Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
1.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34864945

RESUMO

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.


Assuntos
Cromograninas/fisiologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Gonadotrofos/metabolismo , Gonadotropinas/metabolismo , Animais , Castração , Linhagem Celular , Cromograninas/genética , Feminino , Fertilidade/genética , Fertilidade/fisiologia , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Gonadotropinas/genética , Células HEK293 , Humanos , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores LHRH/genética , Receptores LHRH/fisiologia , Maturidade Sexual , Transdução de Sinais/fisiologia
2.
J Neuroendocrinol ; 31(9): e12709, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30882966

RESUMO

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.


Assuntos
Agressão/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Comportamento Materno/fisiologia , Receptores LHRH/fisiologia , Animais , Comportamento Animal , Feminino , Lactação , Motivação/fisiologia , Ratos Wistar , Receptores LHRH/antagonistas & inibidores
3.
Horm Behav ; 98: 77-87, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29269179

RESUMO

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.


Assuntos
Estradiol/farmacologia , Postura/fisiologia , Receptores de Estrogênio/fisiologia , Receptores Acoplados a Proteínas G/agonistas , Receptores LHRH/fisiologia , Receptores de Progesterona/fisiologia , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Feminino , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , Oligopeptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Comportamento Sexual Animal/fisiologia , Tamoxifeno/farmacologia
4.
Reprod Domest Anim ; 52(6): 1104-1112, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28963736

RESUMO

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.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/fisiologia , Pamoato de Triptorrelina/análogos & derivados , Animais , Corpo Lúteo/crescimento & desenvolvimento , Cães , Feminino , Kisspeptinas/análise , Receptores de Superfície Celular , Receptores de Esteroides , Maturidade Sexual/efeitos dos fármacos , Pamoato de Triptorrelina/farmacologia
5.
Eur Surg Res ; 57(1-2): 22-33, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27089503

RESUMO

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.


Assuntos
Trato Gastrointestinal/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Animais , Formação de Anticorpos , Busserrelina/farmacologia , Microbioma Gastrointestinal , Motilidade Gastrointestinal , Hormônio Liberador de Gonadotropina/imunologia , Humanos , Ratos , Receptores LHRH/fisiologia
6.
Hum Reprod Update ; 22(3): 358-81, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26715597

RESUMO

BACKGROUND: Human reproduction depends on an intact hypothalamic-pituitary-gonadal (HPG) axis. Hypothalamic gonadotrophin-releasing hormone (GnRH) has been recognized, since its identification in 1971, as the central regulator of the production and release of the pituitary gonadotrophins that, in turn, regulate the gonadal functions and the production of sex steroids. The characteristic peculiar development, distribution and episodic activity of GnRH-producing neurons have solicited an interdisciplinary interest on the etiopathogenesis of several reproductive diseases. The more recent identification of a GnRH/GnRH receptor (GnRHR) system in both the human endometrium and ovary has widened the spectrum of action of the peptide and of its analogues beyond its hypothalamic function. METHODS: An analysis of research and review articles published in international journals until June 2015 has been carried out to comprehensively summarize both the well established and the most recent knowledge on the physiopathology of the GnRH system in the central and peripheral control of female reproductive functions and diseases. RESULTS: This review focuses on the role of GnRH neurons in the control of the reproductive axis. New knowledge is accumulating on the genetic programme that drives GnRH neuron development to ameliorate the diagnosis and treatment of GnRH deficiency and consequent delayed or absent puberty. Moreover, a better understanding of the mechanisms controlling the episodic release of GnRH during the onset of puberty and the ovulatory cycle has enabled the pharmacological use of GnRH itself or its synthetic analogues (agonists and antagonists) to either stimulate or to block the gonadotrophin secretion and modulate the functions of the reproductive axis in several reproductive diseases and in assisted reproduction technology. Several inputs from other neuronal populations, as well as metabolic, somatic and age-related signals, may greatly affect the functions of the GnRH pulse generator during the female lifespan; their modulation may offer new possible strategies for diagnostic and therapeutic interventions. A GnRH/GnRHR system is also expressed in female reproductive tissues (e.g. endometrium and ovary), both in normal and pathological conditions. The expression of this system in the human endometrium and ovary supports its physiological regulatory role in the processes of trophoblast invasion of the maternal endometrium and embryo implantation as well as of follicular development and corpus luteum functions. The GnRH/GnRHR system that is expressed in diseased tissues of the female reproductive tract (both benign and malignant) is at present considered an effective molecular target for the development of novel therapeutic approaches for these pathologies. GnRH agonists are also considered as a promising therapeutic approach to counteract ovarian failure in young female patients undergoing chemotherapy. CONCLUSIONS: Increasing knowledge about the regulation of GnRH pulsatile release, as well as the therapeutic use of its analogues, offers interesting new perspectives in the diagnosis, treatment and outcome of female reproductive disorders, including tumoral and iatrogenic diseases.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Receptores LHRH/fisiologia , Reprodução/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/análise , Gônadas/metabolismo , Humanos , Hipogonadismo , Hipotálamo , Doenças Ovarianas
7.
Mol Cell Endocrinol ; 402: 95-106, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25583361

RESUMO

Transmembrane helix seven residues of G protein-coupled receptors (GPCRs) couple agonist binding to a conserved receptor activation mechanism. Amino-terminal residues of the GnRH peptide determine agonist activity. We investigated GnRH interactions with the His(7.36(305)) residue of the GnRH receptor, using functional and computational analysis of modified GnRH receptors and peptides. Non-polar His(7.36(305)) substitutions decreased receptor affinity for GnRH four- to forty-fold, whereas GnRH signaling potency was more decreased (~150-fold). Uncharged polar His(7.36(305)) substitutions decreased GnRH potency, but not affinity. [2-Nal(3)]-GnRH retained high affinity at receptors with non-polar His(7.36(305)) substitutions, supporting a role for His(7.36(305)) in recognizing Trp(3) of GnRH. Compared with GnRH, [2-Nal(3)]-GnRH potency was lower at the wild type GnRH receptor, but unchanged or higher at mutant receptors. Results suggest that His(7.36(305)) of the GnRH receptor forms two distinct interactions that determine binding to Trp(3) and couple agonist binding to the conserved transmembrane domain network that activates GPCRs.


Assuntos
Histidina/metabolismo , Receptores LHRH/fisiologia , Sequência de Aminoácidos , Animais , Ligação Competitiva , Células COS , Chlorocebus aethiops , Sequência Conservada , Hormônio Liberador de Gonadotropina/química , Hormônio Liberador de Gonadotropina/fisiologia , Fosfatos de Inositol/biossíntese , Camundongos , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores LHRH/química , Transdução de Sinais
8.
Int J Oncol ; 46(1): 243-53, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25351635

RESUMO

It is well established that gonadotropin-releasing hormone receptors (GnRH-R) are expressed in different types of cancers, including castration-resistant prostate cancer (CRPC) and mediate the antiproliferative effect of GnRH analogs. Thus, these compounds are employed as targeting moieties to selectively deliver chemotherapeutic agents to cancer cells. GnRH-III, the decapeptide isolated from the sea lamprey brain, has lower potency than GnRH in stimulating gonadotropin secretion, but it exerts antiproliferative effects on many tumors expressing the GnRH-R. GnRH-III-based peptides are considered promising targeting moieties for the preparation of anticancer drug delivery systems. These studies were aimed at i) evaluating the antitumor activity of two cytotoxic oxime bond-linked daunorubicin (Dau)-GnRH-III derivative bioconjugates (Dau-GnRH-III, in which daunorubicin was coupled to the 8Lys in the native form of GnRH-III, and Dau-[4Lys(Ac)]-GnRH-III, in which daunorubicin was attached to the 8Lys of a GnRH-III derivative where 4Ser was replaced by an acetylated lysine) on CRPC cells; and ii) to elucidate the involvement of the classical GnRH-R (type I GnRH-R) in this antitumor activity. Our results demonstrated that both Dau-GnRH-III and Dau-[4Lys(Ac)]-GnRH-III were rapidly internalized into DU145 prostate cancer cells and exerted a significant cytostatic effect. Both bioconjugates increased the levels of the active form of caspase-3, indicating the involvement of apoptosis in their antitumor activity. The antiproliferative effect of both Dau-GnRH-III and Dau-[4Lys(Ac)]-GnRH-III was counteracted by the simultaneous treatment of the cells with Antide, an antagonist of the GnRH-R. Moreover, after silencing the type I GnRH-R the antitumor activity of both bioconjugates was completely abolished. These data demonstrate that in CRPC cells, daunorubicin-GnRH-III derivative bioconjugates: i) inhibit tumor cell proliferation, by triggering the apoptosis process; ii) exert their antitumor effect through the activation of the type I GnRH-R expressed on these cells. Cytotoxic-GnRH-III derivative may represent promising targeted chemotherapeutics for the treatment of CRPC patients.


Assuntos
Antineoplásicos/farmacologia , Daunorrubicina/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Oximas/farmacologia , Neoplasias de Próstata Resistentes à Castração/patologia , Ácido Pirrolidonocarboxílico/análogos & derivados , Receptores LHRH/fisiologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Daunorrubicina/química , Relação Dose-Resposta a Droga , Hormônio Liberador de Gonadotropina/química , Humanos , Masculino , Oximas/química , Ácido Pirrolidonocarboxílico/química , Ácido Pirrolidonocarboxílico/farmacologia , Células Tumorais Cultivadas
9.
J Neurosci ; 34(46): 15297-305, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25392497

RESUMO

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.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Área Pré-Óptica/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Diferenciação Sexual/fisiologia , Transdução de Sinais , Animais , Animais Recém-Nascidos , Feminino , Hormônio Liberador de Gonadotropina/genética , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Gravidez , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Receptores LHRH/genética , Receptores LHRH/fisiologia , Caracteres Sexuais , Testosterona/sangue , Tirosina 3-Mono-Oxigenase/metabolismo , Vasopressinas/metabolismo
10.
Theriogenology ; 80(1): 65-9, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23622940

RESUMO

Slow-release GnRH agonist implants are considered an effective, reversible alternative to surgical castration in male tom cats. Individual differences exist regarding the onset of efficacy and might be delayed in some animals. Single measurements of testosterone (T) might result in basal concentrations also in intact male cats. Consequently, GnRH stimulation tests are performed to measure T increase in intact animals and to differentiate castrated from intact male cats. In this study, five tom cats were treated with a 4.7-mg deslorelin implant and GnRH stimulation tests using buserelin were performed before treatment and at 4-week intervals afterward until Week 20. After the last test in Week 20 all animals were castrated. Four of five animals had basal T after 4 weeks and-in contrast to pretreatment-application of buserelin did not result in any further T increase. In one animal, T was low after implant insertion, but not basal; however, a GnRH stimulation test induced a slight increase of T in Week 8 and 16 only and no response in Weeks 4, 12, and 20. Testicular volume was significantly decreased and penile spines disappeared in all cats. Testicular histology showed mixed atrophy, but also fully elongated spermatids in three of five male cats making infertility questionable. Because of the loss of the stimulatory effect of short-term GnRH application (buserelin), it can be assumed that long-term GnRH agonists also act by some mechanisms of downregulation of pituitary GnRH receptors in the tom cat.


Assuntos
Busserrelina/farmacologia , Gatos , Anticoncepção/veterinária , Testosterona/sangue , Pamoato de Triptorrelina/análogos & derivados , Animais , Castração/veterinária , Anticoncepção/métodos , Regulação para Baixo/efeitos dos fármacos , Implantes de Medicamento , Hormônio Liberador de Gonadotropina/agonistas , Masculino , Hipófise/química , Receptores LHRH/efeitos dos fármacos , Receptores LHRH/fisiologia , Pamoato de Triptorrelina/administração & dosagem
11.
Theor Biol Med Model ; 9: 35, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22870942

RESUMO

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.


Assuntos
Ciclo Menstrual/fisiologia , Modelos Biológicos , Ovulação/fisiologia , Ativinas/fisiologia , Evolução Biológica , Estrogênios/fisiologia , Retroalimentação Fisiológica , Feminino , Hormônio Foliculoestimulante/fisiologia , Humanos , Hipotálamo/fisiologia , Inibinas/fisiologia , Leptina/fisiologia , Hormônio Luteinizante/fisiologia , Luteólise/fisiologia , Masculino , Hipófise/fisiologia , Progesterona/fisiologia , Puberdade/fisiologia , Receptores de Estrogênio/fisiologia , Receptores LHRH/fisiologia , Receptores de Progesterona/fisiologia , Caracteres Sexuais
12.
J Clin Endocrinol Metab ; 97(9): E1798-807, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22745237

RESUMO

CONTEXT: A broad spectrum of GnRH-deficient phenotypes has been identified in individuals with both mono- and biallelic GNRHR mutations. OBJECTIVE: The objective of the study was to determine the correlation between the severity of the reproductive phenotype(s) and the number and functional severity of rare sequence variants in GNRHR. SUBJECTS: Eight hundred sixty-three probands with different forms of GnRH deficiency, 46 family members and 422 controls were screened for GNRHR mutations. The 70 subjects (32 patients and 38 family members) harboring mutations were divided into four groups (G1-G4) based on the functional severity of the mutations (complete or partial loss of function) and the number of affected alleles (monoallelic or biallelic) with mutations, and these classes were mapped on their clinical phenotypes. RESULTS: The prevalence of heterozygous rare sequence variants in GNRHR was significantly higher in probands vs. controls (P < 0.01). Among the G1-G3 groups (homozygous subjects with successively decreasing severity and number of mutations), the hypogonadotropic phenotype related to their genetic load. In contrast, subjects in G4, with only monoallelic mutations, demonstrated a greater diversity of clinical phenotypes. CONCLUSIONS: In patients with GnRH deficiency and biallelic mutations in GNRHR, genetic burden defined by severity and dose is associated with clinical phenotype. In contrast, for patients with monoallelic GNRHR mutations this correlation does not hold. Taken together, these data indicate that as-yet-unidentified genetic and/or environmental factors may combine with singly mutated GNRHR alleles to produce reproductive phenotypes.


Assuntos
Carga Genética , Receptores LHRH/genética , Receptores LHRH/fisiologia , Adolescente , Adulto , Amenorreia/genética , DNA/genética , Análise Mutacional de DNA , Etnicidade , Feminino , Hormônio Liberador de Gonadotropina/deficiência , Hormônio Liberador de Gonadotropina/genética , Humanos , Hipogonadismo/genética , Doenças Hipotalâmicas/genética , Masculino , Mutação/genética , Fenótipo , Puberdade Tardia/genética , Adulto Jovem
13.
Mini Rev Med Chem ; 12(9): 841-50, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22681250

RESUMO

Gonadotropin-releasing hormone is a neuropeptide that acts via Gq coupled G-protein coupled receptors in the pituitary that mediate central control of reproduction. GnRH receptors (GnRHR) and GnRH ligands are also found in extra-pituitary sites including the CNS as well as reproductive tissues and cancer cells derived from such tissues. Much of the interest in the extra-pituitary receptors stems from the fact that they mediate anti-proliferative and/or pro-apoptotic effects and may therefore be directly targeted for cancer therapy. Type I mammalian GnRHR are atypical in that they do not bind to (or signal via) arrestins. In spite of this restriction on their signaling repertoire, there is good evidence for existence of multiple active GnRHR conformations and for activation of multiple upstream effectors (heterotrimeric and monomeric G-proteins). In this review GnRHR signaling is described, with emphasis on the relevance of functional selectivity for pharmacological characterization of GnRHR ligands, as well as its possible contribution to contextdependent GnRHR signaling and relevance for GnRHR-mediated effects on cell fate as well as GnRHR trafficking.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Receptores LHRH/fisiologia , Transdução de Sinais/fisiologia , Animais , Antineoplásicos Hormonais/farmacologia , Linhagem Celular , Sistema Nervoso Central/fisiologia , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Ligantes , Neoplasias/tratamento farmacológico , Hipófise/fisiologia , Conformação Proteica , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Receptores LHRH/agonistas , Receptores LHRH/antagonistas & inibidores , Reprodução/fisiologia , Transdução de Sinais/efeitos dos fármacos
14.
Theriogenology ; 77(7): 1442-52, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22284221

RESUMO

The role of LH in luteolysis and development of the ovulatory follicle and the involvement of GnRH receptors in estradiol (E2) stimulation of LH secretion were studied in heifers. A pulse of PGF(2α), as indicated by a metabolite, was induced by E2 treatment on Day 15 (Day 0 = ovulation) and LH concentration was reduced with a GnRH-receptor antagonist (acyline) on Days 15, 16, and 17. Blood samples were collected every 6 h on Days 14-17 and hourly for 10 h beginning at the Day-15 treatments. Four groups were used (n = 6): control, acyline, E2, and E2/acyline. The number of LH pulses/heifer during the 10 h posttreatment was greater (P < 0.0002) in the E2 group (2.3 ± 0.4, mean ± SEM) than in the acyline group (0.2 ± 0.2) and was intermediate in the E2/acyline group (1.4 ± 0.2). Concentrations of progesterone in samples collected every 6 h on Day 15 showed a group-by-hour interaction (P < 0.02); concentrations decreased in the acyline group but not in the control group. The 12 heifers in the combined acyline and E2/acyline groups had three follicular waves compared to two waves in 10 of 12 heifers in the combined control and E2 groups. Results (1) supported the hypothesis that LH delays the progesterone decrease associated with luteolysis, (2) supported the hypothesis that LH has a positive effect on the continued development and growth of the selected ovulatory follicle, and (3) indicated that E2 stimulates LH production through an intracellular pathway that involves GnRH receptors on the gonadotropes and a pathway that does not involve the receptors.


Assuntos
Bovinos/fisiologia , Estradiol/farmacologia , Hormônio Luteinizante/fisiologia , Luteólise/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Animais , Feminino , Hormônio Luteinizante/metabolismo , Ovulação/fisiologia , Progesterona/sangue , Receptores LHRH/fisiologia
15.
Gen Comp Endocrinol ; 175(3): 398-406, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22138555

RESUMO

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.


Assuntos
Bass/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Ácido Pirrolidonocarboxílico/análogos & derivados , Retina/fisiologia , Transdução de Sinais/fisiologia , Animais , Masculino , Receptores LHRH/fisiologia , Células Ganglionares da Retina/fisiologia , Tirosina 3-Mono-Oxigenase/fisiologia
16.
Gen Comp Endocrinol ; 176(3): 321-6, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22172342

RESUMO

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.


Assuntos
Aplysia/fisiologia , Sistema Nervoso Central/fisiologia , Evolução Molecular , Hormônio Liberador de Gonadotropina/fisiologia , Receptores LHRH/fisiologia , Sequência de Aminoácidos , Animais , Aplysia/genética , Hormônio Liberador de Gonadotropina/genética , Dados de Sequência Molecular , Filogenia , Receptores LHRH/genética , Alinhamento de Sequência
17.
Curr Opin Obstet Gynecol ; 23(4): 238-44, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21666463

RESUMO

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.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Implantação do Embrião/efeitos dos fármacos , Feminino , Hormônio Liberador de Gonadotropina/agonistas , Antagonistas de Hormônios/farmacocinética , Antagonistas de Hormônios/farmacologia , Humanos , Masculino , Indução da Ovulação , Receptores LHRH/fisiologia , Transdução de Sinais/fisiologia
18.
Fertil Steril ; 95(2): 465-73, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20888559

RESUMO

OBJECTIVE: To review the physiologic functions and clinical significance of peripheral GnRH receptors. DESIGN: Literature review. All peer-reviewed journal articles published before 2010 on peripheral GnRH receptors were searched for in the Pubmed database, and relevant findings were summarized. RESULT(S): Peripheral GnRH/GnRH receptor systems may serve as regulators of hCG synthesis and implantation, and play crucial roles in antiproliferation and apoptosis. Currently, GnRH agonists have been used in cancer treatment and ovary protection during chemotherapy, taking advantage of the local direct effect mediated by peripheral GnRH receptors. CONCLUSION(S): The ubiquitous GnRH/GnRH receptor system in human tissues has been shown to have some important physiologic functions. Further research to clarify functions of these peripheral GnRH receptors may lead to discovery of new therapeutic options.


Assuntos
Genitália Feminina/metabolismo , Receptores LHRH/fisiologia , Reprodução/fisiologia , Animais , Sistema Nervoso Central/metabolismo , Gonadotropina Coriônica/metabolismo , Feminino , Genitália Feminina/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/fisiologia , Hormônio Liberador de Gonadotropina/uso terapêutico , Humanos , Modelos Biológicos , Receptores LHRH/genética , Receptores LHRH/metabolismo , Reprodução/genética
19.
Gen Comp Endocrinol ; 170(3): 424-48, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21074534

RESUMO

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.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Ovário/fisiologia , Trichosurus/fisiologia , Animais , Ciclo Estral , Feminino , Hormônio Liberador de Gonadotropina/fisiologia , Espécies Introduzidas , Folículo Ovariano/efeitos dos fármacos , Ovulação/fisiologia , RNA Mensageiro/metabolismo , Receptores LHRH/fisiologia
20.
CNS Neurol Disord Drug Targets ; 9(5): 651-60, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20632963

RESUMO

Receptors for hormones of the hypothalamic-pituitary-gonadal axis are expressed throughout the brain. Age-related decline in gonadal reproductive hormones cause imbalances of this axis and many hormones in this axis have been functionally linked to neurodegenerative pathophysiology. Gonadotropin-releasing hormone (GnRH) plays a vital role in both central and peripheral reproductive regulation. GnRH has historically been known as a pituitary hormone; however, in the past few years, interest has been raised in GnRH actions at non-pituitary peripheral targets. GnRH ligands and receptors are found throughout the brain where they may act to control multiple higher functions such as learning and memory function and feeding behavior. The actions of GnRH in mammals are mediated by the activation of a unique rhodopsin-like G protein-coupled receptor that does not possess a cytoplasmic carboxyl terminal sequence. Activation of this receptor appears to mediate a wide variety of signaling mechanisms that show diversity in different tissues. Epidemiological support for a role of GnRH in central functions is evidenced by a reduction in neurodegenerative disease after GnRH agonist therapy. It has previously been considered that these effects were not via direct GnRH action in the brain, however recent data has pointed to a direct central action of these ligands outside the pituitary. We have therefore summarized the evidence supporting a central direct role of GnRH ligands and receptors in controlling central nervous physiology and pathophysiology.


Assuntos
Envelhecimento/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Gônadas/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Doenças Neurodegenerativas/fisiopatologia , Receptores LHRH/fisiologia , Envelhecimento/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Hormônio Liberador de Gonadotropina/metabolismo , Gônadas/metabolismo , Gônadas/fisiopatologia , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Receptores LHRH/efeitos dos fármacos , Receptores LHRH/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA