Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Gen Comp Endocrinol ; 310: 113819, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34015343

RESUMO

Gonadotropin-Releasing Hormone (GnRH), acting via the GnRH receptor (GnRHR), and a member of G-protein coupled receptor (GPCR), plays an essential role in the control of reproduction while operating primarily at the hypothalamic level of the gonadotropic axis. GnRH and its receptor are co-expressed in certain specific cells, suggesting an autocrine regulation of such cells. In the male reproductive system, two forms of GnRH (I and II) and its receptors (GnRHR) are present in the human and non-human primate (NHP) testis, prostate, epididymis, seminal vesicle, and human spermatozoa. In humans, the GnRHR-II receptor gene is disrupted by a frameshift in exon 1 and a stop codon in exon 2, rendering the receptor non-functional, whereas a fully functional GnRHR-II receptor is present in New-World and Old-World monkeys. There is no evidence of the existence of a GnRH receptor in NHP sperm. Since the NHP has a phylogenetic relationship to man and is often used as models in reproductive physiology, this present study aimed to determine GnRHR-I and GnRHR-II in Vervet monkey (Chlorocebus aethiops) spermatozoa. A total of 24 semen samples were obtained from four adult Vervet monkeys through electro-ejaculation and utilized for genotyping and gene expression analysis of GnRHR-I and II. Here we report that both receptors were successfully identified in the Vervet monkey sperm with the abundance of GnRHR-I gene expression compared to GnRHR-II. In comparison to the human, there is no evidence of such a stop codon at position 179 in exon 2 of the Vervet GnRHR-II. These findings suggest that both receptors are transcriptionally functional in Vervet spermatozoa.


Assuntos
Hormônio Liberador de Gonadotropina , Receptores LHRH , Animais , Chlorocebus aethiops , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Masculino , Filogenia , Receptores LHRH/genética , Receptores LHRH/metabolismo , Espermatozoides/metabolismo
2.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33805020

RESUMO

The proper expression of gonadotropin-releasing hormone receptors (GnRHRs) by pituitary gonadotropes is critical for maintaining maximum reproductive capacity. GnRH receptor expression must be tightly regulated in order to maintain the normal pattern of expression through the estrous cycle in rodents, which is believed to be important for interpreting the finely tuned pulses of GnRH from the hypothalamus. Much work has shown that Gnrhr expression is heavily regulated at the level of transcription. However, researchers have also discovered that Gnrhr is regulated post-transcriptionally. This review will discuss how RNA-binding proteins and microRNAs may play critical roles in the regulation of GnRHR expression. We will also discuss how these post-transcriptional regulators may themselves be affected by metabolic cues, specifically with regards to the adipokine leptin. All together, we present evidence that Gnrhr is regulated post-transcriptionally, and that this concept must be further explored in order to fully understand the complex nature of this receptor.


Assuntos
Regulação da Expressão Gênica , Hipotálamo/metabolismo , Receptores LHRH/metabolismo , Reprodução , Regiões 3' não Traduzidas , Adipocinas/metabolismo , Animais , Estro , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Leptina/metabolismo , Camundongos , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Ratos , Receptores LHRH/genética , Transcrição Gênica
3.
Sci Rep ; 10(1): 10579, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601341

RESUMO

Molecules that correct the folding of protein mutants, restoring their functional trafficking, are called pharmacoperones. Most are clinically irrelevant and possess intrinsic antagonist or agonist activity. Here, we identify compounds capable of rescuing the activity of mutant gonadotropin-releasing hormone receptor or GnRHR which, is sequestered within the cell and if dysfunctional leads to Hypogonadotropic Hypogonadism. To do this we screened the E90K GnRHR mutant vs. a library of 645,000 compounds using a cell-based calcium detection system. Ultimately, we identified 399 compounds with EC50 ≤ 5 µM with no effect in counterscreen assays. Medicinal chemistry efforts confirmed activity of 70 pure samples and mode of action studies, including radioligand binding, inositol phosphate, and toxicity assays, proved that we have a series of tractable compounds that can be categorized into structural clusters. These early lead molecules rescue mutant GnRHR function and are neither agonist nor antagonists of the GnRHR cognate receptor, a feature required for potential clinical utility.


Assuntos
Receptores LHRH/agonistas , Receptores LHRH/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Cálcio/metabolismo , Avaliação Pré-Clínica de Medicamentos , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/metabolismo , Células HeLa , Ensaios de Triagem em Larga Escala , Humanos , Fosfatos de Inositol/metabolismo , Mutação , Dobramento de Proteína , Transporte Proteico , Receptores LHRH/genética
4.
Poult Sci ; 98(9): 4172-4181, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31001634

RESUMO

Precise natural anti-oxidative compounds have facilitated the research of infertile gametes and the development of novel bio-therapeutics, especially the molecules that are based on the reduction of oxidative stress, such as L-carnitine (LC). In addition to, the defect in the functioning of sperm mitochondrial and the decreasing seminal antioxidant ability due to aging, its essential role in permitting the mitochondrial import and oxidation of long chain fatty acids is worthy. Therefore, current study was designed to investigate the effects of dietary LC on semen quality, seminal antioxidant activity, and their implications for the fertility in aged cocks for 12 wk. Supplementation of the feed with two different doses of LC (50 and 150 mg/kg body weight/day) for 12 wk showed significantly increased in the reproductive activity of cock, in comparison to the control group. Seminal analysis showed that supplementation of LC significantly increased (P < 0.05) the sperm motility, concentration, livability, semen quality factor, seminal malondialdehyde concentration, catalase, and glutathione peroxidase activities. In addition, addition of LC significantly increased (P < 0.05) the plasma concentration of testosterone and prostaglandin E2 but posed no significant effect on the concentration of follicle-stimulating hormone. Furthermore, the findings of artificial insemination showed significant increased (P < 0.05) in the percentage of fertility in LC groups, while the percentage hatchability and mortality remained unchanged. Immunohistochemistry analysis revealed that LC significantly increased (P < 0.05) the testicular immunopositivity of MT1 and MT2. Moreover, the administration of LC to the aged cocks enhanced (P < 0.05) GnRH1 and GnRHR mRNA levels when compared with untreated cocks. The results of the present study suggest that LC treatment of aged cocks increases the seminal antioxidant enzymes and sexual hormones levels, which may improve the semen quality by increasing the expression of GnRH1 and melatonin receptors (MT1 and MT2) activities. Collectively, LC could be a suitable feed supplementation to increase reproductive activities through enhancing semen quality in aging cocks.


Assuntos
Antioxidantes/metabolismo , Proteínas Aviárias/genética , Carnitina/metabolismo , Galinhas/fisiologia , Expressão Gênica/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Ração Animal/análise , Animais , Antioxidantes/administração & dosagem , Proteínas Aviárias/metabolismo , Carnitina/administração & dosagem , Dieta/veterinária , Suplementos Nutricionais/análise , Relação Dose-Resposta a Droga , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Masculino , Receptor MT1 de Melatonina/genética , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/genética , Receptor MT2 de Melatonina/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo , Análise do Sêmen/veterinária , Espermatozoides/fisiologia , Testículo/metabolismo
5.
BMC Endocr Disord ; 18(1): 30, 2018 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-29793475

RESUMO

BACKGROUND: This study aimed to detect changes in hormone levels in the hypothalamic-pituitary-ovarian axis in Sprague-Dawley (SD) rats with hypothyroidism, and identify differences in the pregnancy and abortion rates of female adult rats. The potential role of gonadotropin releasing hormone (GnRH) as the link between the hypothalamic-pituitary-ovarian axis and reproductive function regulated by thyroid hormones was also investigated. METHODS: Female SD rats (n = 136) were causally classified into two groups: the normal-drinking-water group (n = 60) and the 0.05% propylthiouracil-drinking-water group (PTU 2 mg/kg/day, n = 76) to establish an adult rat model of hypothyroidism (6 weeks). Female and male rats at a ratio of 1:2 were used to establish a hypothyroidism pregnancy model. GnRH mRNA and GnRH receptor (GnRHR) expression in rats was detected using real time quantitative PCR(qRT-PCR) and immunohistochemistry, respectively. RESULTS: The abortion rate differed significantly between the hypothyroidism pregnancy group and the normal pregnancy group (P < 0.05). No significant differences were found in the distribution of the GnRHR among the five nuclei (hypothalamic arcuate nucleus, hypothalamic ventromedial nucleus, hypothalamic anterior nucleus, paraventricular nucleus of the hypothalamus, and ventral premammillary nucleus) of the hypothalamus and ovary (P > 0.05). Hypothyroidism had no significant effect on GnRH mRNA expression in the hypothalamic-pituitary-ovarian axis in the four groups (normal control group, normal pregnancy group, hypothyroidism pregnancy group, and hypothyroidism group) (P > 0.05). CONCLUSIONS: Hypothyroidism had an adverse impact on pregnancy in rats and may affect the distribution of pituitary GnRHR, whereas it did not obviously affect the distribution of GnRHR in the nuclei of the hypothalamus and ovary. Hypothyroidism had no effect on GnRH mRNA expression.


Assuntos
Hipotálamo/patologia , Hipotireoidismo/complicações , Infertilidade Feminina/etiologia , Ovário/patologia , Hipófise/patologia , Reprodução , Animais , Biomarcadores/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hipotireoidismo/metabolismo , Hipotireoidismo/patologia , Infertilidade Feminina/diagnóstico , Infertilidade Feminina/metabolismo , Masculino , Ovário/metabolismo , Hipófise/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores LHRH/genética , Receptores LHRH/metabolismo
6.
Endocrinology ; 159(3): 1496-1510, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29409045

RESUMO

Gonadotropin-releasing hormone (GnRH) from the hypothalamus regulates synthesis and secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) from the anterior pituitary gonadotropes. LH and FSH are heterodimers composed of a common α-subunit and unique ß-subunits, which provide biological specificity and are limiting components of mature hormone synthesis. Gonadotrope cells respond to GnRH via specific expression of the GnRH receptor (Gnrhr). GnRH induces the expression of gonadotropin genes and of the Gnrhr by activation of specific transcription factors. The JUN (c-Jun) transcription factor binds to AP-1 sites in the promoters of target genes and mediates induction of the FSHß gene and of the Gnrhr in gonadotrope-derived cell lines. To analyze the role of JUN in reproductive function in vivo, we generated a mouse model that lacks JUN specifically in GnRH receptor‒expressing cells (conditional JUN knockout; JUN-cKO). JUN-cKO mice displayed profound reproductive anomalies such as reduced LH levels resulting in lower gonadal steroid levels, longer estrous cycles in females, and diminished sperm numbers in males. Unexpectedly, FSH levels were unchanged in these animals, whereas Gnrhr expression in the pituitary was reduced. Steroidogenic enzyme expression was reduced in the gonads of JUN-cKO mice, likely as a consequence of reduced LH levels. GnRH receptor‒driven Cre activity was detected in the hypothalamus but not in the GnRH neuron. Female, but not male, JUN-cKO mice exhibited reduced GnRH expression. Taken together, our results demonstrate that GnRH receptor‒expression levels depend on JUN and are critical for reproductive function.


Assuntos
Gonadotrofos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores LHRH/metabolismo , Reprodução , Animais , Feminino , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hipófise/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Receptores LHRH/genética
7.
J Toxicol Sci ; 42(1): 13-23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28070105

RESUMO

Our previous studies demonstrated that treating pregnant rats with dioxins, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), targets the pituitary expression of luteinizing hormone (LH) to attenuate testicular steroidogenesis in fetuses, resulting in the imprinting of sexual immaturity of the offspring after reaching maturity. Furthermore, we found that although TCDD disturbs the tricarboxylic acid (TCA) cycle in the fetal hypothalamus, maternal co-treatment with α-lipoic acid (α-LA), a cofactor of the TCA cycle, restores a TCDD-produced reduction in the LH-evoked steroidogenesis as well as the TCA cycle activity in fetuses. However, the mechanism underlying the beneficial effect of α-LA remains to be fully elucidated. To address this issue, we compared the effect of α-LA with that of thiamine, another cofactor of the TCA cycle. As with α-LA, supplying thiamine to dams exposed to TCDD alleviates the reduced level of not only hypothalamic ATP but also pituitary LH and testicular steroidogenic protein in fetuses. However, thiamine had a much weaker effect than α-LA. In agreement with ATP attenuation, TCDD activated AMP-activated protein kinase (AMPK), a negative regulator of LH production, whereas the supplementation of α-LA allowed recovery from this defect. Furthermore, α-LA restored the TCDD-produced reduction in the pituitary expression of the receptor for gonadotropin-releasing hormone (GnRH), an upstream regulator of LH synthesis. These results suggest that α-LA rescues TCDD-produced attenuation during fetal steroidogenesis due not only to facilitation of energy production through the TCA cycle but also through suppression of AMPK activation, and the pituitary GnRH receptor may serve as a mediator of these effects.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Encéfalo/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Receptores LHRH/genética , Ácido Tióctico/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/metabolismo , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Hormônio Liberador de Gonadotropina/genética , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Hormônio Luteinizante/sangue , Hormônio Luteinizante/genética , Masculino , Miocárdio/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos Wistar , Testículo/efeitos dos fármacos , Testículo/metabolismo
8.
Mol Cell Endocrinol ; 437: 62-74, 2016 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-27519634

RESUMO

Sporadic epidemics and several researches in rodents indicated that zearalenone (ZEA) and its metabolites, the prevailing oestrogenic mycotoxins in foodstuffs, were a triggering factor for true precocious puberty development in girls. Nevertheless, the neuroendocrine mechanism through which ZEA mycoestrogens advance puberty onset is not fully understood. To elucidate this issue, hypothalamic kisspeptin-G-protein coupled receptor-54 (GPR54) signaling pathway that regulates the onset of puberty was focused on in the present study. Immature female SD rats were given a daily intragastric administration of corn oil (vehicle control), 50 µg/kg body weight (bw) of 17ß-estradiol (E2, positive control), and 3 doses (0.2, 1 and 5 mg/kg bw) of ZEA for consecutive 5 days starting from postnatal day 15, respectively. Puberty onset was evaluated by detecting the physiological and hormonal responses, and hypothalamic kisspeptin-GPR54 pathway was determined to reveal the neuroendocrine mechanism. As the markers of puberty onset, vaginal opening was significantly accelerated and uterine weight was increased in both E2 and 5 mg/kg ZEA groups. Serum levels of follicle stimulating hormone, luteinizing hormone and estradiol were also markedly elevated by E2 and 5 mg/kg ZEA, which is compatible with the changes in peripheral reproductive organs. The mRNA and protein expressions of hypothalamic gonadotropin-releasing hormone (GnRH) were both obviously elevated by E2 and 5 mg/kg ZEA. GnRH expression changes occurred in parallel with increased expressions of hypothalamic Kiss1 and its receptor GPR54 at both mRNA and protein levels. Most of these changes were also noted in 1 mg/kg ZEA group, but none in 0.2 mg/kg group. Therefore, within the context of this study, the No Observed Adverse Effect Level (NOAEL) for ZEA in terms of oestrogenic activity and puberty-promoting effect in immature female rats was considered to be 0.2 mg/kg bw per day, and the Lowest Observed Adverse Effect Level (LOAEL) was 1 mg/kg bw per day. In conclusion, prepubertal exposure to dietary relevant levels of ZEA induced central precocious puberty in female rats by premature activation of hypothalamic kisspeptin-GPR54-GnRH signaling pathway, followed by the stimulation of gonadotropins release at an earlier age, resulting in the advancement of vaginal opening and enlargement of uterus at periphery.


Assuntos
Estrogênios/toxicidade , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Micotoxinas/toxicidade , Puberdade Precoce/induzido quimicamente , Receptores Acoplados a Proteínas G/metabolismo , Maturidade Sexual/efeitos dos fármacos , Zearalenona/toxicidade , Animais , Ciclo Estral/efeitos dos fármacos , Feminino , Genitália Feminina/efeitos dos fármacos , Genitália Feminina/crescimento & desenvolvimento , Genitália Feminina/patologia , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hormônios/sangue , Hipotálamo/efeitos dos fármacos , Masculino , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Puberdade Precoce/sangue , Puberdade Precoce/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de Kisspeptina-1 , Receptores LHRH/genética , Receptores LHRH/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
PLoS One ; 11(7): e0160292, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27467252

RESUMO

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.


Assuntos
Evolução Biológica , Receptores LHRH/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Clonagem Molecular , DNA Complementar/genética , Gastrópodes , Filogenia , Ensaio Radioligante , Receptores LHRH/genética , Receptores LHRH/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
10.
Gen Comp Endocrinol ; 194: 94-101, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24055558

RESUMO

Kisspeptin signaling through its receptor is crucial for many reproductive functions. However, the molecular mechanisms and biomedical significance of the regulation of GnRH neurons by kisspeptin have not been adequately elucidated. In the present study, we found that kisspeptin increases GnRH receptor (GnRHR) expression in a GnRH-producing cell line (GT1-7). Because cellular activity of G protein-coupled receptor 54 (GPR54) and GnRHR was limited in GT1-7 cells, we overexpressed these receptors to clarify receptor function. Using luciferase reporter constructs, the activity of both the serum response element (Sre) promoter, a target for extracellular signal-regulated kinase (ERK), and the cyclic AMP (cAMP) response element (Cre) promoter were increased by kisspeptin. Although GnRH increased Sre promoter activity, the Cre promoter was not significantly activated by GnRH. Kisspeptin, but not GnRH, increased cAMP accumulation in these cells. Kisspeptin also increased the transcriptional activity of GnRHR; however, the effect of GnRH on the GnRHR promoter was limited and not significant. Transfection of GT1-7 cells with constitutively active MEK kinase (MEKK) and protein kinase A (PKA) increased GnRHR expression. In addition, GnRHR expression was further increased by co-overexpression of MEKK and PKA. The Cre promoter, but not the Sre promoter, was also further activated by co-overexpression of MEKK and PKA. GnRH significantly increased the activity of the GnRHR promoter in the presence of cAMP. The present findings suggest that kisspeptin is a potent stimulator of GnRHR expression in GnRH-producing neurons in association with ERK and the cAMP/PKA pathways.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores LHRH/metabolismo , Animais , Linhagem Celular , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Camundongos , Regiões Promotoras Genéticas/genética , Receptores Acoplados a Proteínas G/genética , Receptores LHRH/genética
11.
Endocrinology ; 154(6): 2114-28, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23554453

RESUMO

The sulfated neurosteroids pregnenolone sulfate (Δ(5)PS) and dehydroepiandrosterone sulfate (DHEAS) are known to play a role in the control of reproductive behavior. In the frog Pelophylax ridibundus, the enzyme hydroxysteroid sulfotransferase (HST), responsible for the biosynthesis of Δ(5)PS and DHEAS, is expressed in the magnocellular nucleus and the anterior preoptic area, two hypothalamic regions that are richly innervated by GnRH1-containing fibers. This observation suggests that GnRH1 may regulate the formation of sulfated neurosteroids to control sexual activity. Double labeling of frog brain slices with HST and GnRH1 antibodies revealed that GnRH1-immunoreactive fibers are located in close vicinity of HST-positive neurons. The cDNAs encoding 3 GnRH receptors (designated riGnRHR-1, -2, and -3) were cloned from the frog brain. RT-PCR analyses revealed that riGnRHR-1 is strongly expressed in the hypothalamus and the pituitary whereas riGnRHR-2 and -3 are primarily expressed in the brain. In situ hybridization histochemistry indicated that GnRHR-1 and GnRHR-3 mRNAs are particularly abundant in preoptic area and magnocellular nucleus whereas the concentration of GnRHR-2 mRNA in these 2 nuclei is much lower. Pulse-chase experiments using tritiated Δ(5)P and DHEA as steroid precursors, and 3'-phosphoadenosine 5'-phosphosulfate as a sulfonate moiety donor, showed that GnRH1 stimulates, in a dose-dependent manner, the biosynthesis of Δ(5)PS and DHEAS in frog diencephalic explants. Because Δ(5)PS and DHEAS, like GnRH, stimulate sexual activity, our data strongly suggest that some of the behavioral effects of GnRH could be mediated via the modulation of sulfated neurosteroid production.


Assuntos
Sulfato de Desidroepiandrosterona/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Pregnenolona/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Perfilação da Expressão Gênica , Hormônio Liberador de Gonadotropina/farmacologia , Hibridização In Situ , Masculino , Microscopia Confocal , Dados de Sequência Molecular , Neurônios/metabolismo , Hipófise/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ranidae , Receptores LHRH/genética , Receptores LHRH/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Sulfotransferases/metabolismo
12.
Reprod Domest Anim ; 47(1): 44-52, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21595758

RESUMO

This study was performed to determine the effect of intracerebroventricular (icv) injection of interleukin (IL)-1ß on the gene expression, translation and release of gonadotropin-releasing hormone (GnRH) and the GnRH receptor (GnRHR) gene expression in the hypothalamus of anestrous ewes. In the anterior pituitary gland (AP), the expression of genes encoding: GnRHR, ß subunits of luteinizing hormone (LH) and folliculotropic hormone (FSH) was determined as well as the effect of IL-1ß on pituitary gonadotropins release. The relative mRNA level was determined by real-time PCR, GnRH concentration in the cerebrospinal fluid (CSF) was assayed by ELISA and the plasma concentration of LH and FSH were determined by radioimmunoassay. Our results showed that icv injection of IL-1ß (10 or 50 µg/animal) decreased the GnRH mRNA level in the pre-optic area (POA) (35% and 40% respectively; p ≤ 0.01) and median eminence (ME) (75% and 70% respectively; p ≤ 0.01) and GnRHR gene expression in ME (55% and 50% respectively; p ≤ 0.01). A significant decrease in GnRHR mRNA level in the AP in the group treated with the 50 µg (60%; p ≤ 0.01) but not with the 10 µg dose was observed. The centrally administrated IL-1ß lowered also GnRH concentration in the CSF (60%; p ≤ 0.01) and reduced the intensity of GnRH translation in the POA (p ≤ 0.01). It was not found any effect of icv IL-1ß injection upon the release of LH and FSH. However, the central injection of IL-1ß strongly decreased the LHß mRNA level (41% and 50%; p ≤ 0.01; respectively) and FSHß mRNA in the case of the 50 µg dose (49%; p ≤ 0.01) in the pituitary of anestrous ewes. These results demonstrate that the central IL-1ß is an important modulator of the GnRH biosynthesis and release during immune/inflammatory challenge.


Assuntos
Anestro/fisiologia , Hipotálamo/efeitos dos fármacos , Interleucina-1beta/administração & dosagem , Ovário/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Ovinos/fisiologia , Animais , Feminino , Hormônio Foliculoestimulante/sangue , Subunidade beta do Hormônio Folículoestimulante/genética , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas Hipofisárias/metabolismo , Hipotálamo/metabolismo , Injeções Intraventriculares/veterinária , Hormônio Luteinizante/sangue , Hormônio Luteinizante Subunidade beta/genética , Ovário/metabolismo , Hipófise/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , RNA Mensageiro/análise , Receptores LHRH/genética
13.
J Endocrinol ; 212(3): 353-61, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22159505

RESUMO

The neurotransmitter galanin has been implicated in the steroidogenic regulation of reproduction based on work mainly conducted in rodents. This study investigated the temporal changes in the expression of galanin and its three receptor isoforms and GNRH and GNRHR mRNA in specific hypothalamic nuclei known to be involved in the regulation of reproductive cyclicity, namely the medial pre-optic area (mPOA), the rostral mPOA/organum vasculosum of the lamina terminalis, the paraventricular nucleus and the arcuate nucleus using an ovine model. Following synchronisation of their oestrous cycles, tissues were collected from ewes at five time points: the early follicular, mid follicular (MF) and late follicular phases and the early luteal and mid luteal phases. The results indicated significant differences in regional expression of most of the genes studied, with galanin mRNA expression being highest during the MF phase at the start of the GNRH/LH surge and the expression of the three galanin receptor (GalR) isoforms and GNRH and its receptor highest during the luteal phase. These findings are consistent with a role for galanin in the positive feedback effects of oestradiol (E(2)) on GNRH secretion and a role for progesterone induced changes in the pattern of expression of GalRs in the regulation of the timing of E(2)'s positive feedback through increased sensitivity of galanin-sensitive systems to secreted galanin.


Assuntos
Ciclo Estral/metabolismo , Galanina/genética , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Receptores de Galanina/genética , Ovinos/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/fisiologia , Sincronização do Estro , Retroalimentação Fisiológica , Feminino , Fase Folicular/metabolismo , Expressão Gênica , Hipotálamo/química , Fase Luteal/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Área Pré-Óptica/metabolismo , Progesterona/fisiologia , RNA Mensageiro/análise , Receptor Tipo 1 de Galanina/genética , Receptor Tipo 2 de Galanina/genética , Receptor Tipo 3 de Galanina/metabolismo , Receptores LHRH/genética
14.
ChemMedChem ; 6(11): 2070-80, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21953839

RESUMO

AG-045572 (CMPD1, 1 a) is a nonpeptidic gonadotropin-releasing hormone (GnRH) antagonist that has been investigated for the treatment of sex hormone-related diseases. In the context of systematic studies on sila-substituted drugs, the silicon analogue disila-AG-045572 (1 b) and its derivative 2 were prepared in multi-step syntheses and characterized by elemental analyses (C, H, N), NMR spectroscopic studies (1H, 13C, 29Si), and single-crystal X-ray diffraction. The pharmacological properties of compounds 1 a, 1 b, and 2 were compared in terms of their in vitro potency at cloned human and rat GnRH receptors. Compounds 1 a and 2 were also examined in regard to their pharmacokinetics and in vivo efficacy in both castrated rat (luteinizing hormone (LH) suppression) and intact rat (testosterone suppression) models. The efficacy and pharmacokinetic profiles of 1 a and its silicon-containing analogue 2 appear similar, indicating that replacement of the 5,6,7,8-tetrahydronaphthalene ring system by the 1,3-disilaindane skeleton led to retention of efficacy. Therefore, the silicon compound 2 represents a novel drug prototype for the design of potent, orally available GnRH antagonists suitable for once-daily dosing.


Assuntos
Furanos/química , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/química , Antagonistas de Hormônios/farmacologia , Tetra-Hidronaftalenos/química , Animais , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Furanos/farmacologia , Antagonistas de Hormônios/farmacocinética , Humanos , Hormônio Luteinizante/antagonistas & inibidores , Espectroscopia de Ressonância Magnética , Masculino , Estrutura Molecular , Orquiectomia , Ratos Wistar , Receptores LHRH/genética , Silício/química , Relação Estrutura-Atividade , Tetra-Hidronaftalenos/farmacologia
15.
Endocrinology ; 152(4): 1515-26, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21303944

RESUMO

GnRH signaling regulates reproductive physiology in vertebrates via the hypothalamic-pituitary-gonadal axis. In addition, GnRH signaling has been postulated to act on the brain. However, elucidating its functional role in the central nervous system has been hampered because of the difficulty in identifying direct GnRH signaling targets in live brain tissue. Here we used a binary genetic strategy to visualize GnRH receptor (GnRHR) neurons in the mouse brain and started to characterize these cells. First, we expressed different fluorescent proteins in GnRHR neurons and mapped their precise distribution throughout the brain. Remarkably, neuronal GnRHR expression was only initiated after postnatal day 16, suggesting peri- and postpubertal functions of GnRH signaling in this organ. GnRHR neurons were found in different brain areas. Many GnRHR neurons were identified in areas influencing sexual behaviors. Furthermore, GnRHR neurons were detected in brain areas that process olfactory and pheromonal cues, revealing one efferent pathway by which the neuroendocrine hypothalamus may influence the sensitivity towards chemosensory cues. Using confocal Ca(2+) imaging in brain slices, we show that GnRHR neurons respond reproducibly to extracellular application of GnRH or its analog [D-TRP(6)]-LH-RH, indicating that these neurons express functional GnRHR. Interestingly, the duration and shape of the Ca(2+) responses were similar within and different between brain areas, suggesting that GnRH signaling may differentially influence brain functions to affect reproductive success. Our new mouse model sets the stage to analyze the next level of GnRH signaling in reproductive physiology and behavior.


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Receptores LHRH/metabolismo , Animais , Feminino , Imunofluorescência , Hipotálamo/citologia , Hipotálamo/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Condução Nervosa/genética , Condução Nervosa/fisiologia , Odorantes , Feromônios/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA não Traduzido , Receptores LHRH/genética , Comportamento Sexual Animal/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Tálamo/citologia , Tálamo/metabolismo
16.
Proc Natl Acad Sci U S A ; 107(37): 16372-7, 2010 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-20805495

RESUMO

Gonadotropin-releasing hormone (GnRH) signaling regulates reproductive physiology in mammals. GnRH is released by a subset of hypothalamic neurons and binds to GnRH receptor (GnRHR) on gonadotropes in the anterior pituitary gland to control production and secretion of gonadotropins that in turn regulate the activity of the gonads. Central control of reproduction is well understood in adult animals, but GnRH signaling has also been implicated in the development of the reproductive axis. To investigate the role of GnRH signaling during development, we selectively ablated GnRHR-expressing cells in mice. This genetic strategy permitted us to identify an essential stage in male reproductive axis development, which depends on embryonic GnRH signaling. Our experiments revealed a striking dichotomy in the gonadotrope population of the fetal anterior pituitary gland. We show that luteinizing hormone-expressing gonadotropes, but not follicle-stimulating hormone-expressing gonadotropes, express the GnRHR at embryonic day 16.75. Furthermore, we demonstrate that an embryonic increase in luteinizing hormone secretion is needed to promote development of follicle-stimulating hormone-expressing gonadotropes, which might be mediated by paracrine interactions within the pituitary. Moreover, migration of GnRH neurons into the hypothalamus appeared normal with appropriate axonal connections to the median eminence, providing genetic evidence against autocrine regulation of GnRH neurons. Surprisingly, genetic ablation of GnRHR expressing cells significantly increased the number of GnRH neurons in the anterior hypothalamus, suggesting an unexpected role of GnRH signaling in establishing the size of the GnRH neuronal population. Our experiments define a functional role of embryonic GnRH signaling.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Maturidade Sexual , Transdução de Sinais , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hipogonadismo/genética , Hipogonadismo/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Adeno-Hipófise/embriologia , Adeno-Hipófise/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo
17.
Reprod Biol ; 10(2): 85-124, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20668503

RESUMO

This review is focused on the relationship between neuroendocrine regulation of GnRH/LH secretion and the expression of GnRH and GnRH receptor (GnRHR) genes in the hypothalamic-pituitary unit during different physiological states of animals and under stress. Moreover, the involvement of hypothalamic GABA-ergic, Beta-endorphinergic, CRH-ergic, noradrenergic, dopaminergic and GnRH-ergic systems in the regulation of expression of the GnRH and GnRHR genes as well as secretion of GnRH/LH is analyzed. It appears that the neural mechanisms controlling GnRH gene expression in different physiological states may be distinct from those regulating GnRH/LH release. The hypothalamic GnRHR gene is probably located in different neural systems and may act in a specific way on GnRH gene expression and GnRH release.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Receptores LHRH/genética , Vias Aferentes/fisiologia , Anestro , Animais , Encéfalo/fisiologia , Clonagem Molecular , Endorfinas/fisiologia , Estro , Feminino , Regulação da Expressão Gênica , Humanos , Hipotálamo/fisiologia , Neurônios , Adeno-Hipófise/fisiologia
18.
Neuroendocrinology ; 92(2): 81-99, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20606386

RESUMO

Evolutionary survival of a species is largely a function of its reproductive fitness. In mammals, a sparsely populated and widely dispersed network of hypothalamic neurons, the gonadotropin-releasing hormone (GnRH) neurons, serve as the pilot light of reproduction via coordinated secretion of GnRH. Since it first description, human GnRH deficiency has been recognized both clinically and genetically as a heterogeneous disease. A spectrum of different reproductive phenotypes comprised of congenital GnRH deficiency with anosmia (Kallmann syndrome), congenital GnRH deficiency with normal olfaction (normosmic idiopathic hypogonadotropic hypogonadism), and adult-onset hypogonadotropic hypogonadism has been described. In the last two decades, several genes and pathways which govern GnRH ontogeny have been discovered by studying humans with GnRH deficiency. More importantly, detailed study of these patients has highlighted the emerging theme of oligogenicity and genotypic synergism, and also expanded the phenotypic diversity with the documentation of reversal of GnRH deficiency later in adulthood in some patients. The underlying genetic defect has also helped understand the associated nonreproductive phenotypes seen in some of these patients. These insights now provide practicing clinicians with targeted genetic diagnostic strategies and also impact on clinical management.


Assuntos
Hormônio Liberador de Gonadotropina/deficiência , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/crescimento & desenvolvimento , Síndrome de Kallmann/genética , Animais , Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/genética , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Humanos , Hipogonadismo/genética , Masculino , Camundongos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Transtornos do Olfato/genética , Fenótipo , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores LHRH/genética , Receptores LHRH/metabolismo , Receptores da Neurocinina-3/genética , Receptores da Neurocinina-3/metabolismo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
19.
Anim Reprod Sci ; 120(1-4): 105-11, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20427135

RESUMO

An immune/inflammatory challenge can affect reproduction at the level of the hypothalamus, pituitary gland, or gonads. Nonetheless, the major impact is thought to occur within the brain or the pituitary gland. The present study was designed to examine the effect of intravenous (i.v.) lipopolysaccharide (LPS) injection on the expression of gonadotropin-releasing hormone (GnRH) and the gonadotropin-releasing hormone receptor (GnRHR) genes in the hypothalamic structures where GnRH neurons are located as well as in the anterior pituitary gland (AP) of anestrous ewes. We also determined the effect of LPS on luteinizing hormone (LH) release. It was found that i.v. LPS injection significantly decreased GnRH and GnRHR mRNAs levels in the preoptic area (40%, p

Assuntos
Anestro/efeitos dos fármacos , Endotoxinas/farmacologia , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/efeitos dos fármacos , Adeno-Hipófise/efeitos dos fármacos , Receptores LHRH/genética , Ovinos , Anestro/genética , Anestro/metabolismo , Animais , Endotoxinas/administração & dosagem , Feminino , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/farmacologia , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores LHRH/metabolismo , Ovinos/genética , Ovinos/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética
20.
Endocrinology ; 150(6): 2847-56, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19264870

RESUMO

In vertebrates, activation of the GnRH receptor is necessary to initiate the reproductive cascade. However, little is known about the characteristics of GnRH receptors before the vertebrates evolved. Recently genome sequencing was completed for amphioxus, Branchiostoma floridae. To understand the GnRH receptors (GnRHR) from this most basal chordate, which is also classified as an invertebrate, we cloned and characterized four GnRHR cDNAs encoded in the amphioxus genome. We found that incubation of GnRH1 (mammalian GnRH) and GnRH2 (chicken GnRH II) with COS7 cells heterologously expressing the amphioxus GnRHRs caused potent intracellular inositol phosphate turnover in two of the receptors. One of the two receptors displayed a clear preference for GnRH1 over GnRH2, a characteristic not previously seen outside the type I mammalian GnRHRs. Phylogenetic analysis grouped the four receptors into two paralogous pairs, with one pair grouping basally with the vertebrate GnRH receptors and the other grouping with the octopus GnRHR-like sequence and the related receptor for insect adipokinetic hormone. Pharmacological studies showed that octopus GnRH-like peptide and adipokinetic hormone induced potent inositol phosphate turnover in one of these other two amphioxus receptors. These data demonstrate the functional conservation of two distinct types of GnRH receptors at the base of chordates. We propose that one receptor type led to vertebrate GnRHRs, whereas the other type, related to the mollusk GnRHR-like receptor, was lost in the vertebrate lineage. This is the first report to suggest that distinct invertebrate and vertebrate GnRHRs are present simultaneously in a basal chordate, amphioxus.


Assuntos
Evolução Molecular , Invertebrados , Filogenia , Receptores LHRH/análise , Receptores LHRH/genética , Vertebrados , Sequência de Aminoácidos , Animais , Cordados , DNA Complementar/genética , Humanos , Dados de Sequência Molecular , Receptores LHRH/fisiologia , Reprodução/fisiologia , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA