Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 4776, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36959275

RESUMEN

Decreased estrogens during menopause are associated with increased risk of anxiety, depression, type 2 diabetes and obesity. Similarly, depleting estrogens in rodents by ovariectomy, combined with a high-fat diet (HFD), increases anxiety and adiposity. How estrogens and diet interact to affect anxiety and metabolism is poorly understood. Mounting evidence indicates that gut microbiota influence anxiety and metabolism. Here, we investigated the effects of estradiol (E) and HFD on anxiety, metabolism, and their correlation with changes in gut microbiota in female mice. Adult C57BL/6J mice were ovariectomized, implanted with E or vehicle-containing capsules and fed a standard diet or HFD. Anxiety-like behavior was assessed and neuronal activation was measured by c-fos immunoreactivity throughout the brain using iDISCO. HFD increased anxiety-like behavior, while E reduced this HFD-dependent anxiogenic effect. Interestingly, E decreased neuronal activation in brain regions involved in anxiety and metabolism. E treatment also altered gut microbes, a subset of which were associated with anxiety-like behavior. These findings provide insight into gut microbiota-based therapies for anxiety and metabolic disorders associated with declining estrogens in menopausal women.


Asunto(s)
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Femenino , Animales , Ratones , Estradiol/farmacología , Dieta Alta en Grasa/efectos adversos , Diabetes Mellitus Tipo 2/complicaciones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Ansiedad/etiología , Estrógenos/farmacología , Factores Inmunológicos/farmacología
2.
Front Endocrinol (Lausanne) ; 13: 1010806, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36387852

RESUMEN

Estrogens protect against weight gain and metabolic disruption in women and female rodents. Aberrations in the gut microbiota composition are linked to obesity and metabolic disorders. Furthermore, estrogen-mediated protection against diet-induced metabolic disruption is associated with modifications in gut microbiota. In this study, we tested if estradiol (E2)-mediated protection against obesity and metabolic disorders in female mice is dependent on gut microbiota. Specifically, we tested if fecal microbiota transplantation (FMT) from E2-treated lean female mice, supplemented with or without Akkermansia muciniphila, prevented high fat diet (HFD)-induced body weight gain, fat mass gain, and hyperglycemia in female recipients. FMT from, and cohousing with, E2-treated lean donors was not sufficient to transfer the metabolic benefits to the E2-deficient female recipients. Moreover, FMT from lean donors supplemented with A. muciniphila exacerbated HFD-induced hyperglycemia in E2-deficient recipients, suggesting its detrimental effect on the metabolic health of E2-deficient female rodents fed a HFD. Given that A. muciniphila attenuates HFD-induced metabolic insults in males, the present findings suggest a sex difference in the impact of this microbe on metabolic health.


Asunto(s)
Dieta Alta en Grasa , Hiperglucemia , Femenino , Ratones , Masculino , Animales , Dieta Alta en Grasa/efectos adversos , Akkermansia , Trasplante de Microbiota Fecal , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/terapia , Obesidad/metabolismo , Aumento de Peso
3.
Metabolites ; 11(8)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34436440

RESUMEN

A decrease in ovarian estrogens in postmenopausal women increases the risk of weight gain, cardiovascular disease, type 2 diabetes, and chronic inflammation. While it is known that gut microbiota regulates energy homeostasis, it is unclear if gut microbiota is associated with estradiol regulation of metabolism. In this study, we tested if estradiol-mediated protection from high-fat diet (HFD)-induced obesity and metabolic changes are associated with longitudinal alterations in gut microbiota in female mice. Ovariectomized adult mice with vehicle or estradiol (E2) implants were fed chow for two weeks and HFD for four weeks. As reported previously, E2 increased energy expenditure, physical activity, insulin sensitivity, and whole-body glucose turnover. Interestingly, E2 decreased the tight junction protein occludin, suggesting E2 affects gut epithelial integrity. Moreover, E2 increased Akkermansia and decreased Erysipleotrichaceae and Streptococcaceae. Furthermore, Coprobacillus and Lactococcus were positively correlated, while Akkermansia was negatively correlated, with body weight and fat mass. These results suggest that changes in gut epithelial barrier and specific gut microbiota contribute to E2-mediated protection against diet-induced obesity and metabolic dysregulation. These findings provide support for the gut microbiota as a therapeutic target for treating estrogen-dependent metabolic disorders in women.

4.
Trends Endocrinol Metab ; 32(8): 554-565, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34049772

RESUMEN

This review discusses the interactions of steroids with the gut and vaginal microbiomes within each life phase of adult women and the implications for women's health. Each phase of a woman's life is characterized by distinct hormonal states which drive overall physiology of both host and commensal microbes. These host-microbiome interactions underlie disease pathology in disorders that affect women across their lifetime, including bacterial vaginosis, gestational diabetes, polycystic ovary syndrome (PCOS), anxiety, depression, and obesity. Although many associations between host health and microbiome composition are well defined, the mechanistic role of the microbiome in women's health outcomes is largely unknown. This review addresses potential mechanisms by which the microbiota influences women's health and highlights gaps in current knowledge.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Esteroides , Vagina , Salud de la Mujer , Adulto , Femenino , Humanos , Vagina/microbiología , Vaginosis Bacteriana
6.
J Neuroendocrinol ; 32(10): e12904, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33000549

RESUMEN

Neural progestin receptors (PR) function in reproduction, neural development, neuroprotection, learning, memory and the anxiety response. In the absence of progestins, PR can be activated by dopamine (DA) in the rodent hypothalamus to elicit female sexual behaviour. The present study investigated mechanisms of DA activation of PR by testing the hypothesis that proteins from DA-treated hypothalami interact with PR in the absence of progestins. Ovariectomised, oestradiol-primed mice were infused with a D1-receptor agonist, SKF38393 (SKF), into the third ventricle 30 minutes prior to death. Proteins from SKF-treated hypothalami were pulled-down with glutathione S-transferase-tagged mouse PR-A or PR-B and the interactomes were analysed by mass spectrometry. The largest functional group to interact with PR-A in a DA-dependent manner was synaptic proteins. To test the hypothesis that DA activation of PR regulates synaptic proteins, we developed oestradiol-induced PR-expressing hypothalamic-like neurones derived from human-induced pluripotent stem cells (hiPSCs). Similar to progesterone (P4), SKF treatment of hiPSCs increased synapsin1/2 expression. This SKF-dependent effect was blocked by the PR antagonist RU486, suggesting that PR are necessary for this DA-induced increase. The second largest DA-dependent PR-A protein interactome comprised metabolic regulators involved in glucose metabolism, lipid synthesis and mitochondrial energy production. Interestingly, hypothalamic proteins interacted with PR-A, but not PR-B, in an SKF-dependent manner, suggesting that DA promotes the interaction of multiple hypothalamic proteins with PR-A. These in vivo and in vitro results indicate novel mechanisms by which DA can differentially activate PR isoforms in the absence of P4 and provide a better understanding of ligand-independent PR activation in reproductive, metabolic and mental health disorders in women.


Asunto(s)
Dopamina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Progesterona/metabolismo , Animales , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Progesterona/farmacología , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
7.
mSphere ; 5(4)2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641429

RESUMEN

The microorganisms of the vaginal tract are critical for vaginal and reproductive health. However, the regulation of these microorganisms is not well understood. Therefore, we investigated whether different factors regulate the vaginal microbiota of healthy college-aged women (n = 26) with high temporal resolution by collecting daily self-administered vaginal swabs and using 16S rRNA sequencing for bacterial identification. As expected, vaginal microbiota clustered into five predefined community state types. Vaginal microbial diversity, stability, and Lactobacillus abundances were associated with the menstrual cycle and hormonal contraceptive use. Vaginal microbial diversity, as measured using the Shannon index, increased during menses (P < 0.001), while Lactobacillus abundances decreased (P = 0.01). The covariance of these microbial measures with previously established estradiol levels suggests that estrogens can regulate vaginal microbiota. Moreover, the use of hormonal contraceptives may alter the temporal dynamics of the vaginal microbiota and decrease Lactobacillus abundances, depending on hormonal content and release method. Interestingly, intrasample diversity was greater in participants on a vegetarian diet (P = 0.004) and among participants who exercised more (P = 0.04). These findings indicate that ovarian hormones, diet, and exercise can regulate vaginal microbial composition and stability and may impact vaginal and reproductive health.IMPORTANCE The vaginal microbiome is a critical component of women's sexual and reproductive health, with variations in microbial composition, particularly the loss of Lactobacillus species, being implicated in gynecologic and obstetric diseases. Given that the vaginal microbiome is so crucial, why do vaginal microbial profiles vary strikingly from person to person and even change over time within the same person? In the present study, which tracked the daily vaginal microbiomes of young healthy women through different lifestyles, we found that use of a locally released progestin contraceptive, a vegetarian diet, and intense exercise appear to lead to vaginal microbiome alterations and loss of Lactobacillus species. The impact of these vaginal microbiome changes on immediate and long-term health remain to be investigated.


Asunto(s)
Anticonceptivos , Dieta , Ejercicio Físico , Hormonas Esteroides Gonadales/fisiología , Ciclo Menstrual/fisiología , Microbiota/genética , Vagina/microbiología , Adolescente , Femenino , Variación Genética , Humanos , Lactobacillus/genética , Estudios Longitudinales , Embarazo , ARN Ribosómico 16S/genética , Salud de la Mujer , Adulto Joven
8.
Sci Rep ; 9(1): 20192, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882890

RESUMEN

Estrogens protect against diet-induced obesity in women and female rodents. For example, a lack of estrogens in postmenopausal women is associated with an increased risk of weight gain, cardiovascular diseases, low-grade inflammation, and cancer. Estrogens act with leptin to regulate energy homeostasis in females. Leptin-deficient mice (ob/ob) exhibit morbid obesity and insulin resistance. The gut microbiome is also critical in regulating metabolism. The present study investigates whether estrogens and leptin modulate gut microbiota in ovariectomized ob/ob (obese) or heterozygote (lean) mice fed high-fat diet (HFD) that received either 17ß-Estradiol (E2) or vehicle implants. E2 attenuated weight gain in both genotypes. Moreover, both obesity (ob/ob mice) and E2 were associated with reduced gut microbial diversity. ob/ob mice exhibited lower species richness than control mice, while E2-treated mice had reduced evenness compared with vehicle mice. Regarding taxa, E2 was associated with an increased abundance of the S24-7 family, while leptin was associated with increases in Coriobacteriaceae, Clostridium and Lactobacillus. Some taxa were affected by both E2 and leptin, suggesting these hormones alter gut microbiota of HFD-fed female mice. Understanding the role of E2 and leptin in regulating gut microbiota will provide important insights into hormone-dependent metabolic disorders in women.


Asunto(s)
Dieta Alta en Grasa , Estradiol/administración & dosificación , Microbioma Gastrointestinal , Animales , Estradiol/metabolismo , Conducta Alimentaria , Femenino , Resistencia a la Insulina , Leptina/genética , Leptina/metabolismo , Ratones , Ratones Obesos , Aumento de Peso
9.
eNeuro ; 4(5)2017.
Artículo en Inglés | MEDLINE | ID: mdl-28955722

RESUMEN

Progestins bind to the progestin receptor (PR) isoforms, PR-A and PR-B, in brain to influence development, female reproduction, anxiety, and stress. Hormone-activated PRs associate with multiple proteins to form functional complexes. In the present study, proteins from female mouse hypothalamus that associate with PR were isolated using affinity pull-down assays with glutathione S-transferase-tagged mouse PR-A and PR-B. Using complementary proteomics approaches, reverse phase protein array (RPPA) and mass spectrometry, we identified hypothalamic proteins that interact with PR in a ligand-dependent and isoform-specific manner and were confirmed by Western blot. Synaptic proteins, including synapsin-I and synapsin-II, interacted with agonist-bound PR isoforms, suggesting that both isoforms function in synaptic plasticity. In further support, synaptogyrin-III and synapsin-III associated with PR-A and PR-B, respectively. PR also interacted with kinases, including c-Src, mTOR, and MAPK1, confirming phosphorylation as an integral process in rapid effects of PR in the brain. Consistent with a role in transcriptional regulation, PR associated with transcription factors and coactivators in a ligand-specific and isoform-dependent manner. Interestingly, both PR isoforms associated with a key regulator of energy homeostasis, FoxO1, suggesting a novel role for PR in energy metabolism. Because many identified proteins in this PR interactome are synaptic proteins, we tested the hypothesis that progestins function in synaptic plasticity. Indeed, progesterone enhanced synaptic density, by increasing synapsin-I-positive synapses, in rat primary cortical neuronal cultures. This novel combination of RPPA and mass spectrometry allowed identification of PR action in synaptic remodeling and energy homeostasis and reveals unique roles for progestins in brain function and disease.


Asunto(s)
Hipotálamo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Progesterona/metabolismo , Sinapsis/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Estradiol/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión Transferasa/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Ovariectomía , Unión Proteica , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcripción Genética
10.
eNeuro ; 3(4)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27679811

RESUMEN

Estrogens and leptins act in the hypothalamus to maintain reproduction and energy homeostasis. Neurogenesis in the adult mammalian hypothalamus has been implicated in the regulation of energy homeostasis. Recently, high-fat diet (HFD) and estradiol (E2) have been shown to alter cell proliferation and the number of newborn leptin-responsive neurons in the hypothalamus of adult female mice. The current study tested the hypothesis that new cells expressing estrogen receptor α (ERα) are generated in the arcuate nucleus (ARC) and the ventromedial nucleus of the hypothalamus (VMH) of the adult female mouse, hypothalamic regions that are critical in energy homeostasis. Adult mice were ovariectomized and implanted with capsules containing E2 or oil. Within each hormone group, mice were fed an HFD or standard chow for 6 weeks and treated with BrdU to label new cells. Newborn cells that respond to estrogens were identified in the ARC and VMH, of which a subpopulation was leptin sensitive, indicating that the subpopulation consists of neurons. Moreover, there was an interaction between diet and hormone with an effect on the number of these newborn ERα-expressing neurons that respond to leptin. Regardless of hormone treatment, HFD increased the number of ERα-expressing cells in the ARC and VMH. E2 decreased hypothalamic fibroblast growth factor 10 (Fgf10) gene expression in HFD mice, suggesting a role for Fgf10 in E2 effects on neurogenesis. These findings of newly created estrogen-responsive neurons in the adult brain provide a novel mechanism by which estrogens can act in the hypothalamus to regulate energy homeostasis in females.

11.
eNeuro ; 2(4)2015.
Artículo en Inglés | MEDLINE | ID: mdl-26465008

RESUMEN

Estrogens act in brain to profoundly influence neurogenesis, sexual differentiation, neuroprotection, cognition, energy homeostasis, and female reproductive behavior and physiology through a variety of mechanisms, including the induction of progestin receptors (PRs). PRs are expressed as two isoforms, PR-A and PR-B, that have distinct functions in physiology and behavior. Because these PR isoforms cannot be distinguished using cellular resolution techniques, the present study used isoform-specific null mutant mice that lack PR-A or PR-B for the first time to investigate whether 17ß-estradiol benzoate (EB) regulates the differential expression of the PR isoforms in the ventromedial nucleus of the hypothalamus (VMN), arcuate nucleus, and medial preoptic area, brain regions that are rich in EB-induced PRs. Interestingly, EB induced more PR-A than PR-B in all three brain regions, suggesting that PR-A is the predominant isoform in these regions. Given that steroid receptor coactivator (SRC)-1 and SRC-2 are important in estrogen receptor (ER)-dependent transcription in brain, including PR induction, we tested whether the expression of these coactivators was correlated with PR isoform expression. The majority of EB-induced PR cells expressed both SRC-1 and SRC-2 in the three brain regions of all genotypes. Interestingly, the intensity of PR-A immunoreactivity correlated with SRC-2 expression in the VMN, providing a potential mechanism for selective ER-mediated transactivation of PR-A over PR-B in a brain region-specific manner. In summary, these novel findings indicate that estrogens differentially regulate PR-A and PR-B expression in the female hypothalamus, and provide a mechanism by which steroid action in brain can selectively modulate behavior and physiology.

13.
Steroids ; 103: 23-30, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25824325

RESUMEN

In the present review we summarize observations to date supporting the concept that neuroactive steroids are synthesized in the peripheral nervous system, regulate the physiology of peripheral nerves and exert notable neuroprotective actions. Indeed, neuroactive steroids have been recently proposed as therapies for different types of peripheral neuropathy, like for instance those occurring during aging, chemotherapy, physical injury and diabetes. Moreover, pharmacological tools able to increase the synthesis of neuroactive steroids might represent new interesting therapeutic strategy to be applied in case of peripheral neuropathy.


Asunto(s)
Neurotransmisores/farmacología , Sistema Nervioso Periférico/efectos de los fármacos , Esteroides/farmacología , Animales , Humanos
15.
Pharmacol Res ; 79: 13-20, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24184124

RESUMEN

In the initial stages, human prostate cancer (PC) is an androgen-sensitive disease, which can be pharmacologically controlled by androgen blockade. This therapy often induces selection of androgen-independent PC cells with increased invasiveness. We recently demonstrated, both in cells and mice, that a testosterone metabolite locally synthetized in prostate, the 5α-androstane-3ß, 17ß-diol (3ß-Adiol), inhibits PC cell proliferation, migration and invasion, acting as an anti-proliferative/anti-metastatic agent. 3ß-Adiol is unable to bind androgen receptor (AR), but exerts its protection against PC by specifically interacting with estrogen receptor beta (ERß). Because of its potential retro-conversion to androgenic steroids, 3ß-Adiol cannot be used "in vivo", thus, the aims of this study were to investigate the capability of four ligands of ERß (raloxifen, tamoxifen, genistein and curcumin) to counteract PC progression by mimicking the 3ß-Adiol activity. Our results demonstrated that raloxifen, tamoxifen, genistein and curcumin decreased DU145 and PC3 cell proliferation in a dose-dependent manner; in addition, all four compounds significantly decreased the detachment of cells seeded on laminin or fibronectin. Moreover, raloxifen, tamoxifen, genistein and curcumin-treated DU145 and PC3 cells showed a significant decrease in cell migration. Notably, all these effects were reversed by the anti-estrogen, ICI 182,780, suggesting that their actions are mediated by the estrogenic pathway, via the ERß, the only isoform present in these PCs. In conclusion, these data demonstrate that by selectively activating the ERß, raloxifen, tamoxifen, genistein and curcumin inhibit human PC cells proliferation and migration favoring cell adesion. These synthetic and natural modulators of ER action may exert a potent protective activity against the progression of PC even in its androgen-independent status.


Asunto(s)
Antineoplásicos/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Estradiol/análogos & derivados , Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Fulvestrant , Genisteína/farmacología , Humanos , Masculino , Neoplasias de la Próstata , Clorhidrato de Raloxifeno/farmacología , Tamoxifeno/farmacología
16.
Neuropeptides ; 46(4): 167-72, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22672888

RESUMEN

Thimet oligopeptidase (TOP) and prolyl endopeptidase (PEP) are neuropeptidases involved in the hydrolysis of gonadotropin-releasing hormone, a key component of the hypothalamic-pituitary-gonadal axis. GnRH is regulated in part by feedback from steroid hormones such as estradiol. Previously, we demonstrated that TOP levels are down-regulated by estradiol in reproductively-relevant regions of the female rodent brain. The present study supports these findings by showing that TOP enzyme activity, as well as protein levels, in the ventromedial hypothalamic nucleus of female mice is controlled by estradiol. We further demonstrate that PEP levels in this same brain region are down-regulated by estradiol in parallel with those of TOP. These findings provide evidence that these neuropeptidases are part of the fine control of hormone levels in the HPG axis.


Asunto(s)
Estradiol/farmacología , Hipotálamo/enzimología , Metaloendopeptidasas/metabolismo , Serina Endopeptidasas/metabolismo , Núcleo Hipotalámico Ventromedial/enzimología , Animales , Regulación hacia Abajo , Activación Enzimática/efectos de los fármacos , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Ratones , Ratones Endogámicos C57BL , Prolil Oligopeptidasas , Esteroides/metabolismo
17.
Neuroendocrinology ; 94(1): 49-57, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21311177

RESUMEN

BACKGROUND/AIMS: The steroid hormones, including estradiol (E) and progesterone, act in the brain to regulate female reproductive behavior and physiology. These hormones mediate many of their biological effects by binding to their respective intracellular receptors. The receptors for estrogens (ER) and progestins (PR) interact with nuclear receptor coactivators to initiate transcription of steroid-responsive genes. Work from our laboratory and others reveals that nuclear receptor coactivators, including steroid receptor coactivator-1 (SRC-1) and SRC-2, function in brain to modulate ER-mediated induction of the PR gene and hormone-dependent behaviors. In order for steroid receptors and coactivators to function together, both must be expressed in the same cells. METHODS: Triple-label immunofluorescence was used to determine if E-induced PR cells also express SRC-1 or SRC-2 in reproductively relevant brain regions of the female mouse. RESULTS: The majority of E-induced PR cells in the medial preoptic area (61%), ventromedial nucleus of the hypothalamus (63%) and arcuate nucleus (76%) coexpressed both SRC-1 and SRC-2. A smaller proportion of PR cells expressed either SRC-1 or SRC-2, while a few PR cells expressed neither coactivator. In addition, compared to control animals, 17ß-estradiol benzoate (EB) treatment increased SRC-1 levels in the arcuate nucleus, but not the medial preoptic area or the ventromedial nucleus of the hypothalamus. EB did not alter SRC-2 expression in any of the three brain regions analyzed. CONCLUSIONS: Taken together, the present findings identify a population of cells in which steroid receptors and nuclear receptor coactivators may interact to modulate steroid sensitivity in brain and regulate hormone-dependent behaviors in female mice. Given that cell culture studies reveal that SRC-1 and SRC-2 can mediate distinct steroid-signaling pathways, the present findings suggest that steroids can produce a variety of complex responses in these specialized brain cells.


Asunto(s)
Encéfalo/metabolismo , Estradiol/metabolismo , Coactivador 1 de Receptor Nuclear/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Encéfalo/efectos de los fármacos , Estradiol/farmacología , Femenino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/metabolismo
18.
Biochim Biophys Acta ; 1800(10): 1084-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20097268

RESUMEN

Studies of the mechanisms by which estrogens influence brain function and behavior have advanced from the explication of individual hormone receptors, neural circuitry and individual gene expression. Now, we can report patterns of estrogen receptor subtype contributions to patterns of behavior. Moreover, new work demonstrates important contributions of nuclear receptor coactivator expression in the central nervous system. In this paper, our current state of knowledge is reviewed.


Asunto(s)
Conducta Animal/fisiología , Conducta/fisiología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Regulación de la Expresión Génica/fisiología , Animales , Femenino , Humanos , Masculino
19.
Psychoneuroendocrinology ; 34 Suppl 1: S9-19, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19541426

RESUMEN

Steroid hormones act in the central and peripheral nervous systems to regulate a variety of functions, including development, cell proliferation, cognition and behavior. Many of these effects of steroid hormones are mediated by their respective receptors, which are members of the nuclear receptor superfamily of transcriptional activators. A variety of cell culture studies reveal that nuclear receptor coactivators are recruited to the steroid receptor complex and are critical in modulating steroid-dependent transcription. Thus, in addition to the availability of the hormone and its receptor, the expression of nuclear receptor coactivators is essential for modulating steroid receptor-mediated transcription. This review will discuss the significance of nuclear receptor coactivators in modulating steroid-dependent gene expression in the central and peripheral nervous systems and the regulation of behavior.


Asunto(s)
Sistema Nervioso Central/fisiología , Neurotransmisores/fisiología , Coactivadores de Receptor Nuclear/fisiología , Sistema Nervioso Periférico/fisiología , Receptores de Esteroides/fisiología , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Sistema Nervioso Central/metabolismo , Regulación de la Expresión Génica , Modelos Biológicos , Neurotransmisores/genética , Neurotransmisores/metabolismo , Sistema Nervioso Periférico/metabolismo , Receptores de Esteroides/genética , Conducta Sexual Animal/fisiología
20.
Front Neuroendocrinol ; 30(3): 328-42, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19401208

RESUMEN

Steroid hormones act in brain and throughout the body to regulate a variety of functions, including development, reproduction, stress and behavior. Many of these effects of steroid hormones are mediated by their respective receptors, which are members of the steroid/nuclear receptor superfamily of transcriptional activators. A variety of studies in cell lines reveal that nuclear receptor coregulators are critical in modulating steroid receptor-dependent transcription. Thus, in addition to the availability of the hormone and the expression of its receptor, nuclear receptor coregulators are essential for efficient steroid-dependent transactivation of genes. This review will highlight the importance of nuclear receptor coregulators in modulating steroid-dependent gene expression in brain and the regulation of behavior.


Asunto(s)
Conducta/fisiología , Encéfalo/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Proteínas Represoras/fisiología , Transactivadores/fisiología , Animales , Encéfalo/fisiología , Humanos , Modelos Biológicos , Receptores Citoplasmáticos y Nucleares/metabolismo , Activación Transcripcional/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...