Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Adv ; 9(8): eabq6718, 2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36812308

RESUMEN

Asprosin, a recently identified adipokine, activates agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus (ARH) via binding to protein tyrosine phosphatase receptor δ (Ptprd) to increase food intake. However, the intracellular mechanisms responsible for asprosin/Ptprd-mediated activation of AgRPARH neurons remain unknown. Here, we demonstrate that the small-conductance calcium-activated potassium (SK) channel is required for the stimulatory effects of asprosin/Ptprd on AgRPARH neurons. Specifically, we found that deficiency or elevation of circulating asprosin increased or decreased the SK current in AgRPARH neurons, respectively. AgRPARH-specific deletion of SK3 (an SK channel subtype highly expressed in AgRPARH neurons) blocked asprosin-induced AgRPARH activation and overeating. Furthermore, pharmacological blockade, genetic knockdown, or knockout of Ptprd abolished asprosin's effects on the SK current and AgRPARH neuronal activity. Therefore, our results demonstrated an essential asprosin-Ptprd-SK3 mechanism in asprosin-induced AgRPARH activation and hyperphagia, which is a potential therapeutic target for the treatment of obesity.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Obesidad , Humanos , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Proteína Relacionada con Agouti/farmacología , Núcleo Arqueado del Hipotálamo/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Obesidad/metabolismo , Adipoquinas/metabolismo , Fibrilina-1/metabolismo
2.
Cell Rep ; 37(10): 110075, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879284

RESUMEN

The neuroendocrine system coordinates metabolic and behavioral adaptations to fasting, including reducing energy expenditure, promoting counterregulation, and suppressing satiation and anxiety to engage refeeding. Here, we show that steroid receptor coactivator-2 (SRC-2) in pro-opiomelanocortin (POMC) neurons is a key regulator of all these responses to fasting. POMC-specific deletion of SRC-2 enhances the basal excitability of POMC neurons; mutant mice fail to efficiently suppress energy expenditure during food deprivation. SRC-2 deficiency blunts electric responses of POMC neurons to glucose fluctuations, causing impaired counterregulation. When food becomes available, these mutant mice show insufficient refeeding associated with enhanced satiation and discoordination of anxiety and food-seeking behavior. SRC-2 coactivates Forkhead box protein O1 (FoxO1) to suppress POMC gene expression. POMC-specific deletion of SRC-2 protects mice from weight gain induced by an obesogenic diet feeding and/or FoxO1 overexpression. Collectively, we identify SRC-2 as a key molecule that coordinates multifaceted adaptive responses to food shortage.


Asunto(s)
Metabolismo Energético , Ayuno/metabolismo , Conducta Alimentaria , Hipotálamo/metabolismo , Neuronas/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , Obesidad/metabolismo , Hipernutrición/metabolismo , Proopiomelanocortina/metabolismo , Animales , Ansiedad/metabolismo , Ansiedad/fisiopatología , Ansiedad/psicología , Modelos Animales de Enfermedad , Ayuno/psicología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Humanos , Hipotálamo/fisiopatología , Masculino , Ratones Noqueados , Coactivador 2 del Receptor Nuclear/genética , Obesidad/genética , Obesidad/fisiopatología , Obesidad/psicología , Hipernutrición/genética , Hipernutrición/fisiopatología , Hipernutrición/psicología , Proopiomelanocortina/genética , Respuesta de Saciedad , Transducción de Señal , Aumento de Peso
3.
J Clin Invest ; 129(9): 3786-3791, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31403469

RESUMEN

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.


Asunto(s)
Hipotálamo/metabolismo , Incretinas/metabolismo , Leptina/metabolismo , Obesidad/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap1/metabolismo , Adiposidad/genética , Animales , Incretinas/genética , Leptina/genética , Ratones , Obesidad/genética , Receptores de la Hormona Gastrointestinal/genética , Receptores de la Hormona Gastrointestinal/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteínas de Unión al GTP rap1/genética
4.
EMBO Rep ; 20(9): e47892, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31318145

RESUMEN

The conversion of skeletal muscle fiber from fast twitch to slow-twitch is important for sustained and tonic contractile events, maintenance of energy homeostasis, and the alleviation of fatigue. Skeletal muscle remodeling is effectively induced by endurance or aerobic exercise, which also generates several tricarboxylic acid (TCA) cycle intermediates, including succinate. However, whether succinate regulates muscle fiber-type transitions remains unclear. Here, we found that dietary succinate supplementation increased endurance exercise ability, myosin heavy chain I expression, aerobic enzyme activity, oxygen consumption, and mitochondrial biogenesis in mouse skeletal muscle. By contrast, succinate decreased lactate dehydrogenase activity, lactate production, and myosin heavy chain IIb expression. Further, by using pharmacological or genetic loss-of-function models generated by phospholipase Cß antagonists, SUNCR1 global knockout, or SUNCR1 gastrocnemius-specific knockdown, we found that the effects of succinate on skeletal muscle fiber-type remodeling are mediated by SUNCR1 and its downstream calcium/NFAT signaling pathway. In summary, our results demonstrate succinate induces transition of skeletal muscle fiber via SUNCR1 signaling pathway. These findings suggest the potential beneficial use of succinate-based compounds in both athletic and sedentary populations.


Asunto(s)
Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Ácido Succínico/farmacología , Animales , Ciclo del Ácido Cítrico/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Fatiga Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Cadenas Pesadas de Miosina/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
Nat Commun ; 10(1): 1718, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30979869

RESUMEN

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.


Asunto(s)
Hipotálamo/metabolismo , Neuronas/metabolismo , Coactivador 1 de Receptor Nuclear/genética , Coactivador 1 de Receptor Nuclear/metabolismo , Obesidad/genética , Alelos , Animales , Peso Corporal , Línea Celular Tumoral , Cruzamientos Genéticos , Eliminación de Gen , Técnicas de Sustitución del Gen , Variación Genética , Células HEK293 , Heterocigoto , Homeostasis , Humanos , Leptina/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Ratones Transgénicos , Mutación Missense , Obesidad/metabolismo , Fenotipo
6.
Nat Commun ; 9(1): 1544, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29670083

RESUMEN

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.


Asunto(s)
Neuronas/metabolismo , Fosfoproteínas/fisiología , Proopiomelanocortina/metabolismo , Caracteres Sexuales , Transactivadores/fisiología , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Peso Corporal , Metabolismo Energético/fisiología , Estrógenos/metabolismo , Femenino , Homeostasis , Hipotálamo/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Receptores de Leptina/metabolismo , Factores Sexuales
7.
Diabetes ; 65(8): 2169-78, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27207529

RESUMEN

Ghrelin signaling has major effects on energy and glucose homeostasis, but it is unknown whether ghrelin's functions are centrally and/or peripherally mediated. The ghrelin receptor, growth hormone secretagogue receptor (GHS-R), is highly expressed in the brain and detectable in some peripheral tissues. To understand the roles of neuronal GHS-R, we generated a mouse line where Ghsr gene is deleted in all neurons using synapsin 1 (Syn1)-Cre driver. Our data showed that neuronal Ghsr deletion abolishes ghrelin-induced spontaneous food intake but has no effect on total energy intake. Remarkably, neuronal Ghsr deletion almost completely prevented diet-induced obesity (DIO) and significantly improved insulin sensitivity. The neuronal Ghsr-deleted mice also showed improved metabolic flexibility, indicative of better adaption to different fuels. In addition, gene expression analysis suggested that hypothalamus and/or midbrain might be the sites that mediate the effects of GHS-R in thermogenesis and physical activity, respectively. Collectively, our results indicate that neuronal GHS-R is a crucial regulator of energy metabolism and a key mediator of DIO. Neuronal Ghsr deletion protects against DIO by regulating energy expenditure, not by energy intake. These novel findings suggest that suppressing central ghrelin signaling may serve as a unique antiobesity strategy.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Neuronas/metabolismo , Obesidad/metabolismo , Obesidad/prevención & control , Receptores de Ghrelina/metabolismo , Animales , Encéfalo/metabolismo , Calorimetría Indirecta , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Femenino , Prueba de Tolerancia a la Glucosa , Hipotálamo/metabolismo , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Mutantes , Obesidad/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Ghrelina/genética , Sinapsinas/genética , Sinapsinas/metabolismo , Termogénesis/genética , Termogénesis/fisiología
8.
Endocrinology ; 154(1): 150-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23211707

RESUMEN

Estrogen receptor-α (ERα) expressed by hypothalamic proopiomelanocortin and steroidogenic factor-1 neurons largely mediates the antiobesity effects of estrogens in females. However, the critical molecular events that are coupled to ERα and mediate estrogenic effects on energy balance remain unknown. In the current study, we demonstrated that steroid receptor coactivator-1 (SRC1), a nuclear receptor coactivator, is abundantly expressed by both proopiomelanocortin and steroidogenic factor-1 neurons. We further showed that central administration of an ERα agonist, propyl pyrazole triol, acutely increases physical interaction between SRC1 and ERα in the hypothalamus. Finally, we demonstrated that the effects of estrogens on energy homeostasis are significantly blunted in female mice lacking SRC1 globally. Collectively our results indicate that SRC1 is functionally required to mediate the antiobesity effects of estrogen-ERα signals.


Asunto(s)
Estrógenos/farmacología , Coactivador 1 de Receptor Nuclear/metabolismo , Factor Esteroidogénico 1/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/metabolismo , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Coactivador 1 de Receptor Nuclear/genética , Fenoles , Proopiomelanocortina/metabolismo , Unión Proteica/efectos de los fármacos , Pirazoles/farmacología , Factor Esteroidogénico 1/genética
9.
Physiol Behav ; 105(2): 276-82, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21824486

RESUMEN

Unfavorable nutritional conditions during early developmental periods may cause neuronal network remodeling in the hypothalamus, which influences subsequent adaptability to those same stressful conditions. Alterations in hypothalamic plasticity as a result of neuronal remodeling are achieved by variations in the repertoire of proteins expressed via gene transcriptional activation or repression, both of which are modulated by histone methylation status. This study demonstrates that fasting had a stimulatory effect on dimethylation and trimethylation of histone 3 at lysine 27 (H3K27) in preoptic/anterior hypothalamus (PO/AH) of 3-day-old chicks. The expression of enhancer of zeste 2 (EZH2), a H3K27-specific histone methyltransferase (HMT), was significantly increased by fasting in the paraventricular nucleus (PVN) and PO/AH, which is consistent with the upregulation of H3K27 dimethylation and trimethylation. Furthermore, in the PVN, corticotrophin-releasing hormone (CRH) mRNA expression was significantly inhibited, while mRNA expressions of thyrotropin-releasing hormone (TRH) and type 2 deiodinase (D2) were significantly stimulated by fasting. These findings highlight the potential role of H3K27 methylation status in early feed stress responses in chicks and may be indicative of an epigenetic mechanism for later adaptation to feed intake stress.


Asunto(s)
Ayuno/fisiología , Histonas/metabolismo , Hipotálamo/metabolismo , Animales , Animales Recién Nacidos , Pollos , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Lisina/metabolismo , Masculino , Metilación , ARN Mensajero , Hormona Liberadora de Tirotropina/genética , Hormona Liberadora de Tirotropina/metabolismo , Yodotironina Deyodinasa Tipo II
10.
Behav Brain Res ; 221(1): 216-26, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21376757

RESUMEN

The effects of ghrelin and obestatin regulation of food intake are different in mammals and chickens. We investigated central effects of ghrelin and obestatin in lines of chickens selected 50 generations for high (HWS) or low (LWS) body weight. We hypothesized that the effect of ghrelin and obestatin on food intake in 5-day-old chicks is mediated by the AMP-activated protein kinase (AMPK) system and selection for body weight alters the brain's response to ghrelin and obestatin by changing the neuronal AMPK system. Although intracerebroventricular (ICV) ghrelin injection decreased food intake in both lines, the threshold for the anorexigenic effect of central ghrelin was lower in LWS than HWS chicks. Obestatin caused a linear dose-dependent increase in food intake in HWS but not LWS chicks. ICV injection of 0.4 nmol ghrelin inhibited hypothalamic AMPK related gene expression and phosphorylation of AMPK α and acetyl-CoA carboxylase (ACC) with the magnitude of inhibition different in the two lines. In contrast, ICV injection of 4 nmol obestatin did not affect mRNA expression of AMPK system or phosphorylation of AMPK and ACC in either line. These data support the premise of a lower threshold for anorexigenic effect of central ghrelin in LWS than HWS chicks, and this difference may be associated with differential hypothalamic AMPK signaling. Additionally, the hypothalamic mRNA level of ghrelin was significantly higher in LWS than HWS, which may have also contributed to the different threshold response to ghrelin in these two lines. The expression of the ghrelin receptor was also higher in the LWS line, but not until 56 days of age. In summary, selection for body weight has resulted in differences in the central ghrelin and obestatin system, and an altered brain AMPK system may contribute to the different neuronal response to ghrelin, but not obestatin.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Peso Corporal/genética , Ingestión de Alimentos/fisiología , Ghrelina/fisiología , Hipotálamo/enzimología , Selección Genética/fisiología , Acetil-CoA Carboxilasa/metabolismo , Factores de Edad , Animales , Regulación del Apetito , Peso Corporal/fisiología , Pollos , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Líquidos/genética , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Ghrelina/administración & dosificación , Ghrelina/farmacología , Hipotálamo/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Fosforilación , Receptores de Ghrelina/biosíntesis , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA