Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Transl Psychiatry ; 14(1): 122, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38413577

RESUMO

Estrogens promote binge alcohol drinking and contribute to sex differences in alcohol use disorder. However, the mechanisms are largely unknown. This study aims to test if estrogens act on 5-hydroxytryptamine neurons in the dorsal raphe nucleus (5-HTDRN) to promote binge drinking. We found that female mice drank more alcohol than male mice in chronic drinking in the dark (DID) tests. This sex difference was associated with distinct alterations in mRNA expression of estrogen receptor α (ERα) and 5-HT-related genes in the DRN, suggesting a potential role of estrogen/ERs/5-HT signaling. In supporting this view, 5-HTDRN neurons from naïve male mice had lower baseline firing activity but higher sensitivity to alcohol-induced excitation compared to 5-HTDRN neurons from naïve female mice. Notably, this higher sensitivity was blunted by 17ß-estradiol treatment in males, indicating an estrogen-dependent mechanism. We further showed that both ERα and ERß are expressed in 5-HTDRN neurons, whereas ERα agonist depolarizes and ERß agonist hyperpolarizes 5-HTDRN neurons. Notably, both treatments blocked the stimulatory effects of alcohol on 5-HTDRN neurons in males, even though they have antagonistic effects on the activity dynamics. These results suggest that ERs' inhibitory effects on ethanol-induced burst firing of 5-HTDRN neurons may contribute to higher levels of binge drinking in females. Consistently, chemogenetic activation of ERα- or ERß-expressing neurons in the DRN reduced binge alcohol drinking. These results support a model in which estrogens act on ERα/ß to prevent alcohol-induced activation of 5-HTDRN neurons, which in return leads to higher binge alcohol drinking.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas , Receptor alfa de Estrogênio , Camundongos , Feminino , Masculino , Animais , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Núcleo Dorsal da Rafe/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/metabolismo , Serotonina/metabolismo , Estrogênios/farmacologia , Etanol/farmacologia
2.
Cell Biosci ; 12(1): 170, 2022 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-36210455

RESUMO

BACKGROUND: Pro-opiomelanocortin (POMC) neurons play a sexually dimorphic role in body weight and glucose balance. However, the mechanisms for the sex differences in POMC neuron functions are not fully understood. RESULTS: We detected small conductance calcium-activated potassium (SK) current in POMC neurons. Secondary analysis of published single-cell RNA-Seq data showed that POMC neurons abundantly express SK3, one SK channel subunit. To test whether SK3 in POMC neurons regulates POMC neuron functions on energy and glucose homeostasis, we used a Cre-loxP strategy to delete SK3 specifically from mature POMC neurons. POMC-specific deletion of SK3 did not affect body weight in either male or female mice. Interestingly, male mutant mice showed not only decreased food intake but also decreased physical activity, resulting in unchanged body weight. Further, POMC-specific SK3 deficiency impaired glucose balance specifically in female mice but not in male mice. Finally, no sex differences were detected in the expression of SK3 and SK current in total POMC neurons. However, we found higher SK current but lower SK3 positive neuron population in male POMC neurons co-expressing estrogen receptor α (ERα) compared to that in females. CONCLUSION: These results revealed a sexually dimorphic role of SK3 in POMC neurons in both energy and glucose homeostasis independent of body weight control, which was associated with the sex difference of SK current in a subpopulation of POMC + ERα + neurons.

3.
Front Endocrinol (Lausanne) ; 13: 898139, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757435

RESUMO

Obesity has become a global epidemic, and it is a major risk factor for other metabolic disorders such as type 2 diabetes and cardiometabolic disease. Accumulating evidence indicates that there is sex-specific metabolic protection and disease susceptibility. For instance, in both clinical and experimental studies, males are more likely to develop obesity, insulin resistance, and diabetes. In line with this, males tend to have more visceral white adipose tissue (WAT) and less brown adipose tissue (BAT) thermogenic activity, both leading to an increased incidence of metabolic disorders. This female-specific fat distribution is partially mediated by sex hormone estrogens. Specifically, hypothalamic estrogen signaling plays a vital role in regulating WAT distribution, WAT beiging, and BAT thermogenesis. These regulatory effects on adipose tissue metabolism are primarily mediated by the activation of estrogen receptor alpha (ERα) in neurons, which interacts with hormones and adipokines such as leptin, ghrelin, and insulin. This review discusses the contribution of adipose tissue dysfunction to obesity and the role of hypothalamic estrogen signaling in preventing metabolic diseases with a particular focus on the VMH, the central regulator of energy expenditure and glucose homeostasis.


Assuntos
Diabetes Mellitus Tipo 2 , Tecido Adiposo Marrom/metabolismo , Diabetes Mellitus Tipo 2/complicações , Estrogênios/metabolismo , Feminino , Homeostase , Humanos , Masculino , Obesidade/metabolismo
4.
Sci Adv ; 8(3): eabk0185, 2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35044814

RESUMO

Estrogen receptor­α (ERα) expressed by neurons in the ventrolateral subdivision of the ventromedial hypothalamic nucleus (ERαvlVMH) regulates body weight in females, but the downstream neural circuits mediating this biology remain largely unknown. Here we identified a neural circuit mediating the metabolic effects of ERαvlVMH neurons. We found that selective activation of ERαvlVMH neurons stimulated brown adipose tissue (BAT) thermogenesis, physical activity, and core temperature and that ERαvlVMH neurons provide monosynaptic glutamatergic inputs to 5-hydroxytryptamine (5-HT) neurons in the dorsal raphe nucleus (DRN). Notably, the ERαvlVMH → DRN circuit responds to changes in ambient temperature and nutritional states. We further showed that 5-HTDRN neurons mediate the stimulatory effects of ERαvlVMH neurons on BAT thermogenesis and physical activity and that ERα expressed by DRN-projecting ERαvlVMH neurons is required for the maintenance of energy balance. Together, these findings support a model that ERαvlVMH neurons activate BAT thermogenesis and physical activity through stimulating 5-HTDRN neurons.

5.
Dev Cell ; 56(7): 985-999.e4, 2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33711247

RESUMO

Thermogenic beige fat found in white adipose tissue is a potential therapeutic target to curb the global obesity and diabetes epidemic. However, these inducible thermogenic beige adipocytes have been thought to be short-lived and to rapidly convert to "white-like" adipocytes after discontinuing stimuli. In this study, using effective labeling techniques and genetic mouse tools, we demonstrate that a subset of UCP1+ cells that exist within white adipose tissue are able to self-divide and contribute to new beige adipocyte recruitment in response to ß3 stimuli. When these cells are depleted or their adipogenic capability is blocked, ß3-induced beige adipocyte formation is impaired. We also identify a cell-cycle machinery of p21 and CDKN2A as a molecular basis of beige adipocyte regulation. Collectively, our findings provide new insights into the cellular and molecular mechanisms of beige adipocyte regulation and potential therapeutic opportunities to induce the beige phenotype and treat metabolic disease.


Assuntos
Adipócitos Bege/fisiologia , Tecido Adiposo Branco/citologia , Células-Tronco/fisiologia , Proteína Desacopladora 1/análise , Agonistas de Receptores Adrenérgicos beta 3/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Deleção de Genes , Genes cdc , Masculino , Camundongos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
6.
Mol Metab ; 42: 101053, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32712433

RESUMO

OBJECTIVE: Estrogen protects animals from obesity through estrogen receptor α (ERα), partially by inhibiting overeating in animals fed ad libitum. However, the effects of estrogen on feeding behavior in hungry animals remain unclear. In this study, we examined the roles of 17ß-estradiol (E2) and ERα in the regulation of feeding in hungry female animals and explored the underlying mechanisms. METHODS: Wild-type female mice with surgical depletion of endogenous estrogens were used to examine the effects of E2 supplementation on acute refeeding behavior after starvation. ERα-C451A mutant mice deficient in membrane-bound ERα activity and ERα-AF20 mutant mice lacking ERα transcriptional activity were used to further examine mechanisms underlying acute feeding triggered by either fasting or central glucopenia (induced by intracerebroventricular injections of 2-deoxy-D-glucose). We also used electrophysiology to explore the impact of these ERα mutations on the neural activities of ERα neurons in the hypothalamus. RESULTS: In the wild-type female mice, ovariectomy reduced fasting-induced refeeding, which was restored by E2 supplementation. The ERα-C451A mutation, but not the ERα-AF20 mutation, attenuated acute feeding induced by either fasting or central glucopenia. The ERα-C451A mutation consistently impaired the neural responses of hypothalamic ERα neurons to hypoglycemia. CONCLUSION: In addition to previous evidence that estrogen reduces deviations in energy balance by inhibiting eating at a satiated state, our findings demonstrate the unexpected role of E2 that promotes eating in hungry mice, also contributing to the stability of energy homeostasis. This latter effect specifically requires membrane-bound ERα activity.


Assuntos
Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Animais , Estradiol/fisiologia , Receptor alfa de Estrogênio/genética , Comportamento Alimentar/fisiologia , Feminino , Homeostase/efeitos dos fármacos , Fome/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade , Ovariectomia , Transdução de Sinais
7.
J Neurosci ; 40(27): 5196-5207, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32482639

RESUMO

Elevations in estrogen (17ß-estradiol, E2) are associated with increased alcohol drinking by women and experimentally in rodents. E2 alters the activity of the dopamine system, including the VTA and its projection targets, which plays an important role in binge drinking. A previous study demonstrated that, during high E2 states, VTA neurons in female mice are more sensitive to ethanol excitation. However, the mechanisms responsible for the ability of E2 to enhance ethanol sensitivity of VTA neurons have not been investigated. In this study, we used selective agonists and antagonists to examine the role of ER subtypes (ERα and ERß) in regulating the ethanol sensitivity of VTA neurons in female mice and found that ERα promotes the enhanced ethanol response of VTA neurons. We also demonstrated that enhancement of ethanol excitation requires the activity of the metabotropic glutamate receptor, mGluR1, which is known to couple with ERα at the plasma membrane. To investigate the behavioral relevance of these findings, we administered lentivirus-expressing short hairpin RNAs targeting either ERα or ERß into the VTA and found that knockdown of each receptor in the VTA reduced binge-like ethanol drinking in female, but not male, mice. Reducing ERα in the VTA had a more dramatic effect on binge-like drinking than reducing ERß, consistent with the ability of ERα to alter ethanol sensitivity of VTA neurons. These results provide important insight into sex-specific mechanisms that drive excessive alcohol drinking.SIGNIFICANCE STATEMENT Estrogen has potent effects on the dopamine system and increases the vulnerability of females to develop addiction to substances, such as alcohol. We investigated the mechanisms by which estrogen increases the response of neurons in the VTA to ethanol. We found that activation of the ERα increased the ethanol-induced excitation of VTA neurons. 17ß-Estradiol-mediated enhancement of ethanol-induced excitation required the metabotropic glutamate receptor mGluR1. We also demonstrated that ERs in the VTA regulate binge-like alcohol drinking by female, but not male, mice. The influence of ERs on binge drinking in female mice suggests that treatments for alcohol use disorder in women may need to account for this sex difference.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Receptor alfa de Estrogênio/metabolismo , Etanol/farmacologia , Neurônios/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Consumo Excessivo de Bebidas Alcoólicas/psicologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Receptores de AMPA/metabolismo , Área Tegmentar Ventral/citologia
8.
Nat Commun ; 11(1): 2165, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32358493

RESUMO

Brain glucose-sensing neurons detect glucose fluctuations and prevent severe hypoglycemia, but mechanisms mediating functions of these glucose-sensing neurons are unclear. Here we report that estrogen receptor-α (ERα)-expressing neurons in the ventrolateral subdivision of the ventromedial hypothalamic nucleus (vlVMH) can sense glucose fluctuations, being glucose-inhibited neurons (GI-ERαvlVMH) or glucose-excited neurons (GE-ERαvlVMH). Hypoglycemia activates GI-ERαvlVMH neurons via the anoctamin 4 channel, and inhibits GE-ERαvlVMH neurons through opening the ATP-sensitive potassium channel. Further, we show that GI-ERαvlVMH neurons preferentially project to the medioposterior arcuate nucleus of the hypothalamus (mpARH) and GE-ERαvlVMH neurons preferentially project to the dorsal Raphe nuclei (DRN). Activation of ERαvlVMH to mpARH circuit and inhibition of ERαvlVMH to DRN circuit both increase blood glucose. Thus, our results indicate that ERαvlVMH neurons detect glucose fluctuations and prevent severe hypoglycemia in mice.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Glucose/metabolismo , Neurônios/metabolismo , Animais , Eletrofisiologia , Endocrinologia , Feminino , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase em Tempo Real
9.
J Clin Invest ; 129(9): 3786-3791, 2019 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31403469

RESUMO

Nutrient excess, a major driver of obesity, diminishes hypothalamic responses to exogenously administered leptin, a critical hormone of energy balance. Here, we aimed to identify a physiological signal that arises from excess caloric intake and negatively controls hypothalamic leptin action. We found that deficiency of the gastric inhibitory polypeptide receptor (Gipr) for the gut-derived incretin hormone GIP protected against diet-induced neural leptin resistance. Furthermore, a centrally administered antibody that neutralizes GIPR had remarkable antiobesity effects in diet-induced obese mice, including reduced body weight and adiposity, and a decreased hypothalamic level of SOCS3, an inhibitor of leptin actions. In contrast, centrally administered GIP diminished hypothalamic sensitivity to leptin and increased hypothalamic levels of Socs3. Finally, we show that GIP increased the active form of the small GTPase Rap1 in the brain and that its activation was required for the central actions of GIP. Altogether, our results identify GIPR/Rap1 signaling in the brain as a molecular pathway linking overnutrition to the control of neural leptin actions.


Assuntos
Hipotálamo/metabolismo , Incretinas/metabolismo , Leptina/metabolismo , Obesidade/metabolismo , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/metabolismo , Adiposidade/genética , Animais , Incretinas/genética , Leptina/genética , Camundongos , Obesidade/genética , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Proteínas rap1 de Ligação ao GTP/genética
10.
Nat Commun ; 10(1): 1718, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979869

RESUMO

Hypothalamic neurons expressing the anorectic peptide Pro-opiomelanocortin (Pomc) regulate food intake and body weight. Here, we show that Steroid Receptor Coactivator-1 (SRC-1) interacts with a target of leptin receptor activation, phosphorylated STAT3, to potentiate Pomc transcription. Deletion of SRC-1 in Pomc neurons in mice attenuates their depolarization by leptin, decreases Pomc expression and increases food intake leading to high-fat diet-induced obesity. In humans, fifteen rare heterozygous variants in SRC-1 found in severely obese individuals impair leptin-mediated Pomc reporter activity in cells, whilst four variants found in non-obese controls do not. In a knock-in mouse model of a loss of function human variant (SRC-1L1376P), leptin-induced depolarization of Pomc neurons and Pomc expression are significantly reduced, and food intake and body weight are increased. In summary, we demonstrate that SRC-1 modulates the function of hypothalamic Pomc neurons, and suggest that targeting SRC-1 may represent a useful therapeutic strategy for weight loss.


Assuntos
Hipotálamo/metabolismo , Neurônios/metabolismo , Coativador 1 de Receptor Nuclear/genética , Coativador 1 de Receptor Nuclear/metabolismo , Obesidade/genética , Alelos , Animais , Peso Corporal , Linhagem Celular Tumoral , Cruzamentos Genéticos , Deleção de Genes , Técnicas de Introdução de Genes , Variação Genética , Células HEK293 , Heterozigoto , Homeostase , Humanos , Leptina/metabolismo , Masculino , Potenciais da Membrana , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Obesidade/metabolismo , Fenótipo
11.
Nat Commun ; 9(1): 1544, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29670083

RESUMO

Sexual dimorphism exists in energy balance, but the underlying mechanisms remain unclear. Here we show that the female mice have more pro-opiomelanocortin (POMC) neurons in the arcuate nucleus of hypothalamus than males, and female POMC neurons display higher neural activities, compared to male counterparts. Strikingly, deletion of the transcription factor, TAp63, in POMC neurons confers "male-like" diet-induced obesity (DIO) in female mice associated with decreased POMC neural activities; but the same deletion does not affect male mice. Our results indicate that TAp63 in female POMC neurons contributes to the enhanced POMC neuron functions and resistance to obesity in females. Thus, TAp63 in POMC neurons is one key molecular driver for the sexual dimorphism in energy homeostasis.


Assuntos
Neurônios/metabolismo , Fosfoproteínas/fisiologia , Pró-Opiomelanocortina/metabolismo , Caracteres Sexuais , Transativadores/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Metabolismo Energético/fisiologia , Estrogênios/metabolismo , Feminino , Homeostase , Hipotálamo/metabolismo , Masculino , Camundongos , Obesidade/metabolismo , Receptores para Leptina/metabolismo , Fatores Sexuais
12.
Cell Rep ; 20(10): 2455-2467, 2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28877477

RESUMO

Although the widely used anticoagulant drug heparin has been shown to have many other biological functions independent of its anticoagulant role, its effects on energy homeostasis are unknown. Here, we demonstrate that heparin level is negatively associated with nutritional states and that heparin treatment increases food intake and body weight gain. By using electrophysiological, pharmacological, molecular biological, and chemogenetic approaches, we provide evidence that heparin increases food intake by stimulating AgRP neurons and increasing AgRP release. Our results support a model whereby heparin competes with insulin for insulin receptor binding on AgRP neurons, and by doing so it inhibits FoxO1 activity to promote AgRP release and feeding. Heparin may be a potential drug target for food intake regulation and body weight control.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Heparina/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptor de Insulina/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Eletrofisiologia , Proteína Forkhead Box O1/metabolismo , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
Metabolism ; 70: 152-159, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28403939

RESUMO

BACKGROUND: Brain estrogen receptor-α (ERα) is essential for estrogenic regulation of energy homeostasis and reproduction. We previously showed that ERα expressed by pro-opiomelanocortin (POMC) neurons mediates estrogen's effects on food intake, body weight, negative regulation of hypothalamic-pituitary-gonadal axis (HPG axis) and fertility. RESULTS AND CONCLUSIONS: We report here that global deletion of a key downstream receptor for POMC peptide, the melanocortin 4 receptor (MC4R), did not affect normal negative feedback regulation of estrogen on the HPG axis, estrous cyclicity and female fertility. Furthermore, loss of the MC4R did not influence estrogenic regulation on food intake and body weight. These results indicate that the MC4R is not required for estrogen's effects on metabolic and reproductive functions.


Assuntos
Estrogênios , Homeostase , Receptor Tipo 4 de Melanocortina/fisiologia , Reprodução , Animais , Peso Corporal , Ingestão de Alimentos , Metabolismo Energético , Retroalimentação Fisiológica , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Pró-Opiomelanocortina
14.
Cell Rep ; 17(7): 1807-1818, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27829152

RESUMO

Neurons that co-express agouti-related peptide (AgRP) and neuropeptide Y (NPY) are indispensable for normal feeding behavior. Firing activities of AgRP/NPY neurons are dynamically regulated by energy status and coordinate appropriate feeding behavior to meet nutritional demands. However, intrinsic mechanisms that regulate AgRP/NPY neural activities during the fed-to-fasted transition are not fully understood. We found that AgRP/NPY neurons in satiated mice express high levels of the small-conductance calcium-activated potassium channel 3 (SK3) and are inhibited by SK3-mediated potassium currents; on the other hand, food deprivation suppresses SK3 expression in AgRP/NPY neurons, and the decreased SK3-mediated currents contribute to fasting-induced activation of these neurons. Genetic mutation of SK3 specifically in AgRP/NPY neurons leads to increased sensitivity to diet-induced obesity, associated with chronic hyperphagia and decreased energy expenditure. Our results identify SK3 as a key intrinsic mediator that coordinates nutritional status with AgRP/NPY neural activities and animals' feeding behavior and energy metabolism.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Metabolismo Energético , Comportamento Alimentar , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Ritmo Circadiano/efeitos dos fármacos , Dieta Hiperlipídica , Metabolismo Energético/efeitos dos fármacos , Jejum/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Hiperfagia/complicações , Hiperfagia/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mutação/genética , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Obesidade/complicações , Obesidade/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética
15.
Hypertension ; 67(6): 1321-30, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27091896

RESUMO

Psychological stress contributes to the development of hypertension in humans. The ovarian hormone, estrogen, has been shown to prevent stress-induced pressor responses in females by unknown mechanisms. Here, we showed that the antihypertensive effects of estrogen during stress were blunted in female mice lacking estrogen receptor-α in the brain medial amygdala. Deletion of estrogen receptor-α in medial amygdala neurons also resulted in increased excitability of these neurons, associated with elevated ionotropic glutamate receptor expression. We further demonstrated that selective activation of medial amygdala neurons mimicked effects of stress to increase blood pressure in mice. Together, our results support a model where estrogen acts on estrogen receptor-α expressed by medial amygdala neurons to prevent stress-induced activation of these neurons, and therefore prevents pressor responses to stress.


Assuntos
Tonsila do Cerebelo/metabolismo , Estradiol/farmacologia , Hipertensão/fisiopatologia , Receptores de Estrogênio/metabolismo , Estresse Fisiológico , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Determinação da Pressão Arterial , Western Blotting , Corticosterona/sangue , Modelos Animais de Doenças , Feminino , Hipertensão/metabolismo , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/métodos , Valores de Referência
16.
Sci Rep ; 6: 23459, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26988598

RESUMO

Estrogens act in the ventromedial hypothalamic nucleus (VMH) to regulate body weight homeostasis. However, the molecular mechanisms underlying these estrogenic effects are unknown. We show that activation of estrogen receptor-α (ERα) stimulates neural firing of VMH neurons expressing ERα, and these effects are blocked with intracellular application of a pharmacological inhibitor of the phosphatidyl inositol 3-kinase (PI3K). Further, we demonstrated that mice with genetic inhibition of PI3K activity in VMH neurons showed a sexual dimorphic obese phenotype, with only female mutants being affected. In addition, inhibition of VMH PI3K activity blocked effects of 17ß-estradiol to stimulate energy expenditure, but did not affect estrogen-induced anorexia. Collectively, our results indicate that PI3K activity in VMH neurons plays a physiologically relevant role in mediating estrogenic actions on energy expenditure in females.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Núcleo Hipotalâmico Ventromedial/metabolismo , Animais , Peso Corporal , Modelos Animais de Doenças , Metabolismo Energético , Estrogênios/farmacologia , Feminino , Masculino , Camundongos , Obesidade/induzido quimicamente , Caracteres Sexuais
17.
Metabolism ; 65(4): 522-32, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26975544

RESUMO

BACKGROUND: A variety of biological functions of estrogens, including regulation of energy metabolism, are mediated by neurons expressing estrogen receptor-α (ERα) in the brain. However, complex intracellular processes in these ERα-expressing neurons are difficult to unravel, due to the lack of strategy to visualize ERα-expressing neurons, especially in unfixed brain tissues. RESULTS AND CONCLUSIONS: Here we generated a novel ERα-ZsGreen reporter mouse line in which expression of a green fluorescent reporter protein, ZsGreen, is driven by a 241kb ERα gene promoter. We validated that ZsGreen is highly colocalized with endogenous ERα in the brain. Native ZsGreen signals were visualized in unfixed brain tissue, and were used to assist single cell collection and electrophysiological recordings. Finally, we demonstrated that this ERα-ZsGreen mouse allele can be used in combination with other genetic reporter alleles to allow experiments in highly selective neural populations.


Assuntos
Neurônios/metabolismo , Neurônios/ultraestrutura , Receptores de Estrogênio/biossíntese , Alelos , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Química Encefálica/genética , Linhagem Celular , Separação Celular/métodos , Fenômenos Eletrofisiológicos , Genes Reporter , Proteínas de Fluorescência Verde , Camundongos , Camundongos Endogâmicos C57BL , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/biossíntese , Receptores de Estrogênio/genética , Receptor ERRalfa Relacionado ao Estrogênio
18.
Endocrinology ; 156(12): 4474-91, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26375425

RESUMO

Estrogens act upon estrogen receptor (ER)α to inhibit feeding and improve glucose homeostasis in female animals. However, the intracellular signals that mediate these estrogenic actions remain unknown. Here, we report that anorexigenic effects of estrogens are blunted in female mice that lack ERα specifically in proopiomelanocortin (POMC) progenitor neurons. These mutant mice also develop insulin resistance and are insensitive to the glucose-regulatory effects of estrogens. Moreover, we showed that propyl pyrazole triol (an ERα agonist) stimulates the phosphatidyl inositol 3-kinase (PI3K) pathway specifically in POMC progenitor neurons, and that blockade of PI3K attenuates propyl pyrazole triol-induced activation of POMC neurons. Finally, we show that effects of estrogens to inhibit food intake and to improve insulin sensitivity are significantly attenuated in female mice with PI3K genetically inhibited in POMC progenitor neurons. Together, our results indicate that an ERα-PI3K cascade in POMC progenitor neurons mediates estrogenic actions to suppress food intake and improve insulin sensitivity.


Assuntos
Glicemia/metabolismo , Ingestão de Alimentos/genética , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Resistência à Insulina , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinase/genética , Pró-Opiomelanocortina/metabolismo , Animais , Ingestão de Alimentos/efeitos dos fármacos , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Comportamento Alimentar/fisiologia , Feminino , Glucose/metabolismo , Homeostase , Camundongos , Camundongos Knockout , Células-Tronco Neurais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fenóis/farmacologia , Fosfatidilinositol 3-Quinase/efeitos dos fármacos , Pirazóis/farmacologia , Transdução de Sinais
20.
J Clin Invest ; 125(7): 2861-76, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26098212

RESUMO

Estrogen receptor-α (ERα) activity in the brain prevents obesity in both males and females. However, the ERα-expressing neural populations that regulate body weight remain to be fully elucidated. Here we showed that single-minded-1 (SIM1) neurons in the medial amygdala (MeA) express abundant levels of ERα. Specific deletion of the gene encoding ERα (Esr1) from SIM1 neurons, which are mostly within the MeA, caused hypoactivity and obesity in both male and female mice fed with regular chow, increased susceptibility to diet-induced obesity (DIO) in males but not in females, and blunted the body weight-lowering effects of a glucagon-like peptide-1-estrogen (GLP-1-estrogen) conjugate. Furthermore, selective adeno-associated virus-mediated deletion of Esr1 in the MeA of adult male mice produced a rapid body weight gain that was associated with remarkable reductions in physical activity but did not alter food intake. Conversely, overexpression of ERα in the MeA markedly reduced the severity of DIO in male mice. Finally, an ERα agonist depolarized MeA SIM1 neurons and increased their firing rate, and designer receptors exclusively activated by designer drug-mediated (DREADD-mediated) activation of these neurons increased physical activity in mice. Collectively, our results support a model where ERα signals activate MeA neurons to stimulate physical activity, which in turn prevents body weight gain.


Assuntos
Peso Corporal/fisiologia , Complexo Nuclear Corticomedial/metabolismo , Receptor alfa de Estrogênio/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Complexo Nuclear Corticomedial/citologia , Complexo Nuclear Corticomedial/efeitos dos fármacos , Metabolismo Energético , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/genética , Estrogênios/administração & dosagem , Feminino , Peptídeo 1 Semelhante ao Glucagon/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/fisiologia , Neurônios/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Caracteres Sexuais , Transdução de Sinais , Aumento de Peso/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA