Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Diabetes ; 16(4): e13541, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38599822

RESUMEN

Kisspeptins (KPs) are proteins that were first recognized to have antimetastatic action. Later, the critical role of this peptide in the regulation of reproduction was proved. In recent years, evidence has been accumulated supporting a role for KPs in regulating metabolic processes in a sexual dimorphic manner. It has been proposed that KPs regulate metabolism both indirectly via gonadal hormones and/or directly via the kisspeptin receptor in the brain, brown adipose tissue, and pancreas. The aim of the review is to provide both experimental and clinical evidence indicating that KPs are peptides linking metabolism and reproduction. We propose that KPs could be used as a potential target to treat both metabolic and reproductive abnormalities. Thus, we focus on the consequences of disruptions in KPs and their receptors in metabolic conditions such as diabetes, undernutrition, obesity, and reproductive disorders (hypogonadotropic hypogonadism and polycystic ovary syndrome). Data from both animal models and human subjects indicate that alterations in KPs in the case of metabolic imbalance lead also to disruptions in reproductive functions. Changes both in the hypothalamic and peripheral KP systems in animal models of the aforementioned disorders are discussed. Finally, an overview of current clinical studies involving KP in fertility and metabolism show fewer studies on metabolism (15%) and only one to date on both. Presented data indicate a dynamic and emerging field of KP studies as possible therapeutic targets in treatments of both reproductive and metabolic dysfunctions.


Asunto(s)
Kisspeptinas , Reproducción , Animales , Femenino , Humanos , Kisspeptinas/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo , Péptidos
2.
Sci Rep ; 14(1): 8989, 2024 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637687

RESUMEN

In mammals reproduction is regulated by many factors, among others by the peptides belonging to the RFamide peptide family. However, the knowledge concerning on the impact of recently identified member of this family (QRFP43) on the modulation of the gonadotrophic axis activity is still not fully understood and current research results are ambiguous. In the present study we tested the in vivo effect of QRFP43 on the secretory activity of the gonadotrophic axis at the hypothalamic-pituitary level in Polish Merino sheep. The animals (n = 48) were randomly divided into three experimental groups: controls receiving an icv infusion of Ringer-Locke solution, group receiving icv infusion of QRFP43 at 10 µg per day and 50 µg per day. All sheep received four 50 min icv infusions at 30 min intervals, on each of three consecutive days. Hypothalamic and pituitaries were collected and secured for further immunohistochemical and molecular biological analysis. In addition, during the experiment a blood samples have been collected for subsequent RIA determinations. QRFP43 was found to downregulate Kiss mRNA expression in the MBH and reduce the level of IR material in ME. This resulted in a reduction of GnRH IR material in the ME. QRFP43 increased plasma FSH levels while decreasing LH levels. Our findings indicate that QRFP43 inhibits the activity of the gonadotropic axis in the ovine at the level of the hypothalamus and may represent another neuromodulator of reproductive processes in animals.


Asunto(s)
Gonadotrofos , Hormona Luteinizante , Femenino , Ovinos , Animales , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hipófisis/metabolismo , Gonadotrofos/metabolismo , Hormona Folículo Estimulante , Mamíferos/metabolismo
3.
Theriogenology ; 219: 157-166, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38432143

RESUMEN

To understand better the role that kisspeptin plays in regulating seasonal and estrous cycle changes in the mare, this study investigated the number, location and interactions between GnRH, kisspeptin and RFRP-3 neurons in the equine hypothalamus. Hypothalami were collected from mares during the non-breeding season, vernal transition and various stages of the breeding season. Fluorescent immunohistochemistry was used to label the neuropeptides of interest. GnRH cells were observed primarily in the arcuate nucleus (ARC), while very few labeled cells were identified in the pre-optic area (POA). Kisspeptin cells were identified primarily in the ARC, with a small number of cells observed dorsal to the ARC, surrounding the third ventricle (3V). The mean number of kisspeptin cells varied between animals and typically showed no pattern associated with season or stage of estrous cycle, but a seasonal difference was identified in the ARC population. Small numbers of RFRP-3 cells were observed in the ARC, ventromedial hypothalamus (VMH) and dorsomedial hypothalamus (DMH). The mean number of RFRP-3 cells appeared higher in pre-ovulatory animals compared to all other stages. The percentage of GnRH cell bodies with kisspeptin appositions did not change with season or stage of estrous cycle. The percentage of kisspeptin cells receiving inputs from RFRP-3 fibers did not vary with season or stage of estrous cycle. These interactions suggest the possibility of the presence of an ultra-short loop feedback system between these three peptides. The changes in RFRP-3 neurons suggest the possibility of a role in the regulation of reproduction in the horse, but it is unlikely to be as a gonadotropin inhibitory factor.


Asunto(s)
Hormona Liberadora de Gonadotropina , Neuropéptidos , Caballos , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Estaciones del Año , Neuropéptidos/fisiología , Hipotálamo/metabolismo , Ciclo Estral/fisiología , Neuronas
4.
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
5.
Gen Comp Endocrinol ; 350: 114477, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38387532

RESUMEN

Gonadotropin-inhibitory hormone (GnIH) was the first reported hypothalamic neuropeptide inhibiting reproduction in vertebrates. Since its discovery in the quail brain, its orthologs have been identified in a variety of vertebrate species and even protochordates. Depending on the species, the GnIH precursor polypeptides comprise two, three or four mature peptides of the RFamide family. It has been well documented that GnIH inhibits reproduction at the brain-pituitary-gonadal levels and participates in metabolism, stress response, and social behaviors in birds and mammals. However, most studies in fish have mainly been focused on the physiological roles of GnIH in the control of reproduction and results obtained are in some cases conflicting, leaving aside its potential roles in the regulation of other functions. In this manuscript we summarize the information available in fish with respect to the structural diversity of GnIH peptides and functional roles of GnIH in reproduction and other physiological processes. We also highlight the molecular mechanisms of GnIH actions on target cells and possible interactions with other neuroendocrine factors.


Asunto(s)
Gonadotropinas , Hormonas Hipotalámicas , Animales , Gonadotropinas/metabolismo , Vertebrados/metabolismo , Péptidos/metabolismo , Hipotálamo/metabolismo , Reproducción/fisiología , Peces/metabolismo , Mamíferos/metabolismo , Hormonas Hipotalámicas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo
6.
Endocr Rev ; 45(1): 30-68, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-37467734

RESUMEN

Kisspeptin (KP) and neurokinin B (NKB) are neuropeptides that govern the reproductive endocrine axis through regulating hypothalamic gonadotropin-releasing hormone (GnRH) neuronal activity and pulsatile GnRH secretion. Their critical role in reproductive health was first identified after inactivating variants in genes encoding for KP or NKB signaling were shown to result in congenital hypogonadotropic hypogonadism and a failure of pubertal development. Over the past 2 decades since their discovery, a wealth of evidence from both basic and translational research has laid the foundation for potential therapeutic applications. Beyond KP's function in the hypothalamus, it is also expressed in the placenta, liver, pancreas, adipose tissue, bone, and limbic regions, giving rise to several avenues of research for use in the diagnosis and treatment of pregnancy, metabolic, liver, bone, and behavioral disorders. The role played by NKB in stimulating the hypothalamic thermoregulatory center to mediate menopausal hot flashes has led to the development of medications that antagonize its action as a novel nonsteroidal therapeutic agent for this indication. Furthermore, the ability of NKB antagonism to partially suppress (but not abolish) the reproductive endocrine axis has supported its potential use for the treatment of various reproductive disorders including polycystic ovary syndrome, uterine fibroids, and endometriosis. This review will provide a comprehensive up-to-date overview of the preclinical and clinical data that have paved the way for the development of diagnostic and therapeutic applications of KP and NKB.


Asunto(s)
Kisspeptinas , Neuroquinina B , Embarazo , Femenino , Humanos , Neuroquinina B/genética , Neuroquinina B/metabolismo , Kisspeptinas/uso terapéutico , Hormona Liberadora de Gonadotropina/metabolismo , Reproducción/fisiología , Hipotálamo
7.
Endocrine ; 83(3): 733-746, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37966704

RESUMEN

OBJECTIVE: We examined how the sex steroids influence the synthesis of gonadotropins. MATERIALS AND METHODS: The effects of sex steroids estradiol (E2), progesterone (P4), and dihydrotestosterone (DHT) in pituitary gonadotroph cell model (LßT2 cells) in vitro and ovary-intact rats in vivo were examined. The effects of sex steroids on Kiss1 gene expression in the hypothalamus were also examined in ovary-intact rats. RESULTS: In LßT2 cells, E2 increased common glycoprotein alpha (Cga) and luteinizing hormone beta (Lhb) subunit promoter activity as well as their mRNA expression. Although gonadotropin subunit promoter activity was not modulated by P4, Cga and Lhb mRNA expression was increased by P4. DHT inhibited Cga and Lhb mRNA expression with a concomitant decrease in their promoter activity. During the 2-week administration of exogenous E2 to ovary-intact rats, the estrous cycle determined by vaginal smears was disrupted. P4 or DHT administration completely eliminated the estrous cycle. Protein expression of all three gonadotropin subunits within the pituitary gland was inhibited by E2 or P4 treatment in vivo; however, DHT reduced Cga expression but did not modulate Lhb or follicle-stimulating hormone beta subunit expression. E2 administration significantly repressed Kiss1 mRNA expression in a posterior hypothalamic region that included the arcuate nucleus. P4 and DHT did not modulate Kiss1 mRNA expression in this region. In contrast, P4 administration significantly inhibited Kiss1 mRNA expression in the anterior region of the hypothalamus that included the anteroventral periventricular nucleus. The expression of gonadotropin-releasing hormone (Gnrh) mRNA in the anterior hypothalamic region, where the preoptic area is located, appeared to be decreased by treatment with E2 and P4. CONCLUSION: Our findings suggest that sex steroids have different effects in the hypothalamus and pituitary gland.


Asunto(s)
Kisspeptinas , Ovario , Ratas , Femenino , Animales , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hipotálamo/metabolismo , Gonadotropinas Hipofisarias/genética , Gonadotropinas Hipofisarias/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Estradiol/farmacología , ARN Mensajero/metabolismo , Dihidrotestosterona/farmacología , Expresión Génica
8.
Neurosci Lett ; 819: 137578, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38048875

RESUMEN

Persistent post-ischemic alterations to the hypothalamic-pituitary-adrenal (HPA) axis occur following global cerebral ischemia (GCI) in rodents. However, similar effects on hypothalamic-pituitary-gonadal (HPG) axis activation remain to be determined. Therefore, this study evaluated the effects of GCI in adult female rats (via four-vessel occlusion) on the regularity of the estrous cycle for 24-days post ischemia. A second objective aimed to assess persistent alterations of HPG axis activation through determination of the expression of estrogen receptor alpha (ERα), kisspeptin (Kiss1), and gonadotropin-inhibitory hormone (GnIH/RFamide-related peptide; RFRP3) in the medial preoptic area (POA), arcuate nucleus (ARC), dorsomedial nucleus (DMH) of the hypothalamus, and CA1 of the hippocampus 25 days post ischemia. Expression of glucocorticoid receptors (GR) in the paraventricular nucleus of the hypothalamus (PVN) and CA1 served as a proxy of altered HPA axis activation. Our findings demonstrated interruption of the estrous cycle in 87.5 % of ischemic rats, marked by persistent diestrus, lasting on average 11.86 days. Moreover, compared to sham-operated controls, ischemic female rats showed reduced Kiss1 expression in the hypothalamic ARC and POA, concomitant with elevated ERα in the ARC and increased GnIH in the DMH and CA1. Reduced GR expression in the CA1 was associated with increased GR-immunoreactivity in the PVN, indicative of lasting dysregulation of HPA axis activation. Together, these findings demonstrate GCI disruption of female rats' estrous cycle over multiple days, with a lasting impact on HPG axis regulators within the reproductive axis.


Asunto(s)
Isquemia Encefálica , Sistema Hipotálamo-Hipofisario , Ratas , Femenino , Animales , Sistema Hipotálamo-Hipofisario/metabolismo , Kisspeptinas/metabolismo , Eje Hipotálamico-Pituitario-Gonadal , Receptor alfa de Estrógeno/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Hipotálamo/metabolismo , Ciclo Estral/metabolismo , Isquemia Encefálica/metabolismo , Infarto Cerebral/metabolismo , Periodicidad
9.
Front Endocrinol (Lausanne) ; 14: 1269334, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37900144

RESUMEN

Introduction: Male reproduction is under the control of the hypothalamus-pituitary-gonadal (HPG) axis. The endocannabinoid system (ECS) and the kisspeptin system (KS) are two major signaling systems in the central and peripheral control of reproduction, but their possible interaction has been poorly investigated in mammals. This manuscript analyzes their possible reciprocal modulation in the control of the HPG axis. Materials and methods: Adolescent male rats were treated with kisspeptin-10 (Kp10) and endocannabinoid anandamide (AEA), the latter alone or in combination with the type 1 cannabinoid receptor (CB1) antagonist rimonabant (SR141716A). The hypothalamic KS system and GnRH expression, circulating sex steroids and kisspeptin (Kiss1) levels, and intratesticular KS and ECS were evaluated by immunohistochemical and molecular methods. Non-coding RNAs (i.e., miR145-5p, miR-132-3p, let7a-5p, let7b-5p) were also considered. Results: Circulating hormonal values were not significantly affected by Kp10 or AEA; in the hypothalamus, Kp10 significantly increased GnRH mRNA and aromatase Cyp19, Kiss1, and Kiss1 receptor (Kiss1R) proteins. By contrast, AEA treatment affected the hypothalamic KS at the protein levels, with opposite effects on the ligand and receptor, and SR141716A was capable of attenuating the AEA effects. Among the considered non-coding RNA, only the expression of miR145-5p was positively affected by AEA but not by Kp10 treatment. Localization of Kiss1+/Kiss1R+ neurons in the arcuate nucleus revealed an increase of Kiss1R-expressing neurons in Kp10- and AEA-treated animals associated with enlargement of the lateral ventricles in Kp10-treated animals. In the brain and testis, the selected non-coding RNA was differently modulated by Kp10 or AEA. Lastly, in the testis, AEA treatment affected the KS at the protein levels, whereas Kp10 affected the intragonadal levels of CB1 and FAAH, the main modulator of the AEA tone. Changes in pubertal transition-related miRNAs and the intratesticular distribution of Kiss1, Kiss1R, CB1, and CB2 following KP and AEA treatment corroborate the KS-ECS crosstalk also showing that the CB1 receptor is involved in this interplay. Conclusion: For the first time in mammals, we report the modulation of the KS in both the hypothalamus and testis by AEA and revealed the KP-dependent modulation of CB1 and FAAH in the testis. KP involvement in the progression of spermatogenesis is also suggested.


Asunto(s)
Kisspeptinas , MicroARNs , Masculino , Ratas , Animales , Kisspeptinas/genética , Kisspeptinas/metabolismo , Receptores de Kisspeptina-1/genética , Endocannabinoides/farmacología , Endocannabinoides/metabolismo , Rimonabant/metabolismo , Rimonabant/farmacología , Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Mamíferos/metabolismo , Reproducción , ARN no Traducido/metabolismo , MicroARNs/metabolismo
10.
Zhen Ci Yan Jiu ; 48(8): 804-11, 2023 Aug 25.
Artículo en Chino | MEDLINE | ID: mdl-37614139

RESUMEN

OBJECTIVE: To observe the effects of electroacupuncture (EA) on hormone secretion function of ovarian granulosa cells and theca cells, as well as the expression changes of kisspeptin and kiss1r in rats with polycystic ovarian syndrome (PCOS), so as to explore the mechanism of EA for relieving ovarian dysfunction in PCOS rats. METHODS: Forty-eight SD female rats were randomly divided into control group, model group, EA group and flutamide group, with 12 rats in each group. PCOS rat model was replicated with the gavage of letrozole (0.1 mg/mL, 10 mL•kg-1•d-1). In the EA group, EA (2 Hz, 2 mA) was used to stimulate "Guanyuan" (CV4) for 20 min each time. In the flutamide group, flutamide solution (50 mg•kg-1•d-1) was administrated by gavage. The treatments were given once daily for 14 days in each group. After the modeling and treatment, the body and ovarian weights of the rats were measured, and the ovarian index was calculated. Using HE staining, the morphological changes of ovary were observed. ELISA was adopted to detect the contents of testosterone (T), luteinizing hormone (LH) and estradiol (E2) in serum, the contents of E2 and T in the culture medium of ovarian granulosa cells and theca cells, as well as the content of kisspeptin in the ovarian tissue. The positive expression of kisspeptin in ovary was observed by immunohistochemical method, and the protein expression of its receptor kiss1r was detected by Western blot. RESULTS: Compared with the control group, the body and ovarian weights, ovarian index, the contents of T and LH in serum and that of T in the culture medium of theca cells, as well as the content and positive expression of kisspeptin in ovary were all increased (P<0.01, P<0.05); and the content of E2 in the culture medium of granulosa cells was decreased (P<0.01) in the model group. When compared with the model group, in the EA and flutamide groups, the body and ovarian weights, ovarian index, the contents of T and LH in serum and that of T in the culture medium of theca cells, as well as the content and expression of kisspeptin in ovary were all decreased (P<0.01, P<0.05); and the content of E2 in the culture medium of granulosa cells was increased (P<0.05, P<0.01). CONCLUSION: EA regulates the serum sex hormone levels, the secretion function of the ovarian granulosa cells and theca cells, and the ovarian kisspeptin/kiss1r protein expression in PCOS rats, showing the similar effect as receptor blockade intervention. It is suggested that the improvement of EA in ovarian dysfunction of PCOS rats may be related to the kisspeptin/kiss1r system.


Asunto(s)
Electroacupuntura , Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratas , Flutamida , Kisspeptinas/genética , Hormona Luteinizante , Síndrome del Ovario Poliquístico/genética , Síndrome del Ovario Poliquístico/terapia
11.
Expo Health ; : 1-17, 2023 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-37360514

RESUMEN

Brain-derived neurotrophic factor (BDNF) and kisspeptin-1 (KISS-1) regulate placental development and fetal growth. The predictive value of maternal serum BDNF and KISS-1 concentrations for placental and umbilical cord levels has not yet been explored. The influence of prenatal lead (Pb) and cadmium (Cd) exposure and maternal iron status on BDNF and KISS-1 levels is also unclarified and of concern. In a pilot cross-sectional study with 65 mother-newborn pairs, we analyzed maternal and cord serum levels of pro-BDNF, mature BDNF, and KISS-1, BDNF, and KISS-1 gene expression in placenta, Pb and Cd in maternal and umbilical cord blood (erythrocytes), and placenta. We conducted a series of in vitro experiments using human primary trophoblast cells (hTCs) and BeWo cells to verify main findings of the epidemiological analysis. Strong and consistent correlations were observed between maternal serum levels of pro-BDNF, mature BDNF, and KISS-1 and corresponding levels in umbilical serum and placental tissue. Maternal red blood cell Pb levels were inversely correlated with serum and placental KISS-1 levels. Lower expression and release of KISS-1 was also observed in Pb-exposed BeWo cells. In vitro Pb exposure also reduced cellular BDNF levels. Cd-treated BeWo cells showed increased pro-BDNF levels. Low maternal iron status was positively associated with low BDNF levels. Iron-deficient hTCs and BeWo cells showed a consistent decrease in the release of mature BDNF. The correlations between maternal BDNF and KISS-1 levels, placental gene expression, and umbilical cord serum levels, respectively, indicate the strong potential of maternal serum as predictive matrix for BDNF and KISS-1 levels in placentas and fetal sera. Pb exposure and iron status modulate BDNF and KISS-1 levels, but a clear direction of modulations was not evident. The associations need to be confirmed in a larger sample and validated in terms of placental and neurodevelopmental function. Supplementary Information: The online version contains supplementary material available at 10.1007/s12403-023-00565-w.

12.
Reprod Toxicol ; 119: 108410, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37211340

RESUMEN

We previously reported that female rats placed on a diet containing refined carbohydrates (HCD) resulted in obesity and reproductive abnormalities, such as high serum LH concentration and abnormal ovarian function. However, the impacts at the hypothalamic-pituitary (HP) function, specifically regarding pathways linked to reproductive axis modulation are unknown. In this study, we assessed whether subacute feeding with HCD results in abnormal reproductive control in the HP axis. Female rats were fed with HCD for 15 days and reproductive HP axis morphophysiology was assessed. HCD reduced hypothalamic mRNA expression (Kiss1, Lepr, and Amhr2) and increased pituitary LHß+ cells. These changes likely contribute to the increase in serum LH concentration observed in HCD. Blunted estrogen negative feedback was observed in HCD, with increased kisspeptin protein expression in the arcuate nucleus of the hypothalamus (ARH), lower LHß+ cells and LH concentration in ovariectomized (OVX)+HCD rats. Thus, these data suggest that HCD feeding led to female abnormal reproductive control of HP axis.


Asunto(s)
Hipotálamo , Obesidad , Ratas , Femenino , Animales , Hipotálamo/metabolismo , Obesidad/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Dieta , Carbohidratos , Kisspeptinas/genética , Kisspeptinas/metabolismo
13.
Gen Comp Endocrinol ; 337: 114260, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36933747

RESUMEN

mHypoA-55 cells are kisspeptin-expressing neuronal cells originating from the arcuate nucleus of the mouse hypothalamus. These cells are called KNDy neurons because they co-express kisspeptin, neurokinin B, and dynorphin A. In addition, they express gonadotropin-releasing hormone (GnRH). Here, we found that kisspeptin 10 (KP10) increased Kiss-1 (encoding kisspeptin) and GnRH gene expression in kisspeptin receptor (Kiss-1R)-overexpressing mHypoA-55 cells. KP10 greatly increased serum response element (SRE) promoter activity, which is a target of extracellular signal-regulated kinase (ERK) (20.0 ± 2.54-fold). KP10 also increased cAMP-response element (CRE) promoter activity in these cells (2.32 ± 0.36-fold). KP10-increased SRE promoter activity was significantly prevented in the presence of PD098095, a MEK kinase (MEKK) inhibitor, and KP10-induced CRE promoter activity was also inhibited by PD098059. Similarly, H89, a protein kinase A (PKA) inhibitor, significantly inhibited the KP10 induction of SRE and CRE promoters. KP10-induced Kiss-1 and GnRH gene expressions were inhibited in the presence of PD098059. Likewise, H89 significantly inhibited the KP10-induced increase in Kiss-1 and GnRH. Transfection of mHypoA-55 cells with constitutively active MEKK (pFC-MEKK) increased SRE and CRE promoter activities by 9.75 ± 1.77- and 1.36 ± 0.12-fold, respectively. Induction of constitutively active PKA (pFC-PKA) also increased SRE and CRE promoter activities by 2.41 ± 0.42- and 40.71 ± 7.77-fold, respectively. Furthermore, pFC-MEKK and -PKA transfection of mHypoA-55 cells increased both Kiss-1 and GnRH gene expression. Our current observations suggest that KP10 increases both the ERK and PKA pathways and that both pathways mutually interact in mHypoA-55 hypothalamic cells. Activation of both ERK and PKA signaling might be necessary to induce Kiss-1 and GnRH gene expressions.


Asunto(s)
Hormona Liberadora de Gonadotropina , Kisspeptinas , Animales , Ratones , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/farmacología , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Transducción de Señal
14.
J Ovarian Res ; 16(1): 15, 2023 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-36650561

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is characterized by hyperandrogenism, anovulation, and polycystic ovaries. Electroacupuncture (EA) can effectively improve hyperandrogenism and increase ovulation frequency in patients with PCOS. Pieces of suggest that androgen activity in the brain is associated with impaired steroid negative feedback in such patients. Studies have shown that EA regulated androgen receptor (AR) expression and local factor levels (such as anti-Müllerian hormone and inhibin B) in the ovary of PCOS rats. However, few studies have explored the effect of EA on androgen activity in the brain. OBJECTIVE: This study investigated the effect of EA on the kisspeptin-gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) neural circuit and sex hormone receptor expression in the hypothalamus of PCOS rats. METHODS: PCOS signs were induced by letrozole administration, and the induced rats were treated with low-frequency EA at Guan Yuan acupoint (CV4). The effect of EA on PCOS-like signs was evaluated by observing changes in the body weight, ovarian quality, ovarian morphology, and serum sex hormone levels in rats. To explore the mechanism of the effect of EA on PCOS-like signs, the neuropeptide content of the kisspeptin-GnRH/LH neural circuit was assessed using enzyme-linked immunosorbent assay(ELISA); AR and estrogen receptor α (ERα) coexpression on kisspeptin/neurokinin B/dynorphin (KNDy) neurons was determined via triple-label immunofluorescence; and protein and mRNA expression of Kiss1, Ar, Esr1, and kisspeptin receptor (Kiss1r) was evaluated via western blotting and Reverse Transcription-Polymerase Chain Reaction (RT-PCR). RESULTS: The results revealed that the estrous cycle of rats in the EA treatment group recovered, and their body and ovary weight reduced; ovarian morphology improved; serum testosterone and LH levels significantly decreased; and kisspeptin, GnRH, and dynorphin levels in hypothalamic arcuate nucleus significantly decreased. Compared with controls, the number of AR/Kiss1-positive cells increased, number of ERα/Kiss1-positive cells decreased, and protein and mRNA expression of Kiss1, Ar, and Kiss1r significantly increased in PCOS rats. However, EA treatment reversed these changes and reduced the expression of Kiss1, Ar, and Kiss1r significantly. CONCLUSION: Improvement in the reproductive hallmarks of PCOS rats via EA may be achieved by regulating the kisspeptin-GnRH/LH circuit via androgen activity attenuation. Thus, the results provide an experimental basis for acupuncture as an adjuvant medical therapy on PCOS.


Asunto(s)
Electroacupuntura , Hiperandrogenismo , Síndrome del Ovario Poliquístico , Animales , Femenino , Humanos , Ratas , Andrógenos/metabolismo , Dinorfinas/metabolismo , Receptor alfa de Estrógeno/metabolismo , Hormonas Esteroides Gonadales , Hormona Liberadora de Gonadotropina , Kisspeptinas/metabolismo , Hormona Luteinizante , Neuroquinina B/metabolismo , Neuronas , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/terapia , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Receptores de Kisspeptina-1/metabolismo , ARN Mensajero/metabolismo
15.
Peptides ; 162: 170958, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36682622

RESUMEN

Inflammatory diseases attenuate reproductive functions in humans and domestic animals. Lipopolysaccharide (LPS), an endotoxin released by bacteria, is known to disrupt female reproductive functions in various inflammatory diseases. LPS administration has been used to elucidate the impact of pathophysiological activation of the immune system on reproduction. Hypothalamic kisspeptin neurons are the master regulators of mammalian reproduction, mediating direct stimulation of hypothalamic gonadotropin-releasing hormone (GnRH) release and consequent release of gonadotropins, such as luteinizing hormone (LH) and follicle-stimulating hormone from the pituitary. The discovery of kisspeptin neurons in the mammalian hypothalamus has drastically advanced our understanding of how inflammatory stress causes reproductive dysfunction in both humans and domestic animals. Inflammation-induced ovarian dysfunction could be caused, at least partly, by aberrant GnRH and LH secretion, which is regulated by kisspeptin signaling. In this review, we focus on the effects of LPS on hypothalamic kisspeptin neurons to outline the impact of inflammatory stress on neuroendocrine regulation of mammalian reproductive systems. First, we summarize the attenuation of female reproduction by LPS during inflammation and the effects of LPS on ovarian and pituitary function. Second, we outline the inhibitory effects of LPS on pulsatile- and surge-mode GnRH/LH release. Third, we discuss the LPS-responsive hypothalamic-pituitary-adrenal axis and hypothalamic neural systems in terms of the cytokine-mediated pathway and the possible direct action of LPS via its hypothalamic receptors. This article describes the impact of LPS on hypothalamic kisspeptin neurons and the possible mechanisms underlying LPS-mediated disruption of LH pulses/surge via kisspeptin neurons.


Asunto(s)
Animales Domésticos , Infertilidad , Humanos , Animales , Femenino , Animales Domésticos/metabolismo , Kisspeptinas/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Lipopolisacáridos , Sistema Hipófiso-Suprarrenal/metabolismo , Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina , Hormona Luteinizante/metabolismo , Neuronas/metabolismo , Infertilidad/metabolismo , Mamíferos
16.
Reprod Sci ; 30(3): 802-822, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35799018

RESUMEN

Highly sophisticated and synchronized interactions of various cells and hormonal signals are required to make organisms competent for reproduction. GnRH neurons act as a common pathway for multiple cues for the onset of puberty and attaining reproductive function. GnRH is not directly receptive to most of the signals required for the GnRH secretion during the various phases of the ovarian cycle. Kisspeptin neurons of the hypothalamus convey these signals required for the synchronized release of the GnRH. The steroid-sensitive anteroventral periventricular nucleus (AVPV) kisspeptin and arcuate nucleus (ARC) KNDy neurons convey steroid feedback during the reproductive cycle necessary for GnRH surge and pulse, respectively. AVPV region kisspeptin neurons also communicate with nNOS synthesizing neurons and suprachiasmatic nucleus (SCN) neurons to coordinate the process of the ovarian cycle. Neurokinin B (NKB) and dynorphin play roles in the GnRH pulse stimulation and inhibition, respectively. The loss of NKB and kisspeptin function results in the development of neuroendocrine disorders such as hypogonadotropic hypogonadism (HH) and infertility. Ca2+ signaling is essential for GnRH pulse generation, which is propagated through gap junctions between astrocytes-KNDy and KNDy-KNDy neurons. Impaired functioning of KNDy neurons could develop the characteristics associated with polycystic ovarian syndrome (PCOS) in rodents. Kisspeptin-increased synthesis led to excessive secretion of the LH associated with PCOS. This review provides the latest insights and understanding into the role of the KNDy and AVPV/POA kisspeptin neurons in GnRH secretion and PCOS.


Asunto(s)
Hormona Liberadora de Gonadotropina , Síndrome del Ovario Poliquístico , Femenino , Humanos , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Síndrome del Ovario Poliquístico/metabolismo
17.
Ecotoxicol Environ Saf ; 249: 114387, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36508816

RESUMEN

Increasing evidence shows that the early onset of puberty in female offspring may be caused by maternal prenatal exposure to bisphenol A (BPA) during pregnancy; however, the critical time window of maternal prenatal BPA exposure remains unknown. Here, we identify the critical time window of gestational BPA exposure that induces early onset of puberty in female offspring. Pregnant CD-1 mice were gavaged with BPA (8 mg/kg) daily during the early gestational stage (GD1-GD6), middle gestational stage (GD7-GD12) or late gestational stage (GD13-GD18). We show that maternal BPA exposure during the early and middle gestational stages could advance the vaginal opening time and increase the serum levels of kisspeptin-10 and GnRH in the female offspring at PND 34. Mechanistically, maternal BPA exposure during early and middle gestation could significantly increase CpG island methylation in the Eed gene promoters but reduce the mRNA expression of Eed in the hypothalamus tissues of the female offspring. In conclusion, the critical period of maternal BPA exposure-induced early onset of puberty in female offspring is early and middle gestation; this BPA-induced early onset of puberty might be partly attributed to epigenetic programming of the Eed gene in the hypothalamus. This study provides important insights regarding the relationship and the mechanisms between BPA and offspring pubertal development.


Asunto(s)
Compuestos de Bencidrilo , Exposición Materna , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Humanos , Ratones , Embarazo , Compuestos de Bencidrilo/toxicidad , Compuestos de Bencidrilo/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Maduración Sexual/efectos de los fármacos
18.
Physiol Behav ; 260: 114055, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36563733

RESUMEN

AIMS: This study investigated the possible relationships between the expression of the Kiss1 and Gpr54 gene expressions and the pituitary-gonadal hormones with the female onset of puberty and sexual behavior. The Kiss1 and Gpr54 gene expressions were examined because they are critical to controlling the hypothalamic activation of GnRH neurons and, in turn, the pituitary-gonadal hormones related to the early onset of puberty and sexual behavior. Further, it was evaluated that the pituitary and gonadal hormones involved in the vaginal opening and the expression of sexual behavior. METHODS: Pregnant rats exposed to PRS from gestation days 17 to 20 were evaluated for maternal and open-field behaviors. The maternal behavior was analyzed because it may alter brain sexual organization affecting the pups development. It was observed in female pups the physical and development and, in adult age, the open-field behavior, the anxiety-like behavior, the estrous cycle, the sexual behavior, the serum FSH, LH, estrogen, progesterone, and testosterone levels, and the gene expression of kisspeptin protein (Kiss1) and Gpr54 in the hypothalamus. RESULTS: the maternal and open-field behaviors were unaffected. In the F1 generation, PRS reduced weight at weaning, delayed the day of the vaginal opening and reduced the intensity of lordosis, the estrogen levels, and the Kiss1 and Gpr54 gene expression. These effects were attributed to hypothalamic kisspeptidergic system downregulation of transcripts genes and the reduced estrogen levels affected by the PRS.


Asunto(s)
Kisspeptinas , Maduración Sexual , Embarazo , Ratas , Animales , Femenino , Kisspeptinas/genética , Maduración Sexual/fisiología , Hipotálamo/metabolismo , Estrógenos/farmacología
19.
Endocrinology ; 165(2)2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38170643

RESUMEN

There is considerable evidence that synchronized activity within a reciprocally connected population of cells in the arcuate nucleus (ARC) coexpressing kisspeptin, neurokinin B (NKB), and dynorphin (KNDy cells) is crucial for the generation of gonadotrophin-releasing hormone (GnRH) pulses in mammals. The initial "KNDy hypothesis" proposed that pulsatile GnRH secretion is elicited by episodic kisspeptin release from KNDy cells following synchronized activation and termination of the population by NKB and dynorphin, respectively. Since then, the role of KNDy cells as a critical component of the pulse generator has been further supported by studies at the single-cell level, demonstrating that the population is both necessary and sufficient for pulsatility. In addition, there have been considerable modifications and expansion of the original hypothesis, including work demonstrating the critical role of glutamate in synchronization of the KNDy cell network, functional interactions with other ARC subpopulations, and the existence of species differences in the role of dynorphin in pulse generation. Here we review these recent changes and discuss how the translation of these findings has led to the development of new therapies for disorders related to pulse generation. We also outline critical gaps in knowledge that are currently limiting the application of KNDy research in the clinic, particularly regarding the role of dynorphin in pulse generation in primates.


Asunto(s)
Dinorfinas , Hormona Liberadora de Gonadotropina , Animales , Kisspeptinas , Hipotálamo , Núcleo Arqueado del Hipotálamo , Neuroquinina B , Neuronas , Mamíferos
20.
Aging (Albany NY) ; 14(21): 8615-8632, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36326686

RESUMEN

The process of aging is the result of progressive loss of homeostasis and functional body impairment, including the central nervous system, where the hypothalamus plays a key role in regulating aging mechanisms. The consequences of aging include a chronic proinflammatory environment in the hypothalamus that leads to decreased secretion of gonadotropin-releasing hormone (GnRH) and impairs kisspeptin neuron functionality. In this work, we investigated the effect of insulin-like growth factor 1 (IGF1) gene therapy on hypothalamic kisspeptin/GnRH neurons and on microglial cells, that mediate the inflammatory process related with the aging process. The results show that IGF1 rats have higher kisspeptin expression in the anteroventral periventricular (AVPV) nucleus and higher immunoreactivity of GnRH in the arcuate nucleus and median eminence. In addition, IGF1-treated animals exhibit increased numbers of Iba1+ microglial cells and MHCII+/Iba1+ in the AVPV and arcuate nuclei. In conclusion, IGF1 gene therapy maintains kisspeptin production in the AVPV nucleus, induces GnRH release in the median eminence, and alters the number and reactivity of microglial cells in middle-aged female rats. We suggest that IGF1 gene therapy may have a protective effect against reproductive decline.


Asunto(s)
Hormona Liberadora de Gonadotropina , Kisspeptinas , Femenino , Ratas , Animales , Kisspeptinas/genética , Hormona Liberadora de Gonadotropina/genética , Hormonas Liberadoras de Hormona Hipofisaria , Factor I del Crecimiento Similar a la Insulina/genética , Hipotálamo , Gonadotropinas , Neuronas , Envejecimiento , Terapia Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA