Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nature ; 494(7435): 111-5, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23389544

RESUMO

Insulin resistance represents a hallmark during the development of type 2 diabetes mellitus and in the pathogenesis of obesity-associated disturbances of glucose and lipid metabolism. MicroRNA (miRNA)-dependent post-transcriptional gene silencing has been recognized recently to control gene expression in disease development and progression, including that of insulin-resistant type 2 diabetes. The deregulation of miRNAs miR-143 (ref. 4), miR-181 (ref. 5), and miR-103 and miR-107 (ref. 6) alters hepatic insulin sensitivity. Here we report that the expression of miR-802 is increased in the liver of two obese mouse models and obese human subjects. Inducible transgenic overexpression of miR-802 in mice causes impaired glucose tolerance and attenuates insulin sensitivity, whereas reduction of miR-802 expression improves glucose tolerance and insulin action. We identify Hnf1b (also known as Tcf2) as a target of miR-802-dependent silencing, and show that short hairpin RNA (shRNA)-mediated reduction of Hnf1b in liver causes glucose intolerance, impairs insulin signalling and promotes hepatic gluconeogenesis. In turn, hepatic overexpression of Hnf1b improves insulin sensitivity in Lepr(db/db) mice. Thus, this study defines a critical role for deregulated expression of miR-802 in the development of obesity-associated impairment of glucose metabolism through targeting of Hnf1b, and assigns Hnf1b an unexpected role in the control of hepatic insulin sensitivity.


Assuntos
Inativação Gênica , Glucose/metabolismo , Fator 1-beta Nuclear de Hepatócito/deficiência , MicroRNAs/genética , Obesidade/genética , Animais , Regulação da Expressão Gênica , Gluconeogênese , Glucose/biossíntese , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Fator 1-beta Nuclear de Hepatócito/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Humanos , Insulina/metabolismo , Resistência à Insulina/genética , Fígado/metabolismo , Camundongos , MicroRNAs/biossíntese , Transdução de Sinais
2.
Cell Metab ; 16(2): 144-52, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22883229

RESUMO

Obesity represents a major risk factor for the development of insulin and leptin resistance, ultimately leading to a pleiotropic spectrum of metabolic alterations. However, resistance to both hormones does not uniformly affect all target cells and intracellular signaling pathways. In contrast, numerous clinical phenotypes arise from selective hormone resistance, leading to inhibition of defined intracellular signaling pathways in some tissues, while in other cell types hormone action is maintained or even overactivated. Here, we review the molecular mechanisms and clinical outcomes resulting from selective insulin and leptin resistance, which should ultimately guide future strategies for the treatment of obesity-associated diseases.


Assuntos
Sistema Nervoso Central/fisiopatologia , Resistência à Insulina/fisiologia , Leptina/fisiologia , Doenças Metabólicas/fisiopatologia , Obesidade/metabolismo , Transdução de Sinais/fisiologia , Humanos , Leptina/metabolismo , Doenças Metabólicas/etiologia , Obesidade/complicações
3.
Mol Metab ; 1(1-2): 5-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-24024112
4.
Cell Metab ; 13(6): 720-8, 2011 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-21641553

RESUMO

Dopaminergic midbrain neurons integrate signals on food palatability and food-associated reward into the complex control of energy homeostasis. To define the role of insulin receptor (IR) signaling in this circuitry, we inactivated IR signaling in tyrosine hydroxylase (Th)-expressing cells of mice (IR(ΔTh)). IR inactivation in Th-expressing cells of mice resulted in increased body weight, increased fat mass, and hyperphagia. While insulin acutely stimulated firing frequency in 50% of dopaminergic VTA/SN neurons, this response was abolished in IR(ΔTh) mice. Moreover, these mice exhibited an altered response to cocaine under food-restricted conditions. Taken together, these data provide in vivo evidence for a critical role of insulin signaling in catecholaminergic neurons to control food intake and energy homeostasis.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Insulina/farmacologia , Neurônios/metabolismo , Potenciais de Ação , Adiposidade , Animais , Calorimetria Indireta , Catecolaminas/metabolismo , Cocaína/farmacologia , Ingestão de Alimentos/genética , Expressão Gênica , Hiperinsulinismo/genética , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Neurônios/efeitos dos fármacos , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Transdução de Sinais , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
5.
Trends Endocrinol Metab ; 22(1): 16-23, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20888253

RESUMO

Obesity has been characterized as a state of chronic inflammation. Inflammatory signaling not only causes peripheral insulin resistance, but also promotes neuronal insulin and leptin resistance, which further propagates a positive energy balance. Upon development of obesity, numerous conditions, including increased circulating cytokine concentrations and cell autonomous dysregulation of homeostatic signaling pathways, such as the endoplasmic reticulum stress response, promote activation of stress kinases, to cause peripheral insulin as well as central insulin and leptin resistance. Recently, activation of toll-like receptor (TLR) signaling has been recognized as an alternative activator of obesity-induced inflammation. In this paper, we review recent progress in defining the molecular basis of obesity-associated TLR activation and its role in the development of metabolic syndrome.


Assuntos
Inflamação/imunologia , Receptores Toll-Like/metabolismo , Animais , Humanos , Inflamação/genética , Obesidade/genética , Obesidade/imunologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Receptores Toll-Like/genética
6.
Cell Metab ; 12(3): 237-49, 2010 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-20816090

RESUMO

The contribution of interleukin (IL)-6 signaling in obesity-induced inflammation remains controversial. To specifically define the role of hepatic IL-6 signaling in insulin action and resistance, we have generated mice with hepatocyte-specific IL-6 receptor (IL-6R) alpha deficiency (IL-6Ralpha(L-KO) mice). These animals showed no alterations in body weight and fat content but exhibited a reduction in insulin sensitivity and glucose tolerance. Impaired glucose metabolism originated from attenuated insulin-stimulated glucose transport in skeletal muscle and fat. Surprisingly, hepatic IL-6Ralpha-disruption caused an exaggerated inflammatory response during euglycemic hyperinsulinemic clamp analysis, as revealed by increased expression of IL-6, TNF-alpha, and IL-10, as well as enhanced activation of inflammatory signaling such as phosphorylation of IkappaBalpha. Neutralization of TNF-alpha or ablation of Kupffer cells restored glucose tolerance in IL-6Ralpha(L-KO) mice. Thus, our results reveal an unexpected role for hepatic IL-6 signaling to limit hepatic inflammation and to protect from local and systemic insulin resistance.


Assuntos
Inflamação/metabolismo , Insulina/metabolismo , Interleucina-6/metabolismo , Fígado/citologia , Fígado/patologia , Transdução de Sinais/fisiologia , Adiposidade , Animais , Metabolismo Energético , Glucose/metabolismo , Glicogênio/biossíntese , Homeostase , Humanos , Resistência à Insulina/fisiologia , Interleucina-10/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Cell Mol Life Sci ; 67(19): 3255-73, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20549539

RESUMO

The central nervous system (CNS) is capable of gathering information on the body's nutritional state and it implements appropriate behavioral and metabolic responses to changes in fuel availability. This feedback signaling of peripheral tissues ensures the maintenance of energy homeostasis. The hypothalamus is a primary site of convergence and integration for these nutrient-related feedback signals, which include central and peripheral neuronal inputs as well as hormonal signals. Increasing evidence indicates that glucose and lipids are detected by specialized fuel-sensing neurons that are integrated in these hypothalamic neuronal circuits. The purpose of this review is to outline the current understanding of fuel-sensing mechanisms in the hypothalamus, to integrate the recent findings in this field, and to address the potential role of dysregulation in these pathways in the development of obesity and type 2 diabetes mellitus.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Lipídeos , Neurônios/metabolismo , Estado Nutricional , Obesidade/metabolismo , Transdução de Sinais/fisiologia
8.
Front Neuroendocrinol ; 31(1): 4-15, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19729032

RESUMO

Body weight is tightly controlled in a species-specific range from insects to vertebrates and organisms have developed a complex regulatory network in order to avoid either excessive weight gain or chronic weight loss. Energy homeostasis, a term comprising all processes that aim to maintain stability of the metabolic state, requires a constant communication of the different organs involved; i.e. adipose tissue, skeletal muscle, liver, pancreas and the central nervous system (CNS). A tight hormonal network ensures rapid communication to control initiation and cessation of eating, nutrient processing and partitioning of the available energy within different organs and metabolic pathways. Moreover, recent experiments indicate that many of these homeostatic signals modulate the neural circuitry of food reward and motivation. Disturbances in each individual system can affect the maintenance and regulation of the others, making the analysis of energy homeostasis and its dysregulation highly complex. Though this cross-talk has been intensively studied for many years now, we are far from a complete understanding of how energy balance is maintained and multiple key questions remain unanswered. This review summarizes some of the latest developments in the field and focuses on the effects of leptin, insulin, and nutrient-related signals in the central regulation of feeding behavior. The integrated view, how these signals interact and the definition of functional neurocircuits in control of energy homeostasis, will ultimately help to develop new therapeutic interventions within the current obesity epidemic.


Assuntos
Metabolismo Energético/fisiologia , Homeostase/fisiologia , Sistemas Neurossecretores/fisiologia , Animais , Peso Corporal/fisiologia , Encéfalo/fisiologia , Diabetes Mellitus , Ingestão de Alimentos/fisiologia , Glucose/metabolismo , Humanos , Hipotálamo/fisiologia , Insulina/fisiologia , Leptina/fisiologia , Fenômenos Fisiológicos da Nutrição , Obesidade , Transdução de Sinais
9.
Curr Opin Pharmacol ; 9(6): 794-804, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19884043

RESUMO

The exceeding efforts in understanding the signals initiated by nutrients and hormones in the central nervous system (CNS) to regulate glucose and energy homeostasis have largely revolutionized our understanding of the neurocircuitry in control of peripheral metabolism. The ability of neurons to sense nutrients and hormones and to adopt a coordinated response to these signals is of crucial importance in controlling food intake, energy expenditure, glucose and lipid metabolism. Anatomical lesion experiments, pharmacological inhibition of signaling pathways, and, more recently, the analysis of conditional mouse mutants with modifications of hormone and nutrient signaling in defined neuronal populations have broadened our understanding of these complex neurocircuits. This review summarizes recent findings regarding the role of the CNS in sensing and transmitting nutritional and hormonal signals to control energy and glucose homeostasis and aims to define them as potential novel drug targets for the treatment of obesity and type 2 diabetes mellitus.


Assuntos
Sistema Nervoso Central/fisiologia , Metabolismo Energético/fisiologia , Glucose/metabolismo , Homeostase/fisiologia , Vias Neurais/fisiologia , Animais , Peso Corporal/fisiologia , Sistema Nervoso Central/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Humanos , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Modelos Neurológicos , Vias Neurais/metabolismo , Neurotransmissores/fisiologia , Obesidade/complicações , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Transdução de Sinais/fisiologia
10.
Cell Metab ; 10(4): 249-59, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19808018

RESUMO

Obesity-associated activation of inflammatory pathways represents a key step in the development of insulin resistance in peripheral organs, partially via activation of TLR4 signaling by fatty acids. Here, we demonstrate that palmitate acting in the central nervous system (CNS) inhibits leptin-induced anorexia and Stat3 activation. To determine the functional significance of TLR signaling in the CNS in the development of leptin resistance and diet-induced obesity in vivo, we have characterized mice deficient for the TLR adaptor molecule MyD88 in the CNS (MyD88(DeltaCNS)). Compared to control mice, MyD88(DeltaCNS) mice are protected from high-fat diet (HFD)-induced weight gain, from the development of HFD-induced leptin resistance, and from the induction of leptin resistance by acute central application of palmitate. Moreover, CNS-restricted MyD88 deletion protects from HFD- and icv palmitate-induced impairment of peripheral glucose metabolism. Thus, we define neuronal MyD88-dependent signaling as a key regulator of diet-induced leptin and insulin resistance in vivo.


Assuntos
Sistema Nervoso Central/metabolismo , Dieta , Leptina/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Obesidade/metabolismo , Ácido Palmítico/metabolismo , Transdução de Sinais/fisiologia , Animais , Gorduras na Dieta/metabolismo , Ingestão de Alimentos , Metabolismo Energético , Ativação Enzimática , Feminino , Glucose/metabolismo , Homeostase , Humanos , Quinase I-kappa B/metabolismo , Insulina/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/fisiologia , Fator 88 de Diferenciação Mieloide/genética , Aumento de Peso
11.
Physiol Behav ; 97(5): 632-8, 2009 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-19351541

RESUMO

As the obesity epidemic, diabetes mellitus type 2, and associated comorbidities show no signs of abating, large efforts have been put into a better understanding of the homeostatic control mechanisms involved in regulation of body weight and energy homeostasis. For decades, the hypothalamic arcuate nucleus (ARC), which integrates peripheral signals and modulates appetite and metabolism, has been the focus of investigation. Besides these basic homeostatic circuits, food palatability and reward are thought to be major factors involved in the regulation of food intake. Highly palatable food is easily available, and is ingested even when there is no metabolic need for it. Thus, overriding of the homeostatic control systems by the cognitive, rewarding, social, and emotional aspects of palatable food may contribute to the obesity epidemic. This review aims to provide an updated view, how insulin and leptin as signals originating from the periphery of the body and communicating energy availability to the CNS act not only on ARC neurons, but also directly control the activity of neuronal circuits in control of food-associated reward mechanisms.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético/fisiologia , Homeostase/fisiologia , Insulina/metabolismo , Leptina/metabolismo , Animais , Humanos
12.
J Clin Invest ; 118(6): 2132-47, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18451994

RESUMO

Insulin resistance is a hallmark of type 2 diabetes, and many insights into the functions of insulin have been gained through the study of mice lacking the IR. To gain a better understanding of the role of insulin action in the brain versus peripheral tissues, we created 2 mouse models with inducible IR inactivation, 1 in all tissues including brain (IRDeltawb), and 1 restricted to peripheral tissues (IRDeltaper). While downregulation of IR expression resulted in severe hyperinsulinemia in both models, hyperglycemia was more pronounced in IRDeltawb mice. Both strains displayed a dramatic upregulation of hepatic leptin receptor expression, while only IRDeltaper mice displayed increased hepatic Stat3 phosphorylation and Il6 expression. Despite a similar reduction in IR expression in white adipose tissue (WAT) mass in both models, IRDeltawb mice had a more pronounced reduction in WAT mass and severe hypoleptinemia. Leptin replacement restored hepatic Stat3 phosphorylation and normalized glucose metabolism in these mice, indicating that alterations in glucose metabolism occur largely as a consequence of lipoathrophy upon body-wide IR deletion. Moreover, chronic intracerebroventricular insulin treatment of control mice increased fat mass, fat cell size, and adipose tissue lipoprotein lipase expression, indicating that CNS insulin action promotes lipogenesis. These studies demonstrate that central insulin action plays an important role in regulating WAT mass and glucose metabolism via hepatic Stat3 activation.


Assuntos
Tecido Adiposo/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Insulina/metabolismo , Lipogênese , Animais , Sistema Nervoso Central/metabolismo , Deleção de Genes , Homozigoto , Lipase Lipoproteica/biossíntese , Camundongos , Camundongos Knockout , Modelos Biológicos , Modelos Genéticos , Distribuição Tecidual
13.
Am J Physiol Endocrinol Metab ; 294(5): E810-6, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18285523

RESUMO

As obesity, diabetes, and associated comorbidities are on a constant rise, large efforts have been put into better understanding the cellular and molecular mechanisms by which nutrients and metabolic signals influence central and peripheral energy regulation. For decades, peripheral organs as a source and a target of such cues have been the focus of study. Their ability to integrate metabolic signals is essential for balanced energy and glucose metabolism. Only recently has the pivotal role of the central nervous system in the control of fuel partitioning been recognized. The rapidly expanding knowledge on the elucidation of molecular mechanisms and neuronal circuits involved is the focus of this review.


Assuntos
Glucose/metabolismo , Hormônios/fisiologia , Rede Nervosa/fisiologia , Fenômenos Fisiológicos da Nutrição/fisiologia , Transdução de Sinais/fisiologia , Animais , Glucose/biossíntese , Humanos , Canais KATP/fisiologia , Fígado/metabolismo
14.
Cell Metab ; 6(6): 431-45, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18054313

RESUMO

The contribution of different leptin-induced signaling pathways in control of energy homeostasis is only partly understood. Here we show that selective Pten ablation in leptin-sensitive neurons (Pten(DeltaObRb)) results in enhanced Pi3k activation in these cells and reduces adiposity by increasing energy expenditure. White adipose tissue (WAT) of Pten(DeltaObRb) mice shows characteristics of brown adipose tissue (BAT), reflected by increased mitochondrial content and Ucp1 expression resulting from enhanced leptin-stimulated sympathetic nerve activity (SNA) in WAT. In contrast, leptin-deficient ob/ob-Pten(DeltaObRb) mice exhibit unaltered body weight and WAT morphology compared to ob/ob mice, pointing to a pivotal role of endogenous leptin in control of WAT transdifferentiation. Leanness of Pten(DeltaObRb) mice is accompanied by enhanced sensitivity to insulin in skeletal muscle. These data provide direct genetic evidence that leptin-stimulated Pi3k signaling in the CNS regulates energy expenditure via activation of SNA to perigonadal WAT leading to BAT-like differentiation of WAT.


Assuntos
Tecido Adiposo Branco/crescimento & desenvolvimento , Tecido Adiposo Branco/metabolismo , Sistema Nervoso Central/metabolismo , Leptina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Tecido Adiposo Marrom/crescimento & desenvolvimento , Tecido Adiposo Marrom/metabolismo , Animais , Transdiferenciação Celular , Ativação Enzimática , Glucose/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Obesos , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Transdução de Sinais , Magreza
15.
Cell Metab ; 5(6): 438-49, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17550779

RESUMO

Insulin action in the central nervous system regulates energy homeostasis and glucose metabolism. To define the insulin-responsive neurons that mediate these effects, we generated mice with selective inactivation of the insulin receptor (IR) in either pro-opiomelanocortin (POMC)- or agouti-related peptide (AgRP)-expressing neurons of the arcuate nucleus of the hypothalamus. While neither POMC- nor AgRP-restricted IR knockout mice exhibited altered energy homeostasis, insulin failed to normally suppress hepatic glucose production during euglycemic-hyperinsulinemic clamps in AgRP-IR knockout (IR(DeltaAgRP)) mice. These mice also exhibited reduced insulin-stimulated hepatic interleukin-6 expression and increased hepatic expression of glucose-6-phosphatase. These results directly demonstrate that insulin action in POMC and AgRP cells is not required for steady-state regulation of food intake and body weight. However, insulin action specifically in AgRP-expressing neurons does play a critical role in controlling hepatic glucose production and may provide a target for the treatment of insulin resistance in type 2 diabetes.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Glucose/metabolismo , Insulina/farmacologia , Fígado/metabolismo , Neurônios/efeitos dos fármacos , Animais , Western Blotting , Peso Corporal , Eletrofisiologia , Feminino , Teste de Tolerância a Glucose , Glucose-6-Fosfatase/metabolismo , Homeostase , Hiperinsulinismo/metabolismo , Hipotálamo/metabolismo , Técnicas Imunoenzimáticas , Integrases/metabolismo , Interleucina-6/metabolismo , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptor de Insulina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA