Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
Curr Issues Mol Biol ; 43(3): 1669-1684, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34698102

RESUMEN

Postmenopausal syndrome refers to symptoms caused by the gradual decrease in female hormones after mid-40 years. As a target organ of estrogen, decrease in estrogen causes various changes in brain function such as a decrease in choline acetyltransferase and brain-derived neurotrophic factor; thus, postmenopausal women experience cognitive decline and more depressive symptoms than age-matched men. Radix Polygalae has been used for memory boosting and as a mood stabilizer and its components have shown neuroprotective, antidepressant, and stress relief properties. In a mouse model of estrogen depletion induced by 4-vinylcyclohexene diepoxide, Radix Polygalae was orally administered for 3 weeks. In these animals, cognitive and depression-related behaviors and molecular changes related to these behaviors were measured in the prefrontal cortex and hippocampus. Radix Polygalae improved working memory and contextual memory and despair-related behaviors in 4-vinylcyclohexene diepoxide-treated mice without increasing serum estradiol levels in this model. In relation to these behaviors, choline acetyltransferase and brain-derived neurotrophic factor in the prefrontal cortex and hippocampus and bcl-2-associated athanogene expression increased in the hippocampus. These results implicate the possible benefit of Radix Polygalae in use as a supplement of estrogen to prevent conditions such as postmenopausal depression and cognitive decline.


Asunto(s)
Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Depresión/etiología , Depresión/metabolismo , Medicamentos Herbarios Chinos/farmacología , Estradiol/metabolismo , Menopausia/efectos de los fármacos , Menopausia/metabolismo , Animales , Conducta Animal , Disfunción Cognitiva/tratamiento farmacológico , Depresión/tratamiento farmacológico , Modelos Animales de Enfermedad , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Femenino , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Vagina/efectos de los fármacos , Vagina/metabolismo , Vagina/patología
3.
J Integr Neurosci ; 20(1): 77-85, 2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33834693

RESUMEN

The social behavior mechanisms have not been thoroughly reported in the solitary female striped dwarf hamster (Cricetulus barabensis). In this study, the handling bag test and neutral arena measurements were used to detect the changes of aggression in the face of rivals of different genders of wild striped dwarf hamsters. We found that female hamsters had the highest aggressive performance in proestrus, followed by estrus, and the lowest in metestrus and the dioestrus, and the increased aggression during the proestrus or estrus period was low-intensity aggression such as intimidation, shock, boxing and counterattack, or even ritualized non-harmful behaviors to drive away opponents. When confronted with male individuals, aggression in females decreased significantly during estrus. The concentration of plasma estradiol was the highest in estrus and the lowest in metestrus and dioestrus. In contrast, estrogen receptor 2 relative expression in the hypothalamus is the lowest in proestrus and highest in metestrus and dioestrus. Besides, both estradiol levels in plasma and estrogen receptor 2 mRNA in the hypothalamus were associated with aggression. These results will broaden our understanding of the molecular mechanism of how breeding phenotype is an essential driver in changing the social behavior of female Cricetulus barabensis.


Asunto(s)
Agresión/fisiología , Conducta Animal/fisiología , Estradiol/sangre , Receptor beta de Estrógeno/metabolismo , Ciclo Estral/fisiología , Hipotálamo/metabolismo , Conducta Social , Animales , Cricetinae , Ciclo Estral/metabolismo , Femenino , Masculino
4.
Histochem Cell Biol ; 155(6): 655-668, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33641022

RESUMEN

This study measured the sequelae of cholecalciferol (VD3) therapy on ovarian functions in adult VD-replete rats (n = 48). The animals were distributed into the control and VD groups following estrous cycle synchronisation. The VD group received VD3 injections for 4 weeks (600 IU/Kg; 3 times/week). Vaginal cytology and cycle durations were recorded throughout the study. Serum VD (25-OH VD), Ca2+, gonadotrophins (FSH & LH) and sex steroids (E2 & progesterone) were measured following euthanasia. Follicles and corpora lutea were counted in ovarian tissue sections. VD receptor, binding protein, Ca2+-sensing receptor and retinoid X receptor-α genes and proteins were measured by quantitative RT-PCR and immunohistochemistry. Serum VD, LH, E2 and progesterone levels were significantly higher, whereas FSH declined, in the VD group than controls. VD3 therapy was also associated with markedly higher rates alongside shorter durations of estrous cycles than controls. While serum Ca2+ levels were equal between the study groups, they correlated directly with serum 25-OH VD. The numbers of small and medium size ovarian follicles were equal in both study groups, whereas large follicles and corpora lutea counts were significantly higher in the VD group. The mRNAs and proteins of targeted molecules also increased substantially in the VD group than controls. In conclusion, treating VD-sufficient female rats with supraphysiological VD3 supplements was not associated with hypercalcaemia, and could contribute to ovarian functions by regulating the hypothalamic-pituitary-ovarian hormones and ovarian VD-related molecules. However, further studies are still needed to illustrate the clinical significance of VD3 in female reproduction.


Asunto(s)
Colecalciferol/farmacología , Ovario/efectos de los fármacos , Animales , Colecalciferol/administración & dosificación , Colecalciferol/análisis , Suplementos Dietéticos , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Femenino , Ovario/metabolismo , Ratas , Ratas Wistar
5.
Neuroendocrinology ; 111(7): 660-677, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32570260

RESUMEN

INTRODUCTION: The membrane-associated G protein-coupled estrogen receptor 1 (GPER) mediates the regulation by estradiol of arginine-vasopressin immunoreactivity in the supraoptic and paraventricular hypothalamic nuclei of female rats and is involved in the estrogenic control of hypothalamic regulated functions, such as food intake, sexual receptivity, and lordosis behavior. OBJECTIVE: To assess GPER distribution in the rat hypothalamus. METHODS: GPER immunoreactivity was assessed in different anatomical subdivisions of five selected hypothalamic regions of young adult male and cycling female rats: the arcuate nucleus, the lateral hypothalamus, the paraventricular nucleus, the supraoptic nucleus, and the ventromedial hypothalamic nucleus. GPER immunoreactivity was colocalized with NeuN as a marker of mature neurons, GFAP as a marker of astrocytes, and CC1 as a marker of mature oligodendrocytes. RESULTS: GPER immunoreactivity was detected in hypothalamic neurons, astrocytes, and oligodendrocytes. Sex and regional differences and changes during the estrous cycle were detected in the total number of GPER-immunoreactive cells and in the proportion of neurons, astrocytes, and oligodendrocytes that were GPER-immunoreactive. CONCLUSIONS: These findings suggest that estrogenic regulation of hypothalamic function through GPER may be different in males and females and may fluctuate during the estrous cycle in females.


Asunto(s)
Astrocitos/metabolismo , Ciclo Estral/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Caracteres Sexuales , Animales , Femenino , Inmunohistoquímica , Masculino , Ratas , Ratas Wistar
6.
Cell Tissue Res ; 381(3): 451-460, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32710274

RESUMEN

The content of gonadotropin-releasing hormone (GnRH), its mRNA, and estrogen receptor alpha (ERα) and beta (ERß) in the hypothalamus varies throughout the estrous cycle. Furthermore, the abundance of these molecules displays asymmetry between the right and left side. In the present study, we investigated the changes in the content of ERα, ERß, kisspeptin, and GnRH by western blot in the left and right anteromedial hypothalamus, at four different times during each stage of the rat estrous cycle. The serum levels of the follicle-stimulating hormone (FSH) and luteinizing hormone (LH) were also measured. ERα and ERß levels changed depending on the stage of the estrous cycle, meanwhile that of kisspeptin was modified according to both the hour of the day and the stage of the cycle. Except in estrus day, ERß was higher in the right hypothalamus, while ERα was similar in both sides. During both proestrus and estrus, the content of kisspeptin and GnRH was higher in the right hypothalamus. The highest levels of FSH and LH occurred at 17:00 h of proestrus. But at estrus, the highest FSH levels were observed at 08:00 h and the lowest at 17:00 h. Thus, the current results show that the content of ERα, ERß, kisspeptin, and GnRH in the anteromedial hypothalamus are regulated as a function of the stage of the estrous cycle and the hour of the day. Furthermore, the content of these proteins is regularly higher in the right anteromedial hypothalamus, regardless of the stage of the cycle or time of the day.


Asunto(s)
Ciclo Estral/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Femenino , Ratas
7.
Am J Physiol Regul Integr Comp Physiol ; 318(6): R1027-R1035, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32292064

RESUMEN

There are examples of physiological conditions under which thirst is inappropriately exaggerated, and the mechanisms for these paradoxical ingestive behaviors remain unknown. We are interested in thirst mechanisms across the female life cycle and have identified a novel mechanism through which ingestive behavior may be activated. We discovered a previously unrecognized endogenous hypothalamic peptide, phoenixin (PNX), identified physiologically relevant actions of the peptide in brain and pituitary gland to control reproductive hormone secretion in female rodents, and in the process identified the previously orphaned G protein-coupled receptor Gpr173 to be a potential receptor for the peptide. Labeled PNX binding distribution in brain parallels areas known to be important in ingestive behaviors as well in areas where gonadal steroids feedback to control estrous cyclicity (Stein LM, Tullock CW, Mathews SK, Garcia-Galiano D, Elias CF, Samson WK, Yosten GLC, Am J Physiol Regul Integr Comp Physiol 311: R489-R496, 2016). We have demonstrated upregulation of Gpr173 during puberty, fluctuations across the estrous cycle, and, importantly, upregulation during the last third of gestation. It is during this hypervolemic, hyponatremic state that both vasopressin secretion and thirst are inappropriately elevated in humans. Here, we show that central administration of PNX stimulated water drinking in both males and females under ad libitum conditions, increased water drinking after overnight fluid deprivation, and increased both water and 1.5% NaCl ingestion under fed and hydrated conditions. Importantly, losartan pretreatment blocked the effect of PNX on water drinking, and knockdown of Gpr173 by use of short interfering RNA constructs significantly attenuated water drinking in response to overnight fluid deprivation. These actions, together with the stimulatory action of PNX on vasopressin secretion, suggest that this recently discovered neuropeptide may impact the recruitment of critically important neural circuits through which ingestive behaviors and endocrine mechanisms that maintain fluid and electrolyte homeostasis are regulated.


Asunto(s)
Conducta de Ingestión de Líquido/fisiología , Hipotálamo/metabolismo , Hormonas Peptídicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sed/fisiología , Animales , Ciclo Estral/metabolismo , Femenino , Homeostasis/fisiología , Masculino , Hormonas Peptídicas/genética , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética
8.
Reprod Fertil Dev ; 32(6): 610-618, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32209209

RESUMEN

Di-(2-ethylhexyl) phthalate (DEHP) is ubiquitous in the environment and has been proposed to lead to reproductive disruption. In this study, we systematically investigated the effects of different doses of DEHP exposure on female hypothalamic-pituitary-gonadal axis development. Female Sprague-Dawley rats were gavaged with vehicle (corn oil) or DEHP (5 or 500mgkg-1 day-1) during postnatal Days (PNDs) 22-28 or PNDs 22-70. Results demonstrated that the low and high doses of DEHP exerted opposite effects on puberty onset, circulating luteinising hormone, serum oestradiol and progesterone levels, with the low dose (5mgkg-1) promoting and the high dose (500mgkg-1) inhibiting these parameters. Significant dose-related differences were also found in the D500 group with longer oestrous cycle duration, lower ovarian/bodyweight ratio, fewer corpus lutea and more abnormal ovarian stromal tissue in comparison with the oil or D5 groups. Molecular data showed that the hypothalamic Kiss1 mRNA expression in the anteroventral periventricular but not in the arcuate nucleus significantly decreased in the D500 rats and increased in the D5 rats relative to the rats in the oil group. These findings suggested that the kisspeptin system is a potential target for DEHP to disrupt reproductive development and function.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Contaminantes Ambientales/toxicidad , Ciclo Estral/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Kisspeptinas/metabolismo , Periodicidad , Reproducción/efectos de los fármacos , Desarrollo Sexual/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Estradiol/sangre , Ciclo Estral/metabolismo , Femenino , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Progesterona/sangre , Ratas Sprague-Dawley
9.
J Med Food ; 23(4): 375-387, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32045334

RESUMEN

Myo-inositol supplementation may reduce insulin resistance (IR) with few serious side effects in patients with polycystic ovary syndrome (PCOS). To explore the mechanism of this action in an animal model, a PCOS-IR rat model was generated. Enzyme-linked immunosorbent assay was used to assess changes in ovulation function during treatment with a myo-inositol supplement, and Western blotting, real-time polymerase chain reaction, and immunohistochemistry were performed to investigate the underlying molecular mechanisms. The results showed that the myo-inositol supplement decreased the homeostatic model assessment of insulin resistance (HOMA-IR) index and significantly decreased the serum levels of luteinizing hormone (LH), LH/follicle-stimulating hormone ratio, and testosterone, while increasing the serum level of estradiol. Upregulation of interleukin 6 (IL-6), phospho-STAT3 (p-STAT3), Mir-21, and Mir-155 and significant downregulation of PPAR-γ and GLUT4 were detected in the untreated PCOS-IR rat model. However, downregulation of IL-6, p-STAT3, miR-21, and miR-155 and significant upregulation of PPAR-γ and GLUT4 were detected with myo-inositol supplementation. Thus, myo-inositol supplementation may reduce Mir-21 and Mir-155 levels by downregulating IL-6 and p-STAT3 and, subsequently, reverse the expression of PPAR-γ and GLUT4, leading to a decreased HOMA-IR index. In conclusion, the identification of an IL-6/p-STAT3/Mir-155/Mir-21/PPAR-γ/GLUT4 system in the PCOS-IR rat model provides insight into the pathogenesis of PCOS and may indicate a possible therapeutic strategy. Amelioration of the basal serum glucose levels and of the HOMA/HOMA-IR index may be achieved by the reversal of the expression of PPAR-γ and GLUT4 through the downregulation of IL-6, p-STAT3, miR-21, and miR-155 with myo-inositol supplementation.


Asunto(s)
Inositol/farmacología , Resistencia a la Insulina , Interleucina-6/sangre , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Factor de Transcripción STAT3/genética , Animales , Glucemia/metabolismo , Peso Corporal , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Femenino , Hormona Folículo Estimulante/sangre , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/sangre , Hormona Luteinizante/sangre , MicroARNs/genética , MicroARNs/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Testosterona/sangre
10.
Int J Mol Sci ; 20(18)2019 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-31533319

RESUMEN

Induced by a bacterial infection, an immune/inflammatory challenge is a potent negative regulator of the reproduction process in females. The reduction of the synthesis of pro-inflammatory cytokine is considered as an effective strategy in the treatment of inflammatory induced neuroendocrine disorders. Therefore, the effect of direct administration of acetylcholinesterase inhibitor-neostigmine-into the third ventricle of the brain on the gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) secretions under basal and immune stress conditions was evaluated in this study. In the study, 24 adult, 2-years-old Blackhead ewes during the follicular phase of their estrous cycle were used. Immune stress was induced by the intravenous injection of LPS Escherichia coli in a dose of 400 ng/kg. Animals received an intracerebroventricular injection of neostigmine (1 mg/animal) 0.5 h before LPS/saline treatment. It was shown that central administration of neostigmine might prevent the inflammatory-dependent decrease of GnRH/LH secretion in ewes and it had a stimulatory effect on LH release. This central action of neostigmine is connected with its inhibitory action on local pro-inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)α synthesis in the hypothalamus, which indicates the importance of this mediator in the inhibition of GnRH secretion during acute inflammation.


Asunto(s)
Inhibidores de la Colinesterasa/administración & dosificación , Endotoxinas/efectos adversos , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Hormona Liberadora de Gonadotropina/biosíntesis , Hormona Luteinizante/biosíntesis , Neostigmina/administración & dosificación , Fase Folicular/efectos de los fármacos , Fase Folicular/metabolismo , Hidrocortisona/biosíntesis , Hipotálamo/metabolismo , Lipopolisacáridos/efectos adversos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
11.
Am J Physiol Regul Integr Comp Physiol ; 317(2): R328-R336, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31141415

RESUMEN

Nesfatin-1 is a peptide derived from the nucleobindin 2 (Nucb2) precursor protein that has been shown to exert potent effects on appetite and cardiovascular function in male animals. Sex hormones modulate the expression of Nucb2 in several species, including goldfish, mouse, and rat, and human studies have revealed differential expression based on male or female sex. We therefore hypothesized that the ability of nesfatin-1 to increase mean arterial pressure (MAP) would be influenced by stage of the estrous cycle. Indeed, we found that in cycling female Sprague-Dawley rats, nesfatin-1 induced an increase in MAP on diestrus, when both estrogen and progesterone levels are low but not on proestrus or estrus. The effect of nesfatin-1 on MAP was dependent on functional central melanocortin receptors, because the nesfatin-1-induced increase in MAP was abolished by pretreatment with the melanocortin 3/4 receptor antagonist, SHU9119. We previously reported that nesfatin-1 inhibited angiotensin II-induced water drinking in male rats but found no effect of nesfatin-1 in females in diestrus. However, nesfatin-1 enhanced angiotensin II-induced elevations in MAP in females in diestrus but had no effect on males. Finally, in agreement with previous reports, the expression of Nucb2 mRNA in hypothalamus was significantly reduced in female rats in proestrus compared with rats in diestrus. From these data we conclude that the function and expression of nesfatin-1 are modulated by sex hormone status. Further studies are required to determine the contributions of chromosomal sex and individual sex hormones to the cardiovascular effects of nesfatin-1.


Asunto(s)
Ciclo Estral/metabolismo , Hormonas/metabolismo , Nucleobindinas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Femenino , Hipotálamo/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Hormonas Peptídicas/metabolismo , Ratas Sprague-Dawley , Receptores de Melanocortina/metabolismo
12.
Endocrinology ; 160(6): 1547-1560, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31127273

RESUMEN

Sirtuin 1 (Sirt1) is an NAD-dependent class III deacetylase that functions as a cellular energy sensor. In addition to its well-characterized effects in peripheral tissues, evidence suggests that SIRT1 in neurons plays a role in the central regulation of energy balance and reproduction, but no studies have addressed the contribution of astrocytes. We show here that overexpression of SIRT1 in astrocytes causes markedly increased food intake, body weight gain, and glucose intolerance, but expression of a deacetylase-deficient SIRT1 mutant decreases food intake and body weight and improves glucose tolerance, particularly in female mice. Paradoxically, the effect of these SIRT1 mutants on insulin tolerance was reversed, with overexpression showing greater insulin sensitivity. The mice overexpressing SIRT1 were more active, generated more heat, and had elevated oxygen consumption, possibly in compensation for the increased food intake. The female overexpressing mice were also more sensitive to diet-induced obesity. Reproductively, the mice expressing the deacetylase-deficient SIRT1 mutant had impaired estrous cycles, decreased LH surges, and fewer corpora lutea, indicating decreased ovulation. The GnRH neurons were responsive to kisspeptin stimulation, but hypothalamic expression of Kiss1 was reduced in the mutant mice. Our results showed that SIRT1 signaling in astrocytes can contribute to metabolic and reproductive regulation independent of SIRT1 effects in neurons.


Asunto(s)
Astrocitos/metabolismo , Ingestión de Alimentos/fisiología , Ciclo Estral/metabolismo , Intolerancia a la Glucosa/metabolismo , Glucosa/metabolismo , Sirtuina 1/metabolismo , Aumento de Peso/fisiología , Animales , Ciclo Estral/genética , Femenino , Hormona Folículo Estimulante/sangre , Intolerancia a la Glucosa/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Resistencia a la Insulina/fisiología , Leptina/sangre , Hormona Luteinizante/sangre , Masculino , Ratones , Neuronas/metabolismo , Sirtuina 1/genética , Testículo/metabolismo
13.
Reproduction ; 157(4): 399-411, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30763281

RESUMEN

Successful bovine pregnancy establishment hinges on conceptus elongation, a key reproductive phenomenon coinciding with the period during which most pregnancies fail. Elongation is yet to be recapitulated in vitro, whereas in vivo it is directly driven by uterine secretions and indirectly influenced by prior circulating progesterone levels. To better understand the microenvironment evolved to facilitate this fundamental developmental event, uterine fluid was recovered on Days 12-14 of the oestrous cycle - the window of conceptus elongation initiation - from cycling heifers supplemented, or not, with progesterone. Subsequent lipidomic profiling of uterine luminal fluid by advanced high-throughput metabolomics revealed the consistent presence of 75 metabolites, of which 47% were intricately linked to membrane biogenesis, and with seven displaying a day by progesterone interaction (P ≤ 0.05). Four metabolic pathways were correspondingly enriched according to day and P4 - i.e. comprised metabolites whose concentrations differed between groups (normal vs high P4) at different times (Days 12 vs 13 vs 14). These were inositol, phospholipid, glycerolipid and primary bile acid metabolism. Moreover, P4 elevated total uterine luminal fluid lipid content on Day 14 (P < 0.0001) relative to all other comparisons. The data combined suggest that maternal lipid supply during the elongation-initiation window is primarily geared towards conceptus membrane biogenesis. In summary, progesterone supplementation alters the lipidomic profile of bovine uterine fluid during the period of conceptus elongation initiation.


Asunto(s)
Embrión de Mamíferos/metabolismo , Ciclo Estral/metabolismo , Lípidos/análisis , Metaboloma , Progesterona/farmacología , Útero/metabolismo , Animales , Bovinos , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Ciclo Estral/efectos de los fármacos , Femenino , Embarazo , Útero/efectos de los fármacos
14.
Reprod Sci ; 24(9): 1293-1303, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28814190

RESUMEN

It is now established that mycoestrogen zearalenone (ZEN) disrupts reproductive physiology, but the specific mechanisms by which this occurs remain unknown, especially in brain. Growing evidence suggests that populations of estradiol (E2)-sensitive neurons in anteroventral periventricular (AVPV) and arcuate (ARC) nuclei, especially kisspeptin neurons, play a pivotal role in the timing of puberty onset, ovulation, and normal reproduction. The present study was conducted to find whether the ZEN can cause estrogen-like actions during the critical period of neonatal differentiation. In this study, we compared the effect of neonatal exposure to sesame oil, E2 benzoate (EB, 20 µg/kg body weight [bw]), and 3 various doses: 0.2, 1, and 2 mg/kg bw of ZEN (0.2, 1, and 2 ZEN) on the onset of puberty and estrus cyclicity as well as ovarian follicular profile, kisspeptin expression, and neuronal density in AVPV and ARC hypothalamic nuclei and E2 and luteinizing hormone (LH) levels on postnatal day 70. Control mice received no treatment. Vaginal opening was significantly advanced by EB and 2 ZEN. Disrupted estrus cycles and decreased follicular profiles were observed in EB, 1 ZEN, and 2 ZEN animals. In addition, EB, 1 ZEN, and 2 ZEN reduced the expression of kisspeptin and neuronal density of AVPV and ARC nuclei and caused a decrease in the LH and an increase in E2 plasma levels. Taken together, our observations provide physiological evidence that neonatal exposure to ZEN exerts estrogen-like actions in the estrogen-sensitive hypothalamic AVPV and ARC nuclei, controlling reproductive functions in adult female mice.


Asunto(s)
Ciclo Estral/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Reproducción/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Zearalenona/farmacología , Animales , Relación Dosis-Respuesta a Droga , Estradiol/análogos & derivados , Estradiol/sangre , Estradiol/farmacología , Ciclo Estral/metabolismo , Femenino , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormona Luteinizante/sangre , Ratones , Ratones Endogámicos BALB C , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo
15.
Sci Rep ; 7(1): 8096, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28808344

RESUMEN

Progesterone can block the oestradiol-induced GnRH/LH surge and inhibit LH pulse frequency. Recent studies reported that progesterone prevented premature LH surges during ovarian hyperstimulation in women. As the most potent stimulator of GnRH/LH release, kisspeptin is believed to mediate the positive and negative feedback effects of oestradiol in the hypothalamic anteroventral periventricular (AVPV) and arcuate (ARC) nuclei, while the region-specific role of progesterone receptors in these nuclei remains unknown. This study examined the hypothesis that progesterone inhibits LH surge and pulsatile secretion via its receptor in the ARC and/or AVPV nuclei. Adult female rats received a single injection of pregnant mare serum gonadotropin followed by progesterone or vehicle. Progesterone administration resulted in a significant prolongation of the oestrous cycle and blockade of LH surge. However, microinjection of the progesterone receptor antagonist, RU486, into the AVPV reversed the prolonged cycle length and rescued the progesterone blockade LH surge, while RU486 into the ARC shortened LH pulse interval in the progesterone treated rats. These results demonstrated that progesterone's inhibitory effect on the GnRH/LH surge and pulsatile secretion is mediated by its receptor in the kisspeptin enriched hypothalamic AVPV and ARC respectively, which are essential for progesterone regulation of oestrous cyclicity in rats.


Asunto(s)
Hipotálamo/efectos de los fármacos , Hormona Luteinizante/metabolismo , Progesterona/farmacología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Estradiol/metabolismo , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Hipotálamo Anterior/efectos de los fármacos , Hipotálamo Anterior/metabolismo , Mifepristona/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Progesterona/metabolismo
16.
J Comp Neurol ; 525(15): 3360-3387, 2017 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-28685836

RESUMEN

We analyzed the transcriptome of the C57BL/6J mouse hypothalamus, hippocampus, neocortex, and cerebellum to determine estrous cycle-specific changes in these four brain regions. We found almost 16,000 genes are present in one or more of the brain areas but only 210 genes, ∼1.3%, are significantly changed as a result of the estrous cycle. The hippocampus has the largest number of differentially expressed genes (DEGs) (82), followed by the neocortex (76), hypothalamus (63), and cerebellum (26). Most of these DEGs (186/210) are differentially expressed in only one of the four brain regions. A key finding is the unique expression pattern of growth hormone (Gh) and prolactin (Prl). Gh and Prl are the only DEGs to be expressed during only one stage of the estrous cycle (metestrus). To gain insight into the function of the DEGs, we examined gene ontology and phenotype enrichment and found significant enrichment for genes associated with myelination, hormone stimulus, and abnormal hormone levels. Additionally, 61 of the 210 DEGs are known to change in response to estrogen in the brain. 50 of the 210 genes differentially expressed as a result of the estrous cycle are related to myelin and oligodendrocytes and 12 of the 63 DEGs in the hypothalamus are oligodendrocyte- and myelin-specific genes. This transcriptomic analysis reveals that gene expression in the female mouse brain is remarkably stable during the estrous cycle and demonstrates that the genes that do fluctuate are functionally related.


Asunto(s)
Cerebelo/metabolismo , Ciclo Estral/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Neocórtex/metabolismo , Transcriptoma/fisiología , Animales , Femenino , Expresión Génica/fisiología , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN
17.
Neuro Endocrinol Lett ; 38(6): 422-428, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29298283

RESUMEN

OBJECTIVE: We inferred how KISS-1/GPR54 system to involved in precocious puberty by observing hormones level during the process of precocious puberty in model and normal rats during sexual development and the estrus cycle. METHOD: Female rats were divided randomly into CPP and control groups; the former were injected with NMDA twice daily, and control groups were injected with saline. Blood and tissue samples were collected and measured during the stages of prepuberty, vaginal opening, estrus, proestrus and diestrus. RESULTS: The times of onset of puberty and sexual maturity in the CPP group were significantly earlier than in the control groups. Hypothalamic levels of KISS-1 and GPR54 gene expression, kisspeptin, luteinizing hormone, and follicle stimulating hormone started to rise before puberty. In stable estrus cycles, kisspeptin levels were the lowest during proestrus, while gonadotropin-releasing hormone (GnRH) levels rose to the highest during estrus. GnRH levels increased significantly in the estrus cycle compared with the prepubertal stage, but kisspeptin levels did not change significantly. CONCLUSION: the hypothalamic KISS-1/GPR54 system might permit the onset of puberty, but is not its primary trigger. Hormone levels were lower and gonadal maturity markers in the CPP groups were worse than in the control groups.


Asunto(s)
Ciclo Estral/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Receptores de Kisspeptina-1/metabolismo , Maduración Sexual/fisiología , Animales , Ciclo Estral/efectos de los fármacos , Ciclo Estral/genética , Femenino , Hormona Folículo Estimulante/sangre , Hipotálamo/efectos de los fármacos , Kisspeptinas/sangre , Kisspeptinas/genética , Hormona Luteinizante/sangre , N-Metilaspartato/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Kisspeptina-1/genética , Maduración Sexual/efectos de los fármacos , Frotis Vaginal
18.
Aging (Albany NY) ; 9(1): 156-172, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27997360

RESUMEN

Salvia miltiorrhiza bunge(SM) is a popular herb for alleviating menopausal symptoms, although the scientific evidence of applying SM to estrogen replacement therapy is limited. In this study, we characterized the estrogenic activity of SM using in vivo models of immature and ovariectomized (OVX) mice and performed in vitro studies focusing on the estrogen receptor (ER) pathway for further molecular characterizations. SM treatments demonstrated significant estrogenic activity by promoting the development of uterus and vagina in immature mice, restoring the estrus cycle and reversing the atrophy of reproductive tissues in OVX mice, as well as increasing the expressions of ERα and ERß at protein and mRNA level in the reproductive tissues. Meanwhile, SM significantly increased estradiol in serum, and decreased follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in the circulation of immature and OVX mice. SM could stimulate the binding effect of ERα and ERß, and significantly induce ERα/ß-estrogen response element (ERE) luciferase reporter gene expression. All these activities were inhibited by the ER antagonist ICI182, 780. This study demonstrates SM exerts estrogenic effects by stimulating biosynthesis of estrogen and increasing ERs in target tissues without side effects on reproductive tissues and through ER-ERE-dependent pathway.


Asunto(s)
Estradiol/sangre , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Extractos Vegetales/farmacología , Salvia miltiorrhiza , Útero/efectos de los fármacos , Vagina/efectos de los fármacos , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas del Receptor de Estrógeno/farmacología , Ciclo Estral/efectos de los fármacos , Ciclo Estral/metabolismo , Femenino , Fulvestrant , Ratones , Ovariectomía , Útero/metabolismo , Vagina/metabolismo
19.
Endocrinology ; 157(12): 4803-4816, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27732087

RESUMEN

A temporary and reversible inhibition of the hypothalamo-pituitary-gonadal axis is adaptive when energy reserves are diminished, allowing individual survival and energy accumulation for eventual reproduction. The AMP-activated protein kinase (AMPK) works as a cellular sensor of the AMP to ATP ratio and ultimately of energy availability. Activation of AMPK suppresses ATP-consuming processes and stimulates ATP-producing pathways. The AMPK α2 catalytic subunit is expressed in multiple hypothalamic nuclei including those associated with reproductive control, ie, the anteroventral periventricular nucleus and the arcuate nucleus. Subsets of kisspeptin neurons in the anteroventral periventricular nucleus (20% in females) and arcuate nucleus (45% in males and 65% in females) coexpress AMPKα2 mRNA. Using the Cre-loxP approach, we assessed whether AMPKα2 in Kiss1 cells is required for body weight and reproductive function. The AMPKα2-deleted mice show no difference in body weight and time for sexual maturation compared with controls. Males and females are fertile and have normal litter size. The AMPKα2-deleted and control females have similar estradiol feedback responses and show no difference in Kiss1 mRNA expression after ovariectomy or ovariectomy plus estradiol replacement. In males, acute fasting decreased Kiss1 mRNA expression in both groups, but no effect was observed in females. However, after an acute fasting, control mice displayed prolonged diestrous phase, but AMPKα2-deleted females showed no disruption of estrous cycles. Our findings demonstrate that the AMPKα2 catalytic subunit in Kiss1 cells is dispensable for body weight and reproductive function in mice but is necessary for the reproductive adaptations to conditions of acute metabolic distress.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Ayuno/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neuronas/metabolismo , Maduración Sexual/fisiología , Animales , Peso Corporal/fisiología , Ciclo Estral/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados
20.
Endocrinology ; 157(12): 4794-4802, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27715255

RESUMEN

Using a new tail-tip bleeding procedure and a sensitive ELISA, we describe here the patterns of LH secretion throughout the mouse estrous cycle; in ovariectomized mice; in ovariectomized, estradiol-treated mice that model estrogen-negative and -positive feedback; and in transgenic GNR23 mice that exhibit allele-dependent reductions in GnRH neuron number. Pulsatile LH secretion was evident at all stages of the estrous cycle, with LH pulse frequency being approximately one pulse per hour in metestrous, diestrous, and proestrous mice but much less frequent at estrus (less than one pulse per 4 h). Ovariectomy resulted in substantial increases in basal and pulsatile LH secretion with pulses occurring approximately every 21 minutes. Chronic treatment with negative-feedback, estradiol-filled capsules returned LH pulse frequency to intact follicular phase levels, although pulse amplitude remained elevated. On the afternoon of proestrus, the LH surge was found to begin in a highly variable manner over a 4-hour range, lasting for more than 3 hours. In contrast, ovariectomized, estradiol-treated, positive-feedback mice exhibited a relatively uniform surge onset at approximately 0.5 hour prior to lights out. Gonadectomized wild-type and heterozygous GNR23 (∼200 GnRH neurons) male mice exhibited an LH pulse every 60 minutes. Homozygous GNR23 mice (∼80 GnRH neurons) had very low basal LH concentrations but continued to exhibit small amplitude LH pulses every 90 minutes. These studies provide the first characterization in mice of pulse and surge modes of LH secretion across the estrous cycle and demonstrate that very few GnRH neurons are required for pulsatile LH secretion.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/metabolismo , Neuronas/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática , Estradiol/farmacología , Ciclo Estral/metabolismo , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Ovariectomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA