Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biochem Mol Toxicol ; 38(4): e23699, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38532648

RESUMEN

The endocrine disruptor hexavalent chromium [Cr(VI)] is a proven reproductive toxicant. We recently demonstrated that prenatal Cr(VI) exposure causes testicular resistance to gonadotropins, resulting in hypergonadotropic hypoandrogenism in F1 rats. However, the mechanism driving hypergonadotropism in F1 rats exposed to Cr(VI) prenatally remains an enigma. Therefore, we hypothesized that 'Prenatal Cr(VI) exposure may disrupt steroid hormones-mediated negative feedback regulation of the hypothalamic GnRH, and its receptor in the pituitary of F1 rats, leading to hypergonadotropism.' We administered potassium dichromate (50, 100, or 200 mg/L) to pregnant rats through drinking water between days 9 and 14, and their male F1 offspring were euthanized at 60 days of age. Prenatal Cr(VI) exposure in F1 rats resulted in the accumulation of Cr in the hypothalamus and pituitary. Western blot detected decreased hypothalamic GnRH, Kisspeptin1, and its receptor GPR54, along with diminished ERα, AR, aromatase, and 5α reductase, and GnRH regulatory transcription factors Pit-1 and GATA-4 proteins. Immunohistochemical studies revealed increased immunopositivity of GnRH receptor, AR, 5α reductase, ERα, ERß, and aromatase proteins in the pituitary, whereas decreased Kisspeptin1, GPR54, and inhibin ß. Our findings imply that Cr(VI) exposure during the prenatal period disrupts the hypothalamic Kisspeptin-GPR54-Pit-1/GATA4-GnRH network, boosting the pituitary GnRH receptor. We conclude that prenatal exposure to Cr(VI) alters GnRH expression in the hypothalamus and its receptor in the pituitary of F1 progeny through interfering with the negative feedback effect of androgens and estrogens.


Asunto(s)
Cromo , Efectos Tardíos de la Exposición Prenatal , Receptores LHRH , Femenino , Embarazo , Humanos , Ratas , Masculino , Animales , Receptores LHRH/metabolismo , Receptor alfa de Estrógeno/metabolismo , Aromatasa , Efectos Tardíos de la Exposición Prenatal/metabolismo , Hipotálamo , Hormona Liberadora de Gonadotropina/metabolismo
2.
Horm Behav ; 145: 105230, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35809386

RESUMEN

It is widely known that GnRH plays a role in facilitating reproductive function via the HPG axis, and this was once believed to be its only function. However, over the last several decades important neuromodulatory roles of GnRH in multiple brain functions have been elucidated. Multiple GnRH isoforms and receptors have been detected outside the HPG-axis across different species. In this review, we focus on the human CNS where GnRH I and II isoforms and a functional GnRH I receptor have been isolated. We first describe the traditional understanding of GnRH within the hypothalamus and the pituitary and current clinical use of GnRH analogues. We then review the location and function of GnRH-producing neurons and receptors located outside the HPG axis. We next review the GnRH I and II neuron location and quantity and GnRH I receptor gene expression throughout the human brain, using the Allen Brain Map Atlas. This analysis demonstrates a wide expression of GnRH throughout the brain, including prominent expression in the basal forebrain and cerebellum. Lastly, we examine the potential role of GnRH in aging and inflammation and its therapeutic potential for neurodegenerative disease and spinal cord lesions.


Asunto(s)
Enfermedades Neurodegenerativas , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Hipófisis/metabolismo , Receptores LHRH/metabolismo
3.
Gen Comp Endocrinol ; 310: 113819, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34015343

RESUMEN

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.


Asunto(s)
Hormona Liberadora de Gonadotropina , Receptores LHRH , Animales , Chlorocebus aethiops , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Masculino , Filogenia , Receptores LHRH/genética , Receptores LHRH/metabolismo , Espermatozoides/metabolismo
4.
Molecules ; 26(5)2021 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-33803091

RESUMEN

Although melatonin has been extensively studied in animal reproduction, the mechanism of melatonin in puberty remains elusive. This study was designed to explore the effect of intraperitoneal administration of melatonin on puberty onset in female mice. The injection of melatonin into postnatal days 10 mice at a dose of 15 mg/kg accelerated the puberty onset in mice. Mechanistically, there was no difference in physical growth and serum Leptin levels after melatonin administration. Meanwhile, the serum levels of reproductive hormones involved in hypothalamic-pituitary-ovarian axis, such as FSH and estrogen level in serum were increased. The mRNA levels of GnRH and GnRHr were not affected by melatonin, while the expressions of FSHß in pituitary and Cyp19a1 in ovary were significantly up-regulated. In addition, melatonin still promoted FSH synthesis after ovariectomy. Furthermore, the enhanced activity of ERK1/2 signaling verified that the expression of FSHß increased in pituitary. We confirmed that melatonin promoted the FSH synthesis in pituitary, thereby increased serum estrogen levels and ultimately accelerated puberty onset. However, these effects of melatonin may be pharmacological due to the high dose. This study would help us to understand the functions of melatonin in pubertal regulation comprehensively.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Melatonina/farmacología , Maduración Sexual/efectos de los fármacos , Animales , Aromatasa/metabolismo , China , Estrógenos/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Inyecciones Intraperitoneales , Leptina/metabolismo , Hormona Luteinizante/metabolismo , Melatonina/metabolismo , Ratones , Ovario/efectos de los fármacos , Hipófisis/metabolismo , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Receptores LHRH/metabolismo , Maduración Sexual/fisiología
5.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33805020

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Hipotálamo/metabolismo , Receptores LHRH/metabolismo , Reproducción , Regiones no Traducidas 3' , Adipoquinas/metabolismo , Animales , Estro , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Leptina/metabolismo , Ratones , Hipófisis/metabolismo , ARN Mensajero/metabolismo , Ratas , Receptores LHRH/genética , Transcripción Genética
6.
Int J Mol Sci ; 21(22)2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33198405

RESUMEN

Gonadotropin-releasing hormones (GnRHs) play pivotal roles in reproduction via the hypothalamus-pituitary-gonad axis (HPG axis) in vertebrates. GnRHs and their receptors (GnRHRs) are also conserved in invertebrates lacking the HPG axis, indicating that invertebrate GnRHs do not serve as "gonadotropin-releasing factors" but, rather, function as neuropeptides that directly regulate target tissues. All vertebrate and urochordate GnRHs comprise 10 amino acids, whereas amphioxus, echinoderm, and protostome GnRH-like peptides are 11- or 12-residue peptides. Intracellular calcium mobilization is the major second messenger for GnRH signaling in cephalochordates, echinoderms, and protostomes, while urochordate GnRHRs also stimulate cAMP production pathways. Moreover, the ligand-specific modulation of signal transduction via heterodimerization between GnRHR paralogs indicates species-specific evolution in Ciona intestinalis. The characterization of authentic or putative invertebrate GnRHRs in various tissues and their in vitro and in vivo activities indicate that invertebrate GnRHs are responsible for the regulation of both reproductive and nonreproductive functions. In this review, we examine our current understanding of and perspectives on the primary sequences, tissue distribution of mRNA expression, signal transduction, and biological functions of invertebrate GnRHs and their receptors.


Asunto(s)
Hipotálamo/metabolismo , Invertebrados/metabolismo , Receptores LHRH/metabolismo , Animales , Evolución Biológica , Células COS , Calcio/metabolismo , Chlorocebus aethiops , Ciona intestinalis , AMP Cíclico/metabolismo , Equinodermos , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Células HEK293 , Humanos , Ligandos , Masculino , Cadenas de Markov , Moluscos , Transducción de Señal , Distribución Tisular , Urocordados
7.
Sci Rep ; 10(1): 10579, 2020 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-32601341

RESUMEN

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.


Asunto(s)
Receptores LHRH/agonistas , Receptores LHRH/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Calcio/metabolismo , Evaluación Preclínica de Medicamentos , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/metabolismo , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Fosfatos de Inositol/metabolismo , Mutación , Pliegue de Proteína , Transporte de Proteínas , Receptores LHRH/genética
8.
Poult Sci ; 98(9): 4172-4181, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31001634

RESUMEN

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.


Asunto(s)
Antioxidantes/metabolismo , Proteínas Aviares/genética , Carnitina/metabolismo , Pollos/fisiología , Expresión Génica/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Alimentación Animal/análisis , Animales , Antioxidantes/administración & dosificación , Proteínas Aviares/metabolismo , Carnitina/administración & dosificación , Dieta/veterinaria , Suplementos Dietéticos/análisis , Relación Dosis-Respuesta a Droga , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Masculino , Receptor de Melatonina MT1/genética , Receptor de Melatonina MT1/metabolismo , Receptor de Melatonina MT2/genética , Receptor de Melatonina MT2/metabolismo , Receptores LHRH/genética , Receptores LHRH/metabolismo , Análisis de Semen/veterinaria , Espermatozoides/fisiología , Testículo/metabolismo
9.
Anim Sci J ; 90(4): 473-480, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30793438

RESUMEN

This study was aimed to address melatonin receptor expression, mRNA level of hypothalamus and hypophysis hormone receptors (GnRHR, FSHR, and LHR), steroidogenesis, cell cycle, apoptosis, and their regulatory factors after addition of melatonin for 24 hr in cultured buffalo granulosa cells (GCs). The results revealed that direct addition of different concentrations of melatonin (100 pM, 1 nM, and 100 nM) resulted in significant upregulation (p < 0.05) of mRNA level of melatonin receptor 1a (MT1) without affecting melatonin receptor 1b (MT2). Melatonin treatment significantly downregulated (p < 0.05) mRNA level of FSH and GnRH receptors, whereas 100 nM dose of melatonin significantly increased mRNA level of LH receptor. Treatment with 100 nM of melatonin significantly decreased the basal progesterone production with significant decrease (p < 0.05) in mRNA levels of StAR and p450ssc, and lower mRNA level of genes (Insig1, Lipe, and Scrab1) that affect cholesterol availability. Melatonin supplementation suppressed apoptosis (100 nM, p < 0.05) and enhanced G2/M phase (1 nM, 100 nM, p < 0.05) of cell cycle progression which was further corroborated by decrease in protein expression of caspase-3, p21, and p27 and increase in bcl2. Our results demonstrate that melatonin regulates gonadotrophin receptors and ovarian steroidogenesis through MT1. Furthermore, the notion of its incorporation in apoptosis and proliferation of buffalo GCs extends its role in buffalo ovaries.


Asunto(s)
Apoptosis/efectos de los fármacos , Estradiol/metabolismo , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/metabolismo , Células de la Granulosa/patología , Melatonina/farmacología , Progesterona/metabolismo , Animales , Búfalos , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Hormona Folículo Estimulante/genética , Expresión Génica/efectos de los fármacos , Melatonina/fisiología , ARN Mensajero/metabolismo , Receptor de Melatonina MT1/genética , Receptor de Melatonina MT1/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Receptores LHRH/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
J Cell Physiol ; 234(5): 6865-6875, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30417368

RESUMEN

Gonadotropin-releasing hormone (GnRH) is secreted from hypothalamic GnRH neurons and stimulates a GnRH receptor in gonadotroph cells and GnRH neurons. The GnRH receptor belongs to the G-protein-coupled receptors, and stimulation of the GnRH receptor activates extracellular signal-regulated protein kinase (ERK). We reported previously that the δ2 isoform of Ca2+ /calmodulin-dependent protein kinase II (CaM kinase IIδ2) was involved in GnRH-induced ERK activation in cultured GnRH neurons (GT1-7 cells). Recently, we found that GnRH treatment of GT1-7 cells activated proline-rich tyrosine kinase 2 (Pyk2), and Pyk2 was involved in ERK activation. In the current study, we examined the possibility that CaM kinase IIδ2 might activate Pyk2. Knockdown of CaM kinase IIδ2 and KN93, an inhibitor of CaM kinases, inhibited the GnRH-induced activation of Pyk2. In the case of cultured gonadotroph cells (αT3-1 cells), knockdown of CaM kinase IIß'e inhibited GnRH-induced Pyk2 activation. In addition, our inhibitor studies indicated that Pyk2 and CaM kinase II were involved in the GnRH-induced shedding of proHB-EGF in GT1-7 cells. These results suggested that CaM kinase II activated the ERK pathway through Pyk2 activation and HB-EGF production in response to GnRH.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Gonadotrofos/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Animales , Línea Celular , Activación Enzimática/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Receptores LHRH/metabolismo , Transducción de Señal/fisiología
11.
J Physiol Pharmacol ; 69(3)2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30342430

RESUMEN

This study aimed to determine the mechanisms governing Gonadotropin releasing hormone (GnRH) biosynthesis and luteinising hormone (LH) secretion in follicular-phase sheep after infusion of corticotropin releasing hormone (CRH) and/or CRH antagonist corticotropin releasing hormone nist (CRH-A) into the third cerebral ventricle. The study included two experimental approaches: first, we investigated the effect of CRH or CRH-A (α-helical CRH 9-41) on GnRH and GnRH receptor (GnRHR) biosynthesis in the preoptic area (POA), anterior (AH) and ventromedial hypothalamus (VMH), stalk/median eminence (SME), and on GnRHR in the anterior pituitary (AP) using an enzyme-linked immunosorbent assay (ELISA); second, we used real-time PCR to analyse the influence of CRH and CRH-A on the levels of kisspeptin (Kiss1) mRNA in POA and VMH including arcuate nucleus (VMH/ARC), and on Kiss1 receptor (Kiss1r) mRNA abundance in POA-hypothalamic structures. These analyses were supplemented by radioimmunoassay (RIA) and ELISA methods for measurement of LH and cortisol levels in the blood, respectively. Our results show that administration of CRH significantly decreased GnRH biosynthesis in the POA/hypothalamus. CRH also decreased GnRHR abundance in the hypothalamus and in the AP, but increased it in the POA. Furthermore, administration of CRH decreased plasma LH concentration and levels of Kiss1 mRNA in the POA and VMH/ARC as well as Kiss1r mRNA in these structures and in the SME. Significant increase in plasma cortisol concentration in the group treated with CRH was also observed. For CRH-A, all analysed effects were opposite to those induced by CRH. The study demonstrates that intracerebroventricular (i.c.v.) infusion of both CRH and CRH-A affects the GnRH/GnRHR biosynthesis and LH secretion in follicular-phase sheep conceivably via either central and peripheral mechanisms including Kiss1 neurons activity and cortisol signals. It has also been suggested that CRH and CRH-A infusion probably had effects directly at the AP.


Asunto(s)
Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Hormona Liberadora de Corticotropina/farmacología , Hormona Liberadora de Gonadotropina/biosíntesis , Hipotálamo/metabolismo , Receptores LHRH/metabolismo , Animales , Femenino , Fase Folicular/metabolismo , Hidrocortisona/sangre , Hipotálamo/efectos de los fármacos , Kisspeptinas/genética , Hormona Luteinizante/sangre , Receptores de Kisspeptina-1/genética , Ovinos
12.
Med J Aust ; 209(9): 412-416, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30376664

RESUMEN

Alkylating chemotherapy is often used to treat pre-menopausal women for various malignancies and autoimmune diseases. Chemotherapy-associated ovarian failure is a potential consequence of this treatment which can cause infertility, and increases the risk of other long term adverse health sequelae. Randomised trials, predominantly of women undergoing alkylating chemotherapy for breast cancer, have shown evidence for the efficacy of gonadotropin-releasing hormone agonists (GnRHa) in preventing chemotherapy-associated ovarian failure. The European St Gallen and United States National Comprehensive Cancer Network guidelines recommend the use of concurrent GnRHa to reduce the risk of ovarian failure for pre-menopausal women undergoing chemotherapy for breast cancer. The GnRHa goserelin, a monthly 3.6 mg depot subcutaneous injection, has recently been listed on the Australian Pharmaceutical Benefits Scheme to reduce risk of ovarian failure for pre-menopausal women receiving alkylating therapies for malignancy or autoimmune disease. The first dose of goserelin should ideally be administered at least 1 week before commencement of alkylating treatment and continued 4-weekly during chemotherapy. Concurrent goserelin use should now be considered for all pre-menopausal women due to commence alkylating chemotherapy (except those with incurable cancer), regardless of their childbearing status, in an effort to preserve their ovarian function. For women who have not completed childbearing, consideration of other fertility preservation options, such as cryopreservation of embryos or oocytes, is also important.


Asunto(s)
Antineoplásicos Alquilantes/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Hormona Liberadora de Gonadotropina/uso terapéutico , Infertilidad Femenina/prevención & control , Ovario/efectos de los fármacos , Neoplasias de la Mama/patología , Quimioterapia Adyuvante , Femenino , Preservación de la Fertilidad/métodos , Goserelina/uso terapéutico , Humanos , Infertilidad Femenina/inducido químicamente , Ovario/fisiopatología , Embarazo , Índice de Embarazo , Premenopausia , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores LHRH/agonistas
13.
Biol Reprod ; 99(2): 433-445, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30101293

RESUMEN

The pubertal transition of gonadotropin secretion in pigs is metabolically gated. Kisspeptin (KISS1) and neurokinin B (NKB) are coexpressed in neurons within the arcuate nucleus of the hypothalamus (ARC) and are thought to play an important role in the integration of nutrition and metabolic state with the reproductive neuroendocrine axis. The hypothesis that circulating concentrations of luteinizing hormone (LH) and expression of KISS1 and tachykinin 3(TAC3, encodes NKB) in the ARC of female pigs are reduced with negative energy balance was tested using ovariectomized, prepubertal gilts fed to either gain or lose body weight. Restricted feeding of ovariectomized gilts caused a rapid and sustained metabolic response characterized by reduced concentrations of plasma urea nitrogen, insulin, leptin, and insulin-like growth factor-1 and elevated concentrations of free fatty acids. The secretory pattern of LH shifted from one of low amplitude to one of high amplitude, which caused overall circulating concentrations of LH to be greater in restricted gilts. Nutrient-restricted gilts had greater expression of follicle-stimulating hormone and gonadotropin-releasing hormone receptor, but not LH in the anterior pituitary gland. Expression of KISS1 in the ARC was not affected by dietary treatment, but expression of TAC3 was greater in restricted gilts. These data are consistent with the idea that hypothalamic expression of KISS1 is correlated with the number of LH pulse in pig, and further indicate that amplitude of LH pulses may be regulated by NKB in the gilt.


Asunto(s)
Metabolismo Energético/fisiología , Privación de Alimentos/fisiología , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Neuroquinina B/metabolismo , Adenohipófisis/metabolismo , Animales , Ácidos Grasos no Esterificados/sangre , Femenino , Hormona Folículo Estimulante/metabolismo , Insulina/sangre , Kisspeptinas/metabolismo , Leptina/sangre , Neuronas/metabolismo , Receptores LHRH/metabolismo , Porcinos
14.
BMC Endocr Disord ; 18(1): 30, 2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29793475

RESUMEN

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.


Asunto(s)
Hipotálamo/patología , Hipotiroidismo/complicaciones , Infertilidad Femenina/etiología , Ovario/patología , Hipófisis/patología , Reproducción , Animales , Biomarcadores/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hipotiroidismo/metabolismo , Hipotiroidismo/patología , Infertilidad Femenina/diagnóstico , Infertilidad Femenina/metabolismo , Masculino , Ovario/metabolismo , Hipófisis/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores LHRH/genética , Receptores LHRH/metabolismo
15.
Biomed Pharmacother ; 102: 494-501, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29579710

RESUMEN

Luteal phase defects (LPD) are an important etiology of infertility which has increased in recent years. Studies have shown that bu-shen-zhu-yun decoction (BSZY-D) can lower the expression of estrogen receptor and progesterone receptor, in rats endometrium of embryonic implantation period, which upregulated by mifepristone, and improve uterine receptivity. The aim of present study was to determine the effect of BSZY-D on the synthesis and secretion of gonadotropic hormones in the anterior pituitary cells of rats. Rats were treated with saline (control) or BSZY-D two times/day for three estrous cycles by gavage. The cerebrospinal fluid (CSF) were collected for further cell treatment. The components in BSZY-D, serum and CSF were analysed by High Performance Liquid Chromatography (HPLC). Cells were either pretreated with normal CSF or BSZY-D/CSF before being stimulated with or without cetrorelix. The mRNA and proteins levels of receptors, hormones, and transcription factors were detected by RT-PCR, western blot analysis and immunostaining. We show that non-toxic concentrations of cetrorelix, a GnRH antagonist, can reduce the mRNA and protein levels of GnRHR, LH, and FSH. This effect could be reversed by the addition of BSZY-D/CSF. We also show decreased mRNA and protein expression of transcription factors, such as CREB, and Egr-1 and secretory vescicles, including SNAP-25 and Munc-18 upon treatment with cetrorelix could be reversed post co-treatment with BSZY-D/CSF. These results indicate that BSZY-D/CSF treatment led to increased levels of GnRHR, transcription factors, and secretory vesicles leading to increased secretion of FSH and LH. Thus, BSZY-D presents a promising candidate to treat luteal phase defects and infertility.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Hormona Folículo Estimulante de Subunidad beta/biosíntesis , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Luteinizante de Subunidad beta/biosíntesis , Hormona Luteinizante de Subunidad beta/metabolismo , Adenohipófisis/citología , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Cromatografía Líquida de Alta Presión , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Proteínas Munc18/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores LHRH/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos
16.
Endocrinology ; 159(3): 1496-1510, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29409045

RESUMEN

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.


Asunto(s)
Gonadotrofos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptores LHRH/metabolismo , Reproducción , Animales , Femenino , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipófisis/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Receptores LHRH/genética
17.
Am J Reprod Immunol ; 79(1)2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29048721

RESUMEN

PROBLEM: Immunocastration or vaccination against the GnRH-I hormone is a promising alternative to reproductive control in different animal species. Given the low immunogenicity of this hormone, the use of adjuvants becomes necessary. METHOD OF STUDY: This study evaluated the effects of three adjuvants that induce different immune response profiles over gonadal function, fertility, and expression of GnRH-I. Female mice (n = 6) were vaccinated at days 1 and 30 with a recombinant antigen for immunocastration and different adjuvants that induced preferentially Th1/Th2, Th2, and Th1 immune profiles. RESULTS: Th1/Th2 response is the most efficient to block reproductive activity in vaccinated animals, reducing the number of luteal bodies and pre-ovulatory follicles. Th2 and Th1/Th2 responses induced an increase in GnRH-I at the hypothalamus. CONCLUSION: The immune profile induced by different adjuvants is essential on the effects over fertility, gonadal function, and hypothalamic GnRH-I expression in immunocastrated animals.


Asunto(s)
Hormona Liberadora de Gonadotropina/inmunología , Gónadas/fisiología , Hipotálamo/metabolismo , Precursores de Proteínas/inmunología , Células TH1/inmunología , Células Th2/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Fertilidad , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/patología , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos BALB C , Precursores de Proteínas/genética , Receptores LHRH/metabolismo , Balance Th1 - Th2 , Vacunación
18.
Reprod Fertil Dev ; 30(4): 672-680, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29019791

RESUMEN

This study aimed to explain how prolonged inhibition of central dopaminergic activity affects the cellular processes governing gonadotrophin-releasing hormone (GnRH) and LH secretion in anoestrous sheep. For this purpose, the study included two experimental approaches: first, we investigated the effect of infusion of sulpiride, a dopaminergic D2 receptor antagonist (D2R), on GnRH and GnRH receptor (GnRHR) biosynthesis in the hypothalamus and on GnRHR in the anterior pituitary using an immunoassay. This analysis was supplemented by analysis of plasma LH levels by radioimmunoassay. Second, we used real-time polymerase chain reaction to analyse the influence of sulpiride on the levels of kisspeptin (Kiss1) mRNA in the preoptic area and ventromedial hypothalamus including arcuate nucleus (VMH/ARC), and RFamide-related peptide-3 (RFRP-3) mRNA in the paraventricular nucleus (PVN) and dorsomedial hypothalamic nucleus. Sulpiride significantly increased plasma LH concentration and the levels of GnRH and GnRHR in the hypothalamic-pituitary unit. The abolition of dopaminergic activity resulted in a significant increase in transcript level of Kiss1 in VMH/ARC and a decrease of RFRP-3 in PVN. The study demonstrates that dopaminergic neurotransmission through D2R is involved in the regulatory pathways of GnRH and GnRHR biosynthesis in the hypothalamic-pituitary unit of anoestrous sheep, conceivably via mechanisms in which Kiss1 and RFRP-3 participate.


Asunto(s)
Anestro/metabolismo , Antagonistas de los Receptores de Dopamina D2/farmacología , Hormona Liberadora de Gonadotropina/biosíntesis , Kisspeptinas/metabolismo , Neuropéptidos/metabolismo , Receptores LHRH/biosíntesis , Sulpirida/farmacología , Animales , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Adenohipófisis/efectos de los fármacos , Adenohipófisis/metabolismo , Ovinos
19.
Am J Physiol Gastrointest Liver Physiol ; 313(5): G410-G418, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28751425

RESUMEN

Melatonin is a hormone produced by the pineal gland with increased circulating levels shown to inhibit biliary hyperplasia and fibrosis during cholestatic liver injury. Melatonin also has the capability to suppress the release of hypothalamic gonadotropin-releasing hormone (GnRH), a hormone that promotes cholangiocyte proliferation when serum levels are elevated. However, the interplay and contribution of neural melatonin and GnRH to cholangiocyte proliferation and fibrosis in bile duct-ligated (BDL) rats have not been investigated. To test this, cranial levels of melatonin were increased by implanting osmotic minipumps that performed an intracerebroventricular (ICV) infusion of melatonin or saline for 7 days starting at the time of BDL. Hypothalamic GnRH mRNA and cholangiocyte secretion of GnRH and melatonin were assessed. Cholangiocyte proliferation and fibrosis were measured. Primary human hepatic stellate cells (HSCs) were treated with cholangiocyte supernatants, GnRH, or the GnRH receptor antagonist cetrorelix acetate, and cell proliferation and fibrosis gene expression were assessed. Melatonin infusion reduced hypothalamic GnRH mRNA expression and led to decreased GnRH and increased melatonin secretion from cholangiocytes. Infusion of melatonin was found to reduce hepatic injury, cholangiocyte proliferation, and fibrosis during BDL-induced liver injury. HSCs supplemented with BDL cholangiocyte supernatant had increased proliferation, and this increase was reversed when HSCs were supplemented with supernatants from melatonin-infused rats. GnRH stimulated fibrosis gene expression in HSCs, and this was reversed by cetrorelix acetate cotreatment. Increasing bioavailability of melatonin in the brain may improve outcomes during cholestatic liver disease.NEW & NOTEWORTHY We have previously demonstrated that GnRH is expressed in cholangiocytes and promotes their proliferation during cholestasis. In addition, dark therapy, which increases melatonin, reduced cholangiocyte proliferation and fibrosis during cholestasis. This study expands these findings by investigating neural GnRH regulation by melatonin during BDL-induced cholestasis by infusing melatonin into the brain. Melatonin infusion reduced cholangiocyte proliferation and fibrosis, and these effects are due to GNRH receptor 1-dependent paracrine signaling between cholangiocytes and hepatic stellate cells.


Asunto(s)
Conductos Biliares , Colestasis , Hormona Liberadora de Gonadotropina , Cirrosis Hepática , Melatonina , Glándula Pineal/fisiología , Animales , Conductos Biliares/efectos de los fármacos , Conductos Biliares/metabolismo , Conductos Biliares/patología , Proliferación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Depresores del Sistema Nervioso Central/sangre , Depresores del Sistema Nervioso Central/metabolismo , Colestasis/complicaciones , Colestasis/metabolismo , Modelos Animales de Enfermedad , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Antagonistas de Hormonas/farmacología , Humanos , Hiperplasia , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Melatonina/administración & dosificación , Melatonina/sangre , Melatonina/metabolismo , Ratas , Receptores LHRH/antagonistas & inhibidores
20.
Clin Drug Investig ; 37(9): 873-879, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28660498

RESUMEN

BACKGROUND AND OBJECTIVES: Degarelix is a gonadotropin-releasing hormone antagonist registered for the treatment of advanced hormone-dependent prostate cancer. Treatment causing androgen deprivation is associated with QT prolongation and this study investigated whether degarelix at supratherapeutic concentrations has an intrinsic effect per se on cardiac repolarisation and the QT interval. METHODS: This was a single-centre, randomised, crossover study comparing the effect of degarelix, placebo, and the positive control moxifloxacin on the QT interval. Degarelix and placebo treatments were double-blind, whereas moxifloxacin treatment was open-label. Eighty healthy men, aged 18-45 years, received single intravenous doses of degarelix 2.8 mg, and placebo, as well as a single oral dose of moxifloxacin 400 mg. Electrocardiograms were collected up to 24 h after the start of administration, with the QT interval assessed and plasma concentrations of degarelix concomitantly analysed. RESULTS: Time-matched, one-sided 95% upper confidence boundaries for baseline-corrected average changes from placebo for the QT interval, corrected using the Fridericia method (ΔΔQTcF), did not exceed 10 ms at any timepoint, with maximum degarelix concentrations reaching approximately threefold the concentrations seen in the treatment of prostate cancer. Furthermore, concentration-exposure analysis indicated absence of any QT prolongation effects of degarelix. No significant effect on any other cardiac parameter was observed. The lower bound of the 98.3% confidence interval for moxifloxacin ΔΔQTcF exceeded 5 ms, thus verifying assay sensitivity. CONCLUSION: The results showed that the study was validated to detect a significant effect on the QT interval, and that degarelix by itself does not have any effect on the QT interval and cardiac repolarisation at supratherapeutic concentrations.


Asunto(s)
Síndrome de QT Prolongado/inducido químicamente , Oligopéptidos/efectos adversos , Receptores LHRH/antagonistas & inhibidores , Adolescente , Adulto , Estudios Cruzados , Método Doble Ciego , Electrocardiografía , Fluoroquinolonas/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Moxifloxacino , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA