Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
2.
Metabolism ; 153: 155796, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38262576

RESUMO

Imeglimin is a recently developed anti-diabetic drug that could concurrently promote insulin secretion and insulin sensitivity, while its mechanisms of action are not fully understood. Here we show that imeglimin administration could protect mice from high fat diet-induced weight gain with enhanced energy expenditure and attenuated whitening of brown adipose tissue. Imeglimin administration led to significant alteration of gut microbiota, which included an increase of Akkermansia genus, with attenuation of obesity-associated gut pathologies. Ablation of microbiota by antibiotic treatment partially abrogated the insulin sensitizing effects of imeglimin, while not affecting its actions on body weight gain or brown adipose tissue. Collectively, our results characterize imeglimin as a potential agent promoting energy expenditure and gut integrity, providing new insights into its mechanisms of action.


Assuntos
Microbioma Gastrointestinal , Triazinas , Animais , Camundongos , Tecido Adiposo Marrom , Camundongos Obesos , Obesidade/tratamento farmacológico , Aumento de Peso
3.
Nat Commun ; 14(1): 6584, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37852976

RESUMO

Diabetes is known to increase the risk of nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). Here we treat male STAM (STelic Animal Model) mice, which develop diabetes, NASH and HCC associated with dysbiosis upon low-dose streptozotocin and high-fat diet (HFD), with insulin or phlorizin. Although both treatments ameliorate hyperglycemia and NASH, insulin treatment alone lead to suppression of HCC accompanied by improvement of dysbiosis and restoration of antimicrobial peptide production. There are some similarities in changes of microflora from insulin-treated patients comorbid with diabetes and NASH. Insulin treatment, however, fails to suppress HCC in the male STAM mice lacking insulin receptor specifically in intestinal epithelial cells (ieIRKO), which show dysbiosis and impaired gut barrier function. Furthermore, male ieIRKO mice are prone to develop HCC merely on HFD. These data suggest that impaired gut insulin signaling increases the risk of HCC, which can be countered by restoration of insulin action in diabetes.


Assuntos
Carcinoma Hepatocelular , Diabetes Mellitus Experimental , Neoplasias Hepáticas , Hepatopatia Gordurosa não Alcoólica , Humanos , Masculino , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/patologia , Fígado/patologia , Carcinoma Hepatocelular/patologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Disbiose/complicações , Disbiose/patologia , Neoplasias Hepáticas/patologia , Insulina , Camundongos Endogâmicos C57BL , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças
4.
Cell Chem Biol ; 29(4): 680-689.e6, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-34986326

RESUMO

During insulin resistance, lipid uptake by the liver is promoted by peroxisome proliferator-activated protein (PPAR) γ upregulation, leading to hepatic steatosis. Insulin, however, does not directly regulate adipogenic gene expression in liver, and the mechanisms for its upregulation in obesity remain unclear. Here, we show that the Irs2 locus, a critical regulator of insulin actions, encodes an antisense transcript, ASIrs2, whose expression increases in obesity or after refeeding in liver, reciprocal to that of Irs2. ASIrs2 regulates hepatic Pparg expression, and its suppression ameliorates steatosis in obese mice. The human ortholog AL162497.1, whose expression is correlated with that of hepatic PPARG and the severity of non-alcoholic steatohepatitis (NASH), shows genomic organization similar to that of ASIrs2. We also identified HARS2 as a potential binding protein for ASIrs2, functioning as a regulator of Pparg. Collectively, our data reveal a functional duality of the Irs2 gene locus, where reciprocal changes of Irs2 and ASIrs2 in obesity cause insulin resistance and steatosis.


Assuntos
Resistência à Insulina , Hepatopatia Gordurosa não Alcoólica , Animais , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina/genética , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/complicações , Obesidade/genética , PPAR gama/genética
5.
Diabetol Int ; 12(4): 343-348, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34567916

RESUMO

In recent years, the techniques in molecular biology have been dramatically advanced, and consequently the landscape of metabolism research has undergone a remarkable change. One of the emerging pictures as the fruits of these advancements is one depicting the regulation of systemic metabolism through inter-organ networks involving multiple tissues, either via humoral factors, which are secreted from one tissue and conveyed to their remote target tissues, or through neuronal networks which are integrated by the central nervous system. In addition, the progress in high-throughput research tools enabled detailed characterization and deeper understanding of the nature of human genome, which has attracted much attention to the importance of various non-coding RNAs species. These non-coding RNAs are often co-expressed and co-regulated with adjacent protein coding genes, adding higher levels of complexities by them functioning together as a system and often influencing biologically important pathways in a cooperative manner. Here in this review several examples of these regulatory network systems are presented, illustrating the significance of them in systemic metabolism, with a possible future research direction also being proposed.

6.
Diabetes ; 70(7): 1519-1535, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33906910

RESUMO

In diabetic pathology, insufficiency in ß-cell mass, unable to meet peripheral insulin demand, and functional defects of individual ß-cells in production of insulin are often concurrently observed, collectively causing hyperglycemia. Here we show that the phosphorylation of ERK1/2 is significantly decreased in the islets of db/db mice as well as in those of a cohort of subjects with type 2 diabetes. In mice with abrogation of ERK signaling in pancreatic ß-cells through deletion of Mek1 and Mek2, glucose intolerance aggravates under high-fat diet-feeding conditions due to insufficient insulin production with lower ß-cell proliferation and reduced ß-cell mass, while in individual ß-cells dampening of the number of insulin exocytosis events is observed, with the molecules involved in insulin exocytosis being less phosphorylated. These data reveal bifunctional roles for MEK/ERK signaling in ß-cells for glucose homeostasis, i.e., in regulating ß-cell mass as well as in controlling insulin exocytosis in individual ß-cells, thus providing not only a novel perspective for the understanding of diabetes pathophysiology but also a potential clue for new drug development for diabetes treatment.


Assuntos
Glicemia/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Homeostase , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Animais , Linhagem Celular , Dieta Hiperlipídica , Exocitose , Humanos , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais
7.
Nat Commun ; 11(1): 644, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-32005828

RESUMO

Obesity and type 2 diabetes mellitus are global emergencies and long noncoding RNAs (lncRNAs) are regulatory transcripts with elusive functions in metabolism. Here we show that a high fraction of lncRNAs, but not protein-coding mRNAs, are repressed during diet-induced obesity (DIO) and refeeding, whilst nutrient deprivation induced lncRNAs in mouse liver. Similarly, lncRNAs are lost in diabetic humans. LncRNA promoter analyses, global cistrome and gain-of-function analyses confirm that increased MAFG signaling during DIO curbs lncRNA expression. Silencing Mafg in mouse hepatocytes and obese mice elicits a fasting-like gene expression profile, improves glucose metabolism, de-represses lncRNAs and impairs mammalian target of rapamycin (mTOR) activation. We find that obesity-repressed LincIRS2 is controlled by MAFG and observe that genetic and RNAi-mediated LincIRS2 loss causes elevated blood glucose, insulin resistance and aberrant glucose output in lean mice. Taken together, we identify a MAFG-lncRNA axis controlling hepatic glucose metabolism in health and metabolic disease.


Assuntos
Diabetes Mellitus Tipo 2/genética , Glucose/metabolismo , Fígado/metabolismo , Fator de Transcrição MafG/genética , Obesidade/genética , RNA Longo não Codificante/genética , Proteínas Repressoras/genética , Idoso , Animais , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Fator de Transcrição MafG/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Obesidade/metabolismo , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
8.
Cell ; 177(6): 1536-1552.e23, 2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31150623

RESUMO

Ectopic lipid deposition and altered mitochondrial dynamics contribute to the development of obesity and insulin resistance. However, the mechanistic link between these processes remained unclear. Here we demonstrate that the C16:0 sphingolipid synthesizing ceramide synthases, CerS5 and CerS6, affect distinct sphingolipid pools and that abrogation of CerS6 but not of CerS5 protects from obesity and insulin resistance. We identify proteins that specifically interact with C16:0 sphingolipids derived from CerS5 or CerS6. Here, only CerS6-derived C16:0 sphingolipids bind the mitochondrial fission factor (Mff). CerS6 and Mff deficiency protect from fatty acid-induced mitochondrial fragmentation in vitro, and the two proteins genetically interact in vivo in obesity-induced mitochondrial fragmentation and development of insulin resistance. Our experiments reveal an unprecedented specificity of sphingolipid signaling depending on specific synthesizing enzymes, provide a mechanistic link between hepatic lipid deposition and mitochondrial fragmentation in obesity, and define the CerS6-derived sphingolipid/Mff interaction as a therapeutic target for metabolic diseases.


Assuntos
Proteínas de Membrana/metabolismo , Obesidade/metabolismo , Esfingolipídeos/metabolismo , Esfingosina N-Aciltransferase/metabolismo , Animais , Apoptose , Linhagem Celular , Células HeLa , Humanos , Resistência à Insulina/fisiologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Proteínas Mitocondriais/metabolismo , Obesidade/fisiopatologia , Esfingolipídeos/fisiologia , Esfingosina N-Aciltransferase/fisiologia
9.
Cell Rep ; 26(1): 1-10.e7, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30605666

RESUMO

Skeletal muscle accumulates ceramides in obesity, which contribute to the development of obesity-associated insulin resistance. However, it remained unclear which distinct ceramide species in this organ contributes to instatement of systemic insulin resistance. Here, ceramide profiling of high-fat diet (HFD)-fed animals revealed increased skeletal muscle C18:0 ceramide content, concomitant with increased expression of ceramide synthase (CerS)1. Mice lacking CerS1, either globally or specifically in skeletal muscle (CerS1ΔSkM), exhibit reduced muscle C18:0 ceramide content and significant improvements in systemic glucose homeostasis. CerS1ΔSkM mice exhibit improved insulin-stimulated suppression of hepatic glucose production, and lack of CerS1 in skeletal muscle improves systemic glucose homeostasis via increased release of Fgf21 from skeletal muscle. In contrast, muscle-specific deficiency of C16:0 ceramide-producing CerS5 and CerS6 failed to protect mice from obesity-induced insulin resistance. Collectively, these results reveal the tissue-specific function of distinct ceramide species during the development of obesity-associated insulin resistance.


Assuntos
Ceramidas/efeitos adversos , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina/genética , Proteínas de Membrana/metabolismo , Músculo Esquelético/metabolismo , Obesidade/genética , Esfingosina N-Aciltransferase/metabolismo , Animais , Humanos , Masculino , Camundongos
10.
J Med Case Rep ; 12(1): 368, 2018 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-30545408

RESUMO

INTRODUCTION: Acquired partial lipoatrophy has been reported after bone marrow transplantation during childhood; however, no adult cases have previously been reported. We herein report two adult cases of acquired partial lipoatrophy after transplantation. CASE PRESENTATION: A 28-year-old Japanese woman developed diabetic ketoacidosis and received insulin therapy after bone marrow transplantation. She manifested partial lipoatrophy of the extremities, prominent insulin resistance, hyperglycemia, hypertriglyceridemia, and fatty liver. A 40-year-old Japanese woman underwent liver transplantation from a living donor for alcoholic liver disease after abstinence from alcohol. She newly developed non-alcoholic steatohepatitis and diabetes. Non-alcoholic steatohepatitis progressed to liver failure, and a second liver transplantation from a brain-dead donor was performed at 42 years of age. She demonstrated loss of subdermal fat of the upper and lower extremities, prominent insulin resistance, hyperglycemia, and hypertriglyceridemia. In both cases, the injection of recombinant methionyl human leptin reversed all of the metabolic abnormalities. CONCLUSIONS: Acquired partial lipoatrophy after transplantation is a manifestation of chronic graft-versus-host disease in adults. This entity is associated with diabetes with prominent insulin resistance and severe hypertriglycemia and can be successfully treated with metreleptin for the long term.


Assuntos
Doença Enxerto-Hospedeiro/complicações , Doença Enxerto-Hospedeiro/terapia , Leptina/análogos & derivados , Lipodistrofia/etiologia , Lipodistrofia/terapia , Adulto , Feminino , Doença Enxerto-Hospedeiro/diagnóstico , Humanos , Leptina/uso terapêutico , Lipodistrofia/diagnóstico , Resultado do Tratamento
11.
Cell ; 175(5): 1321-1335.e20, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30445039

RESUMO

Adaptation of liver to the postprandial state requires coordinated regulation of protein synthesis and folding aligned with changes in lipid metabolism. Here we demonstrate that sensory food perception is sufficient to elicit early activation of hepatic mTOR signaling, Xbp1 splicing, increased expression of ER-stress genes, and phosphatidylcholine synthesis, which translate into a rapid morphological ER remodeling. These responses overlap with those activated during refeeding, where they are maintained and constantly increased upon nutrient supply. Sensory food perception activates POMC neurons in the hypothalamus, optogenetic activation of POMC neurons activates hepatic mTOR signaling and Xbp1 splicing, whereas lack of MC4R expression attenuates these responses to sensory food perception. Chemogenetic POMC-neuron activation promotes sympathetic nerve activity (SNA) subserving the liver, and norepinephrine evokes the same responses in hepatocytes in vitro and in liver in vivo as observed upon sensory food perception. Collectively, our experiments unravel that sensory food perception coordinately primes postprandial liver ER adaption through a melanocortin-SNA-mTOR-Xbp1s axis. VIDEO ABSTRACT.


Assuntos
Retículo Endoplasmático/metabolismo , Preferências Alimentares , Melanocortinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Feminino , Regulação da Expressão Gênica , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Norepinefrina/farmacologia , Fosfatidilcolinas/análise , Fosfatidilcolinas/metabolismo , Análise de Componente Principal , Receptor Tipo 4 de Melanocortina/deficiência , Receptor Tipo 4 de Melanocortina/genética , Proteína 1 de Ligação a X-Box/genética
12.
Nat Commun ; 9(1): 3622, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30190464

RESUMO

Increasing brown adipose tissue (BAT) thermogenesis in mice and humans improves metabolic health and understanding BAT function is of interest for novel approaches to counteract obesity. The role of long noncoding RNAs (lncRNAs) in these processes remains elusive. We observed maternally expressed, imprinted lncRNA H19 increased upon cold-activation and decreased in obesity in BAT. Inverse correlations of H19 with BMI were also observed in humans. H19 overexpression promoted, while silencing of H19 impaired adipogenesis, oxidative metabolism and mitochondrial respiration in brown but not white adipocytes. In vivo, H19 overexpression protected against DIO, improved insulin sensitivity and mitochondrial biogenesis, whereas fat H19 loss sensitized towards HFD weight gains. Strikingly, paternally expressed genes (PEG) were largely absent from BAT and we demonstrated that H19 recruits PEG-inactivating H19-MBD1 complexes and acts as BAT-selective PEG gatekeeper. This has implications for our understanding how monoallelic gene expression affects metabolism in rodents and, potentially, humans.


Assuntos
Tecido Adiposo Marrom/fisiologia , Impressão Genômica , Obesidade/genética , RNA Longo não Codificante/genética , Tecido Adiposo Marrom/patologia , Tecido Adiposo Branco/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético/genética , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Obesidade/etiologia
13.
Mol Cell Biol ; 38(16)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29866655

RESUMO

Adipocyte differentiation is regulated by various mechanisms, of which mitotic clonal expansion (MCE) is a key step. Although this process is known to be regulated by cell cycle modulators, the precise mechanism remains unclear. N6-Methyladenosine (m6A) posttranscriptional RNA modification, whose methylation and demethylation are performed by respective enzyme molecules, has recently been suggested to be involved in the regulation of adipogenesis. Here, we show that an RNA N6-adenosine methyltransferase complex consisting of Wilms' tumor 1-associating protein (WTAP), methyltransferase like 3 (METTL3), and METTL14 positively controls adipogenesis by promoting cell cycle transition in MCE during adipogenesis. WTAP, coupled with METTL3 and METTL14, is increased and distributed in nucleus by the induction of adipogenesis dependently on RNA in vitro Knockdown of each of these three proteins leads to cell cycle arrest and impaired adipogenesis associated with suppression of cyclin A2 upregulation during MCE, whose knockdown also impairs adipogenesis. Consistent with this, Wtap heterozygous knockout mice are protected from diet-induced obesity with smaller size and number of adipocytes, leading to improved insulin sensitivity. These data provide a mechanism for adipogenesis through the WTAP-METTL3-METTL14 complex and a potential strategy for treatment of obesity and associated disorders.


Assuntos
Adipogenia/fisiologia , Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Metiltransferases/metabolismo , Proteínas Nucleares/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Adipogenia/genética , Animais , Proteínas de Transporte/genética , Contagem de Células , Pontos de Checagem do Ciclo Celular/genética , Pontos de Checagem do Ciclo Celular/fisiologia , Proteínas de Ciclo Celular , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Núcleo Celular/metabolismo , Tamanho Celular , Células Clonais/citologia , Células Clonais/metabolismo , Ciclina A2/genética , Ciclina A2/metabolismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Metiltransferases/deficiência , Metiltransferases/genética , Camundongos , Camundongos Knockout , Mitose/genética , Mitose/fisiologia , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Processamento Pós-Transcricional do RNA , Fatores de Processamento de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
Nat Med ; 23(12): 1466-1473, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29106399

RESUMO

Over 40% of microRNAs (miRNAs) are located in introns of protein-coding genes, and many of these intronic miRNAs are co-regulated with their host genes. In such cases of co-regulation, the products of host genes and their intronic miRNAs can cooperate to coordinately regulate biologically important pathways. Therefore, we screened intronic miRNAs dysregulated in the livers of mouse models of obesity to identify previously uncharacterized protein-coding host genes that may contribute to the pathogenesis of obesity-associated insulin resistance and type 2 diabetes mellitus. Our approach revealed that expression of both the gene encoding ectodysplasin A (Eda), the causal gene in X-linked hypohidrotic ectodermal dysplasia (XLHED), and its intronic miRNA, miR-676, was increased in the livers of obese mice. Moreover, hepatic EDA expression is increased in obese human subjects and reduced upon weight loss, and its hepatic expression correlates with systemic insulin resistance. We also found that reducing miR-676 expression in db/db mice increases the expression of proteins involved in fatty acid oxidation and reduces the expression of inflammatory signaling components in the liver. Further, we found that Eda expression in mouse liver is controlled via PPARγ and RXR-α, increases in circulation under conditions of obesity, and promotes JNK activation and inhibitory serine phosphorylation of IRS1 in skeletal muscle. In accordance with these findings, gain- and loss-of-function approaches reveal that liver-derived EDA regulates systemic glucose metabolism, suggesting that EDA is a hepatokine that can contribute to impaired skeletal muscle insulin sensitivity in obesity.


Assuntos
Ectodisplasinas/genética , Resistência à Insulina/genética , Fígado/metabolismo , MicroRNAs/genética , Músculo Esquelético/metabolismo , Obesidade/genética , Animais , Células Cultivadas , Displasia Ectodérmica Anidrótica Tipo 1/genética , Ectodisplasinas/metabolismo , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Obesos , Camundongos Transgênicos , Obesidade/metabolismo
15.
Mol Cell Biol ; 34(7): 1290-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24469402

RESUMO

A growing body of evidence has underlined the significance of endoplasmic reticulum (ER) stress in the pathogenesis of diabetes mellitus. ER oxidoreductin 1ß (ERO1ß) is a pancreas-specific disulfide oxidase that is known to be upregulated in response to ER stress and to promote protein folding in pancreatic ß cells. It has recently been demonstrated that ERO1ß promotes insulin biogenesis in ß cells and thus contributes to physiological glucose homeostasis, though it is unknown if ERO1ß is involved in the pathogenesis of diabetes mellitus. Here we show that in diabetic model mice, ERO1ß expression is paradoxically decreased in ß cells despite the indications of increased ER stress. However, overexpression of ERO1ß in ß cells led to the upregulation of unfolded protein response genes and markedly enlarged ER lumens, indicating that ERO1ß overexpression caused ER stress in the ß cells. Insulin contents were decreased in the ß cells that overexpressed ERO1ß, leading to impaired insulin secretion in response to glucose stimulation. These data indicate the importance of the fine-tuning of the ER redox state, the disturbance of which would compromise the function of ß cells in insulin synthesis and thus contribute to the pathogenesis of diabetes mellitus.


Assuntos
Diabetes Mellitus Experimental/etiologia , Diabetes Mellitus Experimental/metabolismo , Estresse do Retículo Endoplasmático , Glicoproteínas/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Diabetes Mellitus Experimental/genética , Regulação da Expressão Gênica , Glicoproteínas/genética , Humanos , Insulina/biossíntese , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxirredutases , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/genética , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Dobramento de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Resposta a Proteínas não Dobradas/genética
16.
Cell ; 156(3): 495-509, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24462248

RESUMO

Maternal metabolic homeostasis exerts long-term effects on the offspring's health outcomes. Here, we demonstrate that maternal high-fat diet (HFD) feeding during lactation predisposes the offspring for obesity and impaired glucose homeostasis in mice, which is associated with an impairment of the hypothalamic melanocortin circuitry. Whereas the number and neuropeptide expression of anorexigenic proopiomelanocortin (POMC) and orexigenic agouti-related peptide (AgRP) neurons, electrophysiological properties of POMC neurons, and posttranslational processing of POMC remain unaffected in response to maternal HFD feeding during lactation, the formation of POMC and AgRP projections to hypothalamic target sites is severely impaired. Abrogating insulin action in POMC neurons of the offspring prevents altered POMC projections to the preautonomic paraventricular nucleus of the hypothalamus (PVH), pancreatic parasympathetic innervation, and impaired glucose-stimulated insulin secretion in response to maternal overnutrition. These experiments reveal a critical timing, when altered maternal metabolism disrupts metabolic homeostasis in the offspring via impairing neuronal projections, and show that abnormal insulin signaling contributes to this effect.


Assuntos
Dieta Hiperlipídica , Hiperglicemia/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Lactação , Obesidade/metabolismo , Animais , Axônios/metabolismo , Feminino , Masculino , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Gravidez , Pró-Opiomelanocortina/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais
17.
Cell Metab ; 13(4): 401-412, 2011 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-21459325

RESUMO

Insulin resistance is often associated with impeded insulin signaling due either to decreased concentrations or functional modifications of crucial signaling molecules including insulin receptor substrates (IRS) in the liver. Many actions of adiponectin, a well-recognized antidiabetic adipokine, are currently attributed to the activation of two critical molecules downstream of AdipoR1 and R2: AMP-activated kinase (AMPK) and peroxisome proliferator-activated receptor α (PPARα). However, the direct effects of adiponectin on insulin signaling molecules remain poorly understood. We show here that adiponectin upregulates IRS-2 through activation of signal transducer and activator of transcription-3 (STAT3). Surprisingly, this activation is associated with IL-6 production from macrophages induced by adiponectin through NFκB activation independent of its authentic receptors, AdipoR1 and AdipoR2. These data have unraveled an insulin-sensitizing action initiated by adiponectin leading to upregulation of hepatic IRS-2 via an IL-6 dependent pathway through a still unidentified adiponectin receptor.


Assuntos
Adiponectina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Interleucina-6/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Adiponectina/deficiência , Adiponectina/genética , Animais , Modelos Animais de Doenças , Proteínas Substratos do Receptor de Insulina/genética , Resistência à Insulina , Interleucina-6/deficiência , Interleucina-6/genética , Camundongos , Camundongos Obesos , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Receptores de Adiponectina/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
18.
Proc Natl Acad Sci U S A ; 108(14): 5753-8, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21436039

RESUMO

Obesity and insulin resistance, the key features of metabolic syndrome, are closely associated with a state of chronic, low-grade inflammation characterized by abnormal macrophage infiltration into adipose tissues. Although it has been reported that chemokines promote leukocyte migration by activating class IB phosphoinositide-3 kinase (PI3Kγ) in inflammatory states, little is known about the role of PI3Kγ in obesity-induced macrophage infiltration into tissues, systemic inflammation, and the development of insulin resistance. In the present study, we used murine models of both diet-induced and genetically induced obesity to examine the role of PI3Kγ in the accumulation of tissue macrophages and the development of obesity-induced insulin resistance. Mice lacking p110γ (Pik3cg(-/-)), the catalytic subunit of PI3Kγ, exhibited improved systemic insulin sensitivity with enhanced insulin signaling in the tissues of obese animals. In adipose tissues and livers of obese Pik3cg(-/-) mice, the numbers of infiltrated proinflammatory macrophages were markedly reduced, leading to suppression of inflammatory reactions in these tissues. Furthermore, bone marrow-specific deletion and pharmacological blockade of PI3Kγ also ameliorated obesity-induced macrophage infiltration and insulin resistance. These data suggest that PI3Kγ plays a crucial role in the development of both obesity-induced inflammation and systemic insulin resistance and that PI3Kγ can be a therapeutic target for type 2 diabetes.


Assuntos
Inflamação/tratamento farmacológico , Resistência à Insulina , Obesidade/complicações , Inibidores de Fosfoinositídeo-3 Quinase , Tecido Adiposo/citologia , Animais , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Técnicas Histológicas , Inflamação/etiologia , Fígado/citologia , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Quinoxalinas/farmacologia , Tiazolidinedionas/farmacologia
19.
Cell Metab ; 12(6): 619-32, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21109194

RESUMO

Type 2 diabetes is characterized by insulin resistance and pancreatic ß cell dysfunction, the latter possibly caused by a defect in insulin signaling in ß cells. Inhibition of class IA phosphatidylinositol 3-kinase (PI3K), using a mouse model lacking the pik3r1 gene specifically in ß cells and the pik3r2 gene systemically (ßDKO mouse), results in glucose intolerance and reduced insulin secretion in response to glucose. ß cells of ßDKO mice had defective exocytosis machinery due to decreased expression of soluble N-ethylmaleimide attachment protein receptor (SNARE) complex proteins and loss of cell-cell synchronization in terms of Ca(2+) influx. These defects were normalized by expression of a constitutively active form of Akt in the islets of ßDKO mice, preserving insulin secretion in response to glucose. The class IA PI3K pathway in ß cells in vivo is important in the regulation of insulin secretion and may be a therapeutic target for type 2 diabetes.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/enzimologia , Insulina/metabolismo , Análise de Variância , Animais , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Exocitose/fisiologia , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas SNARE/metabolismo
20.
Biochem Biophys Res Commun ; 382(1): 51-6, 2009 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-19254698

RESUMO

Adiponectin, one of the insulin-sensitizing adipokines, has been shown to activate fatty acid oxidation in liver and skeletal muscle, thus maintaining insulin sensitivity. However, the precise roles of adiponectin in fatty acid synthesis are poorly understood. Here we show that adiponectin administration acutely suppresses expression of sterol regulatory element-binding protein (SREBP) 1c, the master regulator which controls and upregulates the enzymes involved in fatty acid synthesis, in the liver of +Lepr(db)/+Lepr(db) (db/db) mouse as well as in cultured hepatocytes. We also show that adiponectin suppresses SREBP1c by AdipoR1, one of the functional receptors for adiponetin, and furthermore that suppressing either AMP-activated protein kinase (AMPK) via its upstream kinase LKB1 deletion cancels the negative effect of adiponectin on SREBP1c expression. These data show that adiponectin suppresses SREBP1c through the AdipoR1/LKB1/AMPK pathway, and suggest a possible role for adiponectin in the regulation of hepatic fatty acid synthesis.


Assuntos
Ácidos Graxos/metabolismo , Fígado/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/biossíntese , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Adiponectina/genética , Adiponectina/farmacologia , Adiponectina/fisiologia , Animais , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Adiponectina/metabolismo , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA