex Characteristics , Testosterone/blood , Tyrosine 3-Monooxygenase/metabolism , Vasopressins/metabolism
5.
Am J Physiol Endocrinol Metab ; 305(1): E67-77, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23632635

ABSTRACT

The human GnRH receptor (GNRHR1) has a specific set of properties with physiological and pharmacological influences not appropriately modeled in laboratory animals or cell-based systems. To address this deficiency, we have generated human GNRHR1 knock-in mice and described their reproductive phenotype. Measurement of pituitary GNRHR1 transcripts from homozygous human GNRHR1 knock-in (ki/ki) mice revealed a severe reduction (7- to 8-fold) compared with the mouse Gnrhr1 in wild-type mice. ¹²5I-GnRH binding assays on pituitary membrane fractions corroborated reduced human GNRHR1 protein expression in ki/ki mice, as occurs with transfection of human GNRHR1 in cell lines. Female homozygous knock-in mice displayed normal pubertal onset, indicating that a large reduction in GNRHR1 expression is sufficient for this process. However, ki/ki females exhibited periods of prolonged estrous and/or metestrous and reduced fertility. No impairment was found in reproductive maturity or adult fertility in male ki/ki mice. Interestingly, the serum LH response to GnRH challenge was reduced in both knock-in males and females, indicating a reduced GNRHR1 signaling capacity. Small molecules targeting human GPCRs usually have poor activities at homologous rodent receptors, thus limiting their use in preclinical development. Therefore, we tested a human-specific GnRH1 antagonist, NBI-42902, in our mouse model and demonstrated abrogation of a GnRH1-induced serum LH rise in ki/ki mice and an absence of effect in littermates expressing the wild-type murine receptor. This novel model provides the opportunity to study the human receptor in vivo and for screening the activity of human-specific GnRH analogs.


Subject(s)
Estrous Cycle/physiology , Fertility/physiology , Receptors, LHRH/genetics , Receptors, LHRH/physiology , Reproduction/physiology , Animals , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Transgenic , Models, Animal , Phenotype , Pituitary Gland/physiology , Pregnancy , Receptors, LHRH/antagonists & inhibitors , Sexual Maturation/physiology , Testis/growth & development , Testis/physiology , Thymine/analogs & derivatives , Thymine/pharmacology
6.
Theor Biol Med Model ; 9: 35, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22870942

ABSTRACT

When hormones during the ovulatory cycle are shown in phase plane graphs, reported FSH and estrogen values form a specific pattern that resembles the leaning "&" symbol, while LH and progesterone (Pg) values form a "boomerang" shape. Graphs in this paper were made using data reported by Stricker et al. [Clin Chem Lab Med 2006;44:883-887]. These patterns were used to construct a simplistic model of the ovulatory cycle without the conventional "positive feedback" phenomenon. The model is based on few well-established relations:hypothalamic GnRH secretion is increased under estrogen exposure during two weeks that start before the ovulatory surge and lasts till lutheolysis.the pituitary GnRH receptors are so prone to downregulation through ligand binding that this must be important for their function.in several estrogen target tissue progesterone receptor (PgR) expression depends on previous estrogen binding to functional estrogen receptors (ER), while Pg binding to the expressed PgRs reduces both ER and PgR expression.Some key features of the presented model are here listed:High GnRH secretion induced by the recovered estrogen exposure starts in the late follicular phase and lasts till lutheolysis. The LH and FSH surges start due to combination of accumulated pituitary GnRH receptors and increased GnRH secretion. The surges quickly end due to partial downregulation of the pituitary GnRH receptors (64% reduction of the follicular phase pituitary GnRH receptors is needed to explain the reported LH drop after the surge). A strong increase in the lutheal Pg blood level, despite modest decline in LH levels, is explained as delayed expression of pituitary PgRs. Postponed pituitary PgRs expression enforces a negative feedback loop between Pg levels and LH secretions not before the mid lutheal phase.Lutheolysis is explained as a consequence of Pg binding to hypothalamic and pituitary PgRs that reduces local ER expression. When hypothalamic sensitivity to estrogen is diminished due to lack of local ERs, hypothalamus switches back to the low GnRH secretion rate, leading to low secretion of gonadotropins and to lutheolysis. During low GnRH secretion rates, previously downregulated pituitary GnRH receptors recover to normal levels and thus allow the next cycle.Possible implications of the presented model on several topics related to reproductive physiology are shortly discussed with some evolutionary aspects including the emergence of menopause.


Subject(s)
Menstrual Cycle/physiology , Models, Biological , Ovulation/physiology , Activins/physiology , Biological Evolution , Estrogens/physiology , Feedback, Physiological , Female , Follicle Stimulating Hormone/physiology , Humans , Hypothalamus/physiology , Inhibins/physiology , Leptin/physiology , Luteinizing Hormone/physiology , Luteolysis/physiology , Male , Pituitary Gland/physiology , Progesterone/physiology , Puberty/physiology , Receptors, Estrogen/physiology , Receptors, LHRH/physiology , Receptors, Progesterone/physiology , Sex Characteristics
7.
Gen Comp Endocrinol ; 175(3): 398-406, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22138555

ABSTRACT

The European sea bass expresses three GnRH (Gonadotrophin Releasing Hormone) forms that exert pleiotropic actions via several classes of receptors. The GnRH-1 form is responsible for the endogenous regulation of gonadotrophin release by the pituitary gland but the role of GnRH-2 and GnRH-3 remains unclear in fish. In a previous study performed in sea bass, we have provided evidence of direct links between the GnRH-2 cells and the pineal organ and demonstrated a functional role for GnRH-2 in the modulation of the secretory activity of this photoreceptive organ. In this study, we have investigated the possible relationship between the GnRH-3 system and the retina in the same species. Thus, using a biotinylated dextran-amine tract-tracing method, we reveal the presence of retinopetal cells in the terminal nerve of sea bass, a region that also contains GnRH-3-immunopositive cells. Moreover, GnRH-3-immunoreactive fibers were observed at the boundary between the inner nuclear and the inner plexiform layers, and also within the ganglion cell layer. These results strongly suggest that the GnRH-3 neurons located in the terminal nerve area represent the source of GnRH-3 innervation in the retina of this species. In order to clarify whether the retina is a target for GnRH, the expression pattern of GnRH receptors (dlGnRHR) was also analyzed by RT-PCR and in situ hybridization. RT-PCR revealed the retinal expression of dlGnRHR-II-2b, -1a, -1b and -1c, while in situ hybridization only showed positive signals for the receptors dlGnRHR-II-2b and -1a. Finally, double-immunohistochemistry showed that GnRH-3 projections reaching the sea bass retina end in close proximity to tyrosine hydroxylase (dopaminergic) cells, which also expressed the dlGnRHR-II-2b receptor subtype. Taken together, these results suggest an important role for GnRH-3 in the modulation of dopaminergic cell activities and retinal functions in sea bass.


Subject(s)
Bass/physiology , Gonadotropin-Releasing Hormone/physiology , Pyrrolidonecarboxylic Acid/analogs & derivatives , Retina/physiology , Signal Transduction/physiology , Animals , Male , Receptors, LHRH/physiology , Retinal Ganglion Cells/physiology , Tyrosine 3-Monooxygenase/physiology
8.
Gen Comp Endocrinol ; 176(3): 321-6, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22172342

ABSTRACT

Several protostomian molecules that structurally resemble chordate gonadotropin-releasing hormone (GnRH) have been identified through cloning, biochemical purification or data mining. These molecules share considerable sequence and structural similarities with chordate GnRH, leading to the current belief that protostomian and chordate forms of GnRH share a common ancestor. However, the physiological significance of these protostomian GnRH-like molecules remains poorly understood. This knowledge gap hampers our understanding of how GnRH has evolved functionally over time. This review provides a summary of our recent functional characterization of a GnRH-like molecule (ap-GnRH) in a gastropod mollusk, Aplysia californica, and presents preliminary proof for a cognate ap-GnRH receptor (ap-GnRHR). Our data reveal that ap-GnRH is a general neural regulator capable of exerting diverse central and motor effects, but plays little or no role in reproductive activation. This notion is supported by the abundance of a putative ap-GnRHR transcript in the central nervous system and the foot. Comparing these results to the available functional data from a cephalopod mollusk, Octopus vulgaris, we surmise that protostomian GnRH-like molecules are likely to assume a wide range of physiological roles, and reproductive activation is not an evolutionarily conserved role of these molecules. Future functional studies using suitable protostomian models are required to identify functional changes in protostomian GnRH-like molecules that accompany major taxa-level transitions.


Subject(s)
Aplysia/physiology , Central Nervous System/physiology , Evolution, Molecular , Gonadotropin-Releasing Hormone/physiology , Receptors, LHRH/physiology , Amino Acid Sequence , Animals , Aplysia/genetics , Gonadotropin-Releasing Hormone/genetics , Molecular Sequence Data , Phylogeny , Receptors, LHRH/genetics , Sequence Alignment
9.
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
10.
Front Neuroendocrinol ; 31(3): 241-58, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20546773

ABSTRACT

As the final common pathway for the central control of gonadotropin secretion, GnRH neurons are subjected to numerous regulatory homeostatic and external factors to achieve levels of fertility appropriate to the organism. The GnRH system thus provides an excellent model in which to investigate the complex relationships between neurosecretion, morphological plasticity and the expression of a physiological function. Throughout the reproductive cycle beginning from postnatal sexual development and the onset of puberty to reproductive senescence, and even within the ovarian cycle itself, all levels of the GnRH system undergo morphological plasticity. This structural plasticity within the GnRH system appears crucial to the timely control of reproductive competence within the individual, and as such must have coordinated actions of multiple signals secreted from glial cells, endothelial cells, and GnRH neurons. Thus, the GnRH system must be viewed as a complete neuro-glial-vascular unit that works in concert to maintain the reproductive axis.


Subject(s)
Cell Communication/physiology , Endothelial Cells/physiology , Gonadotropin-Releasing Hormone/metabolism , Neuroglia/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Animals , Endothelial Cells/metabolism , Female , Gonadotropin-Releasing Hormone/physiology , Humans , Models, Biological , Neuroglia/metabolism , Neurons/metabolism , Ovary/metabolism , Ovary/physiology , Puberty/metabolism , Puberty/physiology , Receptors, LHRH/metabolism , Receptors, LHRH/physiology
11.
J Pharmacol Exp Ther ; 338(2): 430-42, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21527534

ABSTRACT

G protein-coupled receptors (GPCRs) play central roles in most physiological functions, and mutations in them cause heritable diseases. Whereas crystal structures provide details about the structure of GPCRs, there is little information that identifies structural features that permit receptors to pass the cellular quality control system or are involved in transition from the ground state to the ligand-activated state. The gonadotropin-releasing hormone receptor (GnRHR), because of its small size among GPCRs, is amenable to molecular biological approaches and to computer modeling. These techniques and interspecies comparisons are used to identify structural features that are important for both intracellular trafficking and GnRHR activation yet distinguish between these processes. Our model features two salt (Arg(38)-Asp(98) and Glu(90)-Lys(121)) and two disulfide (Cys(14)-Cys(200) and Cys(114)-Cys(196)) bridges, all of which are required for the human GnRHR to traffic to the plasma membrane. This study reveals that both constitutive and ligand-induced activation are associated with a "coincidence detector" that occurs when an agonist binds. The observed constitutive activation of receptors lacking Glu(90)-Lys(121), but not Arg(38)-Asp(98) ionic bridge, suggests that the role of the former connection is holding the receptor in the inactive conformation. Both the aromatic ring and hydroxyl group of Tyr(284) and the hydrogen bonding of Ser(217) are important for efficient receptor activation. Our modeling results, supported by the observed influence of Lys(191) from extracellular loop 2 (EL2) and a four-residue motif surrounding this loop on ligand binding and receptor activation, suggest that the positioning of EL2 within the seven-α-helical bundle regulates receptor stability, proper trafficking, and function.


Subject(s)
Amino Acids, Basic/chemistry , Amino Acids, Basic/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, LHRH/chemistry , Receptors, LHRH/physiology , Animals , Binding Sites/physiology , COS Cells , Cattle , Chlorocebus aethiops , Crystallography, X-Ray , Extracellular Fluid/metabolism , Humans , Mice , Mutation/physiology , Protein Binding/physiology , Protein Stability , Protein Transport , Rats , Receptors, G-Protein-Coupled/chemistry , Salts/chemistry , Salts/metabolism
12.
Curr Opin Obstet Gynecol ; 23(4): 238-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21666463

ABSTRACT

PURPOSE OF REVIEW: Gonadotropin-releasing hormone (GnRH) receptors are not only detected in the central nervous system but also in tissues such as ovary, endometrium, breast, gastrointestinal system, placenta and malignant tumors of ovary and breast. The direct role of GnRH-antagonists in ovarian function, implantation, cancer pathogenesis and treatment is under extensive investigation. This study reviews the biochemistry and molecular and cellular biology of GnRH-antagonists as well as GnRH types and their receptors. RECENT FINDINGS: The best clinical evidence with GnRH-antagonists has accumulated in controlled ovarian hyperstimulation protocols for prevention of premature luteinizing hormone surge (cetrorelix, ganirelix) and in the treatment of advanced-stage prostate cancer (abarelix and degarelix). GnRH-GnRH receptor pathways may have a role in the embryo implantation. The controversy still exists whether GnRH antagonist protocols result in slightly decreased clinical pregnancy rates compared with the GnRH agonist protocols. GnRH-antagonists could be used in the near future to treat some cancer types that express GnRH receptors. SUMMARY: GnRH-antagonists have various clinical applications in gynecology, reproductive medicine, urology and oncology. The emergence of well tolerated, orally active GnRH-antagonists may provide an alternative to long-term injections and is likely to have a major impact on the utility of GnRH analogues in the treatment of human diseases.


Subject(s)
Breast Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/therapeutic use , Prostatic Neoplasms/drug therapy , Embryo Implantation/drug effects , Female , Gonadotropin-Releasing Hormone/agonists , Hormone Antagonists/pharmacokinetics , Hormone Antagonists/pharmacology , Humans , Male , Ovulation Induction , Receptors, LHRH/physiology , Signal Transduction/physiology
13.
Gen Comp Endocrinol ; 170(3): 424-48, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21074534

ABSTRACT

The main purpose of this review is to provide a comprehensive update on what is known about the regulatory mechanisms of the hypothalamic-pituitary-ovarian axis in the brushtail possum, and to report on the outcomes of attempts made to manipulate by hormonal means, these processes in the possum. Over the last 15 years, several unique features of possum reproductive physiology have been discovered. These include an extended follicular phase despite elevated concentrations of FSH during the luteal phase, and early expression of LH receptors on granulosa cells of small antral follicles, suggesting a different mechanism for the selection of a dominant follicle. The use of routine synchronisation protocols that are effective in eutherians has failed to be effective in possums, and so the ability to reliably synchronise oestrus in this species remains a challenge.


Subject(s)
Hypothalamo-Hypophyseal System/physiology , Ovary/physiology , Trichosurus/physiology , Animals , Estrous Cycle , Female , Gonadotropin-Releasing Hormone/physiology , Introduced Species , Ovarian Follicle/drug effects , Ovulation/physiology , RNA, Messenger/metabolism , Receptors, LHRH/physiology
14.
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
15.
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
16.
Front Neuroendocrinol ; 30(1): 10-29, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18708085

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is the first key hormone of reproduction. GnRH analogs are extensively used in in vitro fertilization, and treatment of sex hormone-dependent cancers, due to their ability to bring about 'chemical castration'. The interaction of GnRH with its cognate type I receptor (GnRHR) in pituitary gonadotropes results in the activation of Gq/G(11), phospholipase Cbeta (PLCbetaI), PLA(2), and PLD. Sequential activation of the phospholipases generates the second messengers inositol 1, 4, 5-trisphosphate (IP(3)), diacylglycerol (DAG), and arachidonic acid (AA), which are required for Ca(2+) mobilization, the activation of various protein kinase C isoforms (PKCs), and the production of prostaglandin (PG) and other metabolites of AA, respectively. PKC isoforms are the major mediators of the downstream activation of a number of mitogen-activated protein kinase (MAPK) cascades by GnRH, namely: extracellular signal-regulated kinase (ERK), jun-N-terminal kinase (JNK), and p38MAPK. The activated MAPKs phosphorylate both cytosolic and nuclear proteins to initiate the transcriptional activation of the gonadotropin subunit genes and the GnRHR. While Ca(2+) mobilization has been found to initiate rapid gonadotropin secretion, Ca(2+), together with various PKC isoforms, MAPKs and AA metabolites also serve as key nodes, in the GnRH-stimulated signaling network that enables the gonadotropes to decode GnRH pulse frequencies and translating that into differential gonadotropin synthesis and release. Even though pulsatility of GnRH is recognized as a major determinant for differential gonadotropin subunit gene expression and gonadotropin secretion very little is yet known about the signaling circuits governing GnRH action at the 'Systems Biology' level. Direct apoptotic and metastatic effects of GnRH analogs in gonadal steroid-dependent cancers expressing the GnRHR also seem to be mediated by the activation of the PKC/MAPK pathways. However, the mechanisms dictating life (pituitary) vs. death (cancer) decisions made by the same GnRHR remain elusive. Understanding these molecular mechanisms triggered by the GnRHR through biochemical and 'Systems Biology' approaches would provide the basis for the construction of the dynamic connectivity maps, which operate in the various cell types (endocrine, cancer, and immune system) targeted by GnRH. The connectivity maps will open a new vista for exploring the direct effects of GnRH analogs in tumors and the design of novel combined therapies for fertility control, reproductive disorders and cancers.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Receptors, LHRH/physiology , Signal Transduction/physiology , Adenylyl Cyclases/physiology , Animals , Calcium Signaling/physiology , Enzyme Activation , Guanylate Cyclase/physiology , Humans , MAP Kinase Signaling System/physiology , Phospholipases/metabolism , Pituitary Gland/physiology , Protein Kinase C/physiology , Receptors, Prostaglandin/physiology
17.
Gen Comp Endocrinol ; 167(2): 331-7, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20350548

ABSTRACT

Gonadotropin-inhibitory hormone (GnIH) is a hypothalamic peptide from the RFamide peptide family that has been identified in multiple avian species. Although GnIH has clearly been shown to reduce LH release from the anterior pituitary gland, its mechanism of action remains to be determined. The overall objectives of this study were (1) to characterize the GnIH receptor (GnIH-R) signaling pathway, (2) to evaluate potential interactions with gonadotropin releasing hormone type III receptor (GnRH-R-III) signaling, and (3) to determine the molecular mechanisms by which GnIH and GnRH regulate pituitary gonadotrope function during a reproductive cycle in the chicken. Using real-time PCR, we showed that in the chicken pituitary gland, GnIH-R mRNA levels fluctuate in an opposite manner to GnRH-R-III, with higher and lower levels observed during inactive and active reproductive stages, respectively. We demonstrated that the chicken GnIH-R signals by inhibiting adenylyl cyclase cAMP production, most likely by coupling to G(alphai). We also showed that this inhibition is sufficient to significantly reduce GnRH-induced cAMP responsive element (CRE) activation in a dose-dependent manner, and that the ratio of GnRH/GnIH receptors is a significant factor. We propose that in avian species, sexual maturation is characterized by a change in GnIH/GnRH receptor ratio, resulting in a switch in pituitary sensitivity from inhibitory (involving GnIH) to stimulatory (involving GnRH). In turn, decreasing GnIH-R signaling, combined with increasing GnRH-R-III signaling, results in significant increases in CRE activation, possibly initiating gonadotropin synthesis.


Subject(s)
Chickens/physiology , Cyclic AMP/physiology , Glycoproteins/physiology , Gonadotropin-Releasing Hormone/physiology , Pyrrolidonecarboxylic Acid/analogs & derivatives , Receptors, LHRH/physiology , Animals , Cell Line , Cloning, Molecular , Cyclic AMP/antagonists & inhibitors , Female , Gene Expression Profiling , Male , Pituitary Gland/physiology , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
18.
Gynecol Endocrinol ; 26(9): 669-72, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20236028

ABSTRACT

Most surgical procedures performed by obstetrician-gynecologists are associated with pelvic adhesions that cause subsequent serious sequelae, including small bowel obstruction, infertility, chronic pelvic pain, and difficulty in postoperative treatment, including complexity during subsequent surgical procedures. This study was conducted to determine if gonadotropin-releasing hormone analogues (GnRHa) affect the expressing tissue-type plasminogen activator (t-PA) and its inhibitor-1 (PAI-1) in peritoneal cells in culture. Human peritoneal Met5A cells were used to examine the effects of GnRHa leuprolide, buserelin and goserelin on the levels of t-PA and PA-1. Antigen concentrations were measured in conditioned media and cell lysates by real-time PCR and ELISA. GnRH receptor (GnRHR) mRNA was determined by RT-PCR. GnRHR mRNA was detected in Met5A cells. Exposure of Met5A cells to GnRHa induced a rapid decrease of PAI-1 level in cultured medium but not in cell lysate (protein and mRNA). These effects of GnRHa on PAI-1 were not associated with any changes in t-PA level. These results suggest that GnRHa may be an effective stimulator of local peritoneal fibrinolytic activity, as it decreases PAI-1 secretion in peritoneal Met5A cells by a mechanism linked to GnRHR.


Subject(s)
Fibrinolysin/metabolism , Peritoneum/metabolism , Receptors, LHRH/physiology , Buserelin/pharmacology , Cells, Cultured , Enzyme Activation , Gene Expression/drug effects , Gene Expression/physiology , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Goserelin/pharmacology , Humans , Leuprolide/pharmacology , Peritoneum/enzymology , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism
19.
Front Neuroendocrinol ; 29(1): 17-35, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17976709

ABSTRACT

Geoffrey Wingfield Harris' demonstration of hypothalamic hormones regulating pituitary function led to their structural identification and therapeutic utilization in a wide spectrum of diseases. Amongst these, Gonadotropin Releasing Hormone (GnRH) and its analogs are widely employed in modulating gonadotropin and sex steroid secretion to treat infertility, precocious puberty and many hormone-dependent diseases including endometriosis, uterine fibroids and prostatic cancer. While these effects are all mediated via modulation of the pituitary gonadotrope GnRH receptor and the G(q) signaling pathway, it has become increasingly apparent that GnRH regulates many extrapituitary cells in the nervous system and periphery. This review focuses on two such examples, namely GnRH analog effects on reproductive behaviors and GnRH analog effects on the inhibition of cancer cell growth. For both effects the relative activities of a range of GnRH analogs is distinctly different from their effects on the pituitary gonadotrope and different signaling pathways are utilized. As there is only a single functional GnRH receptor type in man we have proposed that the GnRH receptor can assume different conformations which have different selectivity for GnRH analogs and intracellular signaling proteins complexes. This ligand-induced selective-signaling recruits certain pathways while by-passing others and has implications in developing more selective GnRH analogs for highly specific therapeutic intervention.


Subject(s)
Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/physiology , Ligands , Receptors, LHRH/agonists , Reproductive Behavior/drug effects , Signal Transduction/drug effects , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Gene Silencing , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/chemistry , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Neoplasms/pathology , Protein Isoforms/chemistry , Protein Isoforms/physiology , Receptors, LHRH/physiology , Reproductive Behavior/physiology , Sequence Homology, Amino Acid , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Signal Transduction/physiology , Structure-Activity Relationship , Substrate Specificity
20.
Endocr Rev ; 26(2): 283-306, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15561800

ABSTRACT

In human beings, two forms of GnRH, termed GnRH-I and GnRH-II, encoded by separate genes have been identified. Although these hormones share comparable cDNA and genomic structures, their tissue distribution and regulation of gene expression are significantly dissimilar. The actions of GnRH are mediated by the GnRH receptor, which belongs to a member of the rhodopsin-like G protein-coupled receptor superfamily. However, to date, only one conventional GnRH receptor subtype (type I GnRH receptor) uniquely lacking a carboxyl-terminal tail has been found in the human body. Studies on the transcriptional regulation of the human GnRH receptor gene have indicated that tissue-specific gene expression is mediated by differential promoter usage in various cell types. Functionally, there is growing evidence showing that both GnRH-I and GnRH-II are potentially important autocrine and/or paracrine regulators in some extrapituitary compartments. Recent cloning of a second GnRH receptor subtype (type II GnRH receptor) in nonhuman primates revealed that it is structurally and functionally distinct from the mammalian type I receptor. However, the human type II receptor gene homolog carries a frameshift and a premature stop codon, suggesting that a full-length type II receptor does not exist in humans.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Receptors, LHRH/physiology , Signal Transduction/physiology , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/genetics , Humans , Male , Protein Isoforms , Receptors, LHRH/genetics , Reproduction/genetics , Reproduction/physiology , Signal Transduction/genetics , Transcription, Genetic/genetics , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL