Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
4.
Curr Opin Clin Nutr Metab Care ; 17(4): 312-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24848531

RESUMO

PURPOSE OF REVIEW: Rising evidence suggest that variation in the gut microbiome at gene and species levels defines subsets of individuals who have increased risk of obesity-related metabolic disorders, including insulin resistance and type 2 diabetes, which is influenced by diet and genetic profile of the host. Our goal in this review is gathering the newest findings concerning gut microbiota composition and effects on host's metabolism. RECENT FINDINGS: Dietary changes have been shown as the most prominent shaper of gut microbiota composition, reflecting major phenotypes, which can also be transmitted to other individuals, in spite of genetic variances. Gut microbiota composition has also been presented as diversity, which may have important implications in metabolite production and consequent interference with inflammatory activation, insulin resistance, and obesity. SUMMARY: Specific approaches made it possible to comprehend some of the interactions between certain bacterial strains and their host, and how their metabolites may interfere with host's cell signaling, changing its metabolic profile. Herein, we discuss some of the mechanisms by which alterations in the gut microbiota composition may contribute to the pathophysiology of obesity and its related comorbidities.


Assuntos
Trato Gastrointestinal/microbiologia , Resistência à Insulina , Microbiota , Obesidade/microbiologia , Ácidos e Sais Biliares/metabolismo , Diabetes Mellitus Tipo 2/microbiologia , Humanos
5.
Nutrients ; 5(3): 829-51, 2013 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-23482058

RESUMO

The development of obesity and insulin resistance has been extensively studied in the last decades, but the mechanisms underlying these alterations are still not completely understood. The gut microbiota has been identified as a potential contributor to metabolic diseases. It has been shown that obese individuals present different proportions of bacterial phyla compared with lean individuals, with an increase in Firmicutes and Actinobacteria and a decrease in Bacteroidetes. This alteration seems to interfere with intestinal permeability, increasing the absorption of lipopolysaccharide (LPS), which reaches circulation and initiates activation of Toll-like receptor (TLR) 4 and 2 and LPS receptor CD14, leading to increased activation of inflammatory pathways. With these activations, an impairment of the insulin signaling is observed, with decreased phosphorylation of the insulin receptor, insulin receptor substrate (IRS) and Akt, as well as increased inhibitory serine phosphorylation of IRS-1. Altered proportions of bacterial phyla have also been demonstrated to interfere with host's biochemical pathways, increasing energy extraction and depot in adipose tissue. Therefore, understanding the mechanisms by which the alteration in the gut microbiota produces different signaling activations and phenotype changes may offer an interesting opportunity for the treatment of obesity and type 2 diabetes.


Assuntos
Bactérias/classificação , Resistência à Insulina/fisiologia , Intestinos/microbiologia , Animais , Bactérias/isolamento & purificação , Regulação da Expressão Gênica , Humanos , Inflamação , Resistência à Insulina/genética , Obesidade/etiologia , Obesidade/microbiologia
6.
J Endocrinol ; 217(2): 131-40, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23349329

RESUMO

The 72 kDa inositol polyphosphate 5-phosphatase E (72k-5ptase) controls signal transduction through the catalytic dephosphorylation of the 5-position of membrane-bound phosphoinositides. The reduction of 72k-5ptase expression in the hypothalamus results in improved hypothalamic insulin signal transduction and reduction of food intake and body mass. Here, we evaluated the tissue distribution and the impact of obesity on the expression of 72k-5ptase in peripheral tissues of experimental animals. In addition, insulin signal transduction and action were determined in an animal model of obesity and insulin resistance treated with an antisense (AS) oligonucleotide that reduces 72k-5ptase expression. In lean Wistar rats, 72k-5ptase mRNA and protein are found in highest levels in heart, skeletal muscle, and white adipose tissue. In three distinct models of obesity, Wistar rats, Swiss mice fed on high-fat diet, and leptin-deficient ob/ob mice, the expression of 72k-5ptase is increased in skeletal muscle and adipose tissue. The treatment of obese Wistar rats with an anti-72k-5ptase AS oligonucleotide results in significant reduction of 72k-5ptase catalytic activity, which is accompanied by reduced food intake and body mass and improved insulin signal transduction and action as determined by immunoblotting and clamp studies respectively. 72k-5ptase expression is increased in obesity and its AS inhibition resulted in a significant improvement in insulin signal transduction and restoration of glucose homeostasis.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Insulina/fisiologia , Obesidade/etiologia , Obesidade/fisiopatologia , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Transdução de Sinais/fisiologia , Tecido Adiposo Branco/enzimologia , Animais , Modelos Animais de Doenças , Inositol Polifosfato 5-Fosfatases , Resistência à Insulina/fisiologia , Leptina/deficiência , Masculino , Camundongos , Camundongos Obesos , Músculo Esquelético/enzimologia , Miocárdio/enzimologia , Obesidade/metabolismo , Oligorribonucleotídeos Antissenso/farmacologia , Monoéster Fosfórico Hidrolases/efeitos dos fármacos , Monoéster Fosfórico Hidrolases/metabolismo , Ratos , Ratos Wistar
7.
Diabetes ; 62(2): 466-70, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22991447

RESUMO

Accumulating evidence has demonstrated that S-nitrosation of proteins plays a critical role in several human diseases. Here, we explored the role of inducible nitric oxide synthase (iNOS) in the S-nitrosation of proteins involved in the early steps of the insulin-signaling pathway and insulin resistance in the skeletal muscle of aged mice. Aging increased iNOS expression and S-nitrosation of major proteins involved in insulin signaling, thereby reducing insulin sensitivity in skeletal muscle. Conversely, aged iNOS-null mice were protected from S-nitrosation-induced insulin resistance. Moreover, pharmacological treatment with an iNOS inhibitor and acute exercise reduced iNOS-induced S-nitrosation and increased insulin sensitivity in the muscle of aged animals. These findings indicate that the insulin resistance observed in aged mice is mainly mediated through the S-nitrosation of the insulin-signaling pathway.


Assuntos
Envelhecimento/metabolismo , Resistência à Insulina/fisiologia , Músculo Esquelético/enzimologia , Óxido Nítrico Sintase Tipo II/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Inibidores Enzimáticos/farmacologia , Insulina/metabolismo , Lisina/análogos & derivados , Lisina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Nitrosação , Condicionamento Físico Animal , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
8.
Diabetes ; 62(1): 137-48, 2013 01.
Artigo em Inglês | MEDLINE | ID: mdl-22966070

RESUMO

Mutation of tub gene in mice induces obesity, suggesting that tub could be an important regulator of energy balance. In the current study, we investigated whether insulin, leptin, and obesity can modulate Tub in vivo in hypothalamic nuclei, and we investigated possible consequences on energy balance, neuropeptide expression, and hepatic glucose metabolism. Food intake, metabolic characteristics, signaling proteins, and neuropeptide expression were measured in response to fasting and refeeding, intracerebroventricular insulin and leptin, and Tub antisense oligonucleotide (ASO). Tub tyrosine phosphorylation (Tub-p-tyr) is modulated by nutritional status. Tub is a substrate of insulin receptor tyrosine kinase (IRTK) and leptin receptor (LEPR)-Janus kinase 2 (JAK2) in hypothalamic nuclei. After leptin or insulin stimulation, Tub translocates to the nucleus. Inhibition of Tub expression in hypothalamus by ASO increased food intake, fasting blood glucose, and hepatic glucose output, decreased O(2) consumption, and blunted the effect of insulin or leptin on proopiomelanocortin, thyroid-releasing hormone, melanin-concentrating hormone, and orexin expression. In hypothalamus of mice administered a high-fat diet, there is a reduction in leptin and insulin-induced Tub-p-tyr and nuclear translocation, which is reversed by reducing protein tyrosine phosphatase 1B expression. These results indicate that Tub has a key role in the control of insulin and leptin effects on food intake, and the modulation of Tub may contribute to insulin and leptin resistance in DIO mice.


Assuntos
Hipotálamo/fisiologia , Insulina/farmacologia , Leptina/farmacologia , Proteínas/fisiologia , Transdução de Sinais/fisiologia , Transporte Ativo do Núcleo Celular , Proteínas Adaptadoras de Transdução de Sinal , Animais , Jejum , Janus Quinase 2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligonucleotídeos Antissenso/farmacologia , Fosfolipase C beta/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Proteínas/antagonistas & inibidores
9.
Endocrinology ; 153(9): 4401-11, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22822160

RESUMO

Topiramate (TPM) treatment has been shown to reduce adiposity in humans and rodents. The reduction in adiposity is related to decreased food intake and increased energy expenditure. However, the molecular mechanisms through which TPM induces weight loss are contradictory and remain to be clarified. Whether TPM treatment alters hypothalamic insulin, or leptin signaling and action, is not well established. Thus, we investigate herein whether short-term TPM treatment alters energy balance by affecting insulin and leptin signaling, action, or neuropeptide expression in the hypothalamus of mice fed with a high-fat diet. As expected, short-term treatment with TPM diminished adiposity in obese mice mainly due to reduced food intake. TPM increased anorexigenic signaling by enhancing the leptin-induced leptin receptor/Janus kinase 2/signal transducer and activator of transcription 3 pathway and the insulin-induced insulin receptor substrate/Akt/forkhead box O1 pathway in parallel to reduced phosphatase protein expression in the hypothalamus of obese mice. These effects were independent of body weight. TPM also raised anorexigenic neuropeptides such as POMC, TRH, and CRH mRNA levels in obese mice. In addition, TPM increased the activation of the hypothalamic MAPK/ERK pathway induced by leptin, accompanied by an increase in peroxisome proliferator-activated receptor-coactivator α and uncoupling protein 1 protein levels in brown adipose tissue. Furthermore, TPM increased AMP-activated protein kinase and acetyl-coenzyme A carboxylase phosphorylation in peripheral tissues, which may help improve energy metabolism in these tissues. Together, these results provide novel insights into the molecular mechanisms through which TPM treatment reduces adiposity.


Assuntos
Frutose/análogos & derivados , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Animais , Frutose/uso terapêutico , Masculino , Camundongos , Topiramato
10.
PLoS One ; 7(5): e36974, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22662132

RESUMO

BACKGROUND: Wound healing is impaired in diabetes mellitus, but the mechanisms involved in this process are virtually unknown. Proteins belonging to the insulin signaling pathway respond to insulin in the skin of rats. OBJECTIVE: The purpose of this study was to investigate the regulation of the insulin signaling pathway in wound healing and skin repair of normal and diabetic rats, and, in parallel, the effect of a topical insulin cream on wound healing and on the activation of this pathway. RESEARCH DESIGN AND METHODS: We investigated insulin signaling by immunoblotting during wound healing of control and diabetic animals with or without topical insulin. Diabetic patients with ulcers were randomized to receive topical insulin or placebo in a prospective, double-blind and placebo-controlled, randomized clinical trial (NCT 01295177) of wound healing. RESULTS AND CONCLUSIONS: Expression of IR, IRS-1, IRS-2, SHC, ERK, and AKT are increased in the tissue of healing wounds compared to intact skin, suggesting that the insulin signaling pathway may have an important role in this process. These pathways were attenuated in the wounded skin of diabetic rats, in parallel with an increase in the time of complete wound healing. Upon topical application of insulin cream, the wound healing time of diabetic animals was normalized, followed by a reversal of defective insulin signal transduction. In addition, the treatment also increased expression of other proteins, such as eNOS (also in bone marrow), VEGF, and SDF-1α in wounded skin. In diabetic patients, topical insulin cream markedly improved wound healing, representing an attractive and cost-free method for treating this devastating complication of diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT01295177.


Assuntos
Complicações do Diabetes/tratamento farmacológico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Insulina/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Administração Tópica , Idoso , Animais , Medula Óssea/metabolismo , Quimiocina CXCL12/metabolismo , Cromonas/farmacologia , Complicações do Diabetes/metabolismo , Diabetes Mellitus Experimental/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Morfolinas/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Ratos Wistar , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Diabetes ; 61(6): 1455-62, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22522614

RESUMO

Defective liver gluconeogenesis is the main mechanism leading to fasting hyperglycemia in type 2 diabetes, and, in concert with steatosis, it is the hallmark of hepatic insulin resistance. Experimental obesity results, at least in part, from hypothalamic inflammation, which leads to leptin resistance and defective regulation of energy homeostasis. Pharmacological or genetic disruption of hypothalamic inflammation restores leptin sensitivity and reduces adiposity. Here, we evaluate the effect of a hypothalamic anti-inflammatory approach to regulating hepatic responsiveness to insulin. Obese rodents were treated by intracerebroventricular injections, with immunoneutralizing antibodies against Toll-like receptor (TLR)4 or tumor necrosis factor (TNF)α, and insulin signal transduction, hepatic steatosis, and gluconeogenesis were evaluated. The inhibition of either TLR4 or TNFα reduced hypothalamic inflammation, which was accompanied by the reduction of hypothalamic resistance to leptin and improved insulin signal transduction in the liver. This was accompanied by reduced liver steatosis and reduced hepatic expression of markers of steatosis. Furthermore, the inhibition of hypothalamic inflammation restored defective liver glucose production. All these beneficial effects were abrogated by vagotomy. Thus, the inhibition of hypothalamic inflammation in obesity results in improved hepatic insulin signal transduction, leading to reduced steatosis and reduced gluconeogenesis. All these effects are mediated by parasympathetic signals delivered by the vagus nerve.


Assuntos
Anticorpos Neutralizantes/administração & dosagem , Hipotálamo/metabolismo , Inflamação/metabolismo , Resistência à Insulina/fisiologia , Fígado/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/fisiologia , Homeostase/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Inflamação/tratamento farmacológico , Insulina/metabolismo , Leptina/metabolismo , Fígado/efeitos dos fármacos , Masculino , Camundongos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
12.
PLoS Biol ; 9(12): e1001212, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22162948

RESUMO

Environmental factors and host genetics interact to control the gut microbiota, which may have a role in the development of obesity and insulin resistance. TLR2-deficient mice, under germ-free conditions, are protected from diet-induced insulin resistance. It is possible that the presence of gut microbiota could reverse the phenotype of an animal, inducing insulin resistance in an animal genetically determined to have increased insulin sensitivity, such as the TLR2 KO mice. In the present study, we investigated the influence of gut microbiota on metabolic parameters, glucose tolerance, insulin sensitivity, and signaling of TLR2-deficient mice. We investigated the gut microbiota (by metagenomics), the metabolic characteristics, and insulin signaling in TLR2 knockout (KO) mice in a non-germ free facility. Results showed that the loss of TLR2 in conventionalized mice results in a phenotype reminiscent of metabolic syndrome, characterized by differences in the gut microbiota, with a 3-fold increase in Firmicutes and a slight increase in Bacteroidetes compared with controls. These changes in gut microbiota were accompanied by an increase in LPS absorption, subclinical inflammation, insulin resistance, glucose intolerance, and later, obesity. In addition, this sequence of events was reproduced in WT mice by microbiota transplantation and was also reversed by antibiotics. At the molecular level the mechanism was unique, with activation of TLR4 associated with ER stress and JNK activation, but no activation of the IKKß-IκB-NFκB pathway. Our data also showed that in TLR2 KO mice there was a reduction in regulatory T cell in visceral fat, suggesting that this modulation may also contribute to the insulin resistance of these animals. Our results emphasize the role of microbiota in the complex network of molecular and cellular interactions that link genotype to phenotype and have potential implications for common human disorders involving obesity, diabetes, and even other immunological disorders.


Assuntos
Resistência à Insulina , Intestinos/microbiologia , Síndrome Metabólica/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Antibacterianos/uso terapêutico , Cruzamentos Genéticos , Dieta Hiperlipídica/efeitos adversos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Intestinos/imunologia , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Síndrome Metabólica/tratamento farmacológico , Metagenômica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória , Organismos Livres de Patógenos Específicos , Linfócitos T Reguladores/imunologia , Receptor 2 Toll-Like/genética
13.
J Endocrinol ; 199(3): 399-406, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18787058

RESUMO

The aims of the present study were to investigate the expression of toll-like receptor 2 (TLR2) in muscle and white adipose tissue (WAT) of diet-induced obesity (DIO) mice, and also the effects of its inhibition, with the use of TLR2 antisense oligonucleotide (ASON), on insulin sensitivity and signaling. The expression of TLR2 was increased in muscle and WAT of DIO mice, compared with those that received standard chow. Inhibition of TLR2 in DIO mice, by TLR2 ASON, improved insulin sensitivity and signaling in muscle and WAT. In addition, data show that the inhibition of TLR2 expression prevents the activation of IKBKB, MAPK8, and serine phosphorylation of IRS1 in DIO mice, suggesting that TLR2 is a key modulator of the crosstalk between inflammatory and metabolic pathways. We, therefore, suggest that a selective interference with TLR2 presents an attractive opportunity for the treatment of insulin resistance in obesity and type 2 diabetes.


Assuntos
Tecido Adiposo Branco/metabolismo , Gorduras na Dieta/farmacologia , Expressão Gênica/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptor 2 Toll-Like/antagonistas & inibidores , Tecido Adiposo Branco/efeitos dos fármacos , Animais , Glicemia/efeitos dos fármacos , Immunoblotting , Imunoprecipitação , Insulina/sangue , Resistência à Insulina , Masculino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...