Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Biol Chem ; 296: 100482, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33647317

RESUMO

Skeletal muscle is responsible for the majority of glucose disposal following meals, and this is achieved by insulin-mediated trafficking of glucose transporter type 4 (GLUT4) to the cell membrane. The eight-protein exocyst trafficking complex facilitates targeted docking of membrane-bound vesicles, a process underlying the regulated delivery of fuel transporters. We previously demonstrated the role of exocyst subunit EXOC5 in insulin-stimulated GLUT4 exocytosis and glucose uptake in cultured rat skeletal myoblasts. However, the in vivo role of EXOC5 in skeletal muscle remains unclear. Using mice with inducible, skeletal-muscle-specific knockout of exocyst subunit EXOC5 (Exoc5-SMKO), we examined how muscle-specific disruption of the exocyst would affect glucose homeostasis in vivo. We found that both male and female Exoc5-SMKO mice displayed elevated fasting glucose levels. Additionally, male Exoc5-SMKO mice had impaired glucose tolerance and lower serum insulin levels. Using indirect calorimetry, we observed that male Exoc5-SMKO mice have a reduced respiratory exchange ratio during the light period and lower energy expenditure. Using the hyperinsulinemic-euglycemic clamp method, we further showed that insulin-stimulated skeletal muscle glucose uptake is reduced in Exoc5-SMKO males compared with wild-type controls. Overall, our findings indicate that EXOC5 and the exocyst are necessary for insulin-stimulated glucose uptake in skeletal muscle and regulate glucose homeostasis in vivo.


Assuntos
Glucose/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Metabolismo dos Carboidratos , Membrana Celular/metabolismo , Citoplasma/metabolismo , Exocitose , Feminino , Intolerância à Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Homeostase , Insulina/análise , Insulina/sangue , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos , Músculo Esquelético/fisiologia , Mioblastos Esqueléticos/metabolismo , Transporte Proteico , Proteínas de Transporte Vesicular/fisiologia
2.
Arterioscler Thromb Vasc Biol ; 31(8): 1805-13, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21636809

RESUMO

OBJECTIVE: There are several pathways that mediate the aberrant metabolism of glucose and that might induce greater vascular damage in the setting of diabetes. The polyol pathway mediated by aldose reductase (AR) has been postulated to be one such pathway. However, it has been reported that AR reduces toxic lipid aldehydes and, under some circumstances, might be antiatherogenic. METHODS AND RESULTS: Atherosclerosis development was quantified in 2 lines of transgenic mice expressing human AR (hAR) crossed on the apolipoprotein E knockout background. The transgenes were used to increase the normally low levels of this enzyme in wild-type mice. Both generalized hAR overexpression and hAR expression via the Tie 2 promoter increased lesion size in streptozotocin diabetic mice. In addition, pharmacological inhibition of AR reduced lesion size. CONCLUSIONS: Although in some settings AR expression might reduce levels of toxic aldehydes, transgenic expression of this enzyme within the artery wall leads to greater atherosclerosis.


Assuntos
Aldeído Redutase/metabolismo , Aterosclerose/etiologia , Diabetes Mellitus Experimental/metabolismo , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/genética , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Sequência de Bases , Linhagem Celular , DNA Complementar/genética , Complicações do Diabetes/etiologia , Complicações do Diabetes/genética , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Diabetes Mellitus Experimental/genética , Células Endoteliais/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Expressão Gênica , Glucose/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Interferente Pequeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vasodilatação/fisiologia
3.
Cells ; 10(8)2021 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-34440862

RESUMO

CEACAM1 regulates endothelial barrier integrity. Because insulin signaling in extrahepatic target tissues is regulated by insulin transport through the endothelium, we aimed at investigating the metabolic role of endothelial CEACAM1. To this end, we generated endothelial cell-specific Ceacam1 null mice (VECadCre+Cc1fl/fl) and carried out their metabolic phenotyping and mechanistic analysis by comparison to littermate controls. Hyperinsulinemic-euglycemic clamp analysis showed intact insulin sensitivity in VECadCre+Cc1fl/fl mice. This was associated with the absence of visceral obesity and lipolysis and normal levels of circulating non-esterified fatty acids, leptin, and adiponectin. Whereas the loss of endothelial Ceacam1 did not affect insulin-stimulated receptor phosphorylation, it reduced IRS-1/Akt/eNOS activation to lower nitric oxide production resulting from limited SHP2 sequestration. It also reduced Shc sequestration to activate NF-κB and increase the transcription of matrix metalloproteases, ultimately inducing plasma IL-6 and TNFα levels. Loss of endothelial Ceacam1 also induced the expression of the anti-inflammatory CEACAM1-4L variant in M2 macrophages in white adipose tissue. Together, this could cause endothelial barrier dysfunction and facilitate insulin transport, sustaining normal glucose homeostasis and retaining fat accumulation in adipocytes. The data assign a significant role for endothelial cell CEACAM1 in maintaining insulin sensitivity in peripheral extrahepatic target tissues.


Assuntos
Antígeno Carcinoembrionário/metabolismo , Células Endoteliais/metabolismo , Resistência à Insulina , Adipócitos/metabolismo , Animais , Antígeno Carcinoembrionário/genética , Endotélio Vascular/metabolismo , Gorduras/metabolismo , Glucose/metabolismo , Inflamação , Insulina/metabolismo , Resistência à Insulina/genética , Fígado/citologia , Fígado/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais
4.
Sci Rep ; 10(1): 4561, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32165710

RESUMO

Insulin resistance is associated with aging in mice and humans. We have previously shown that administration of recombinant GDF11 (rGDF11) to aged mice alters aging phenotypes in the brain, skeletal muscle, and heart. While the closely related protein GDF8 has a role in metabolism, limited data are available on the potential metabolic effects of GDF11 or GDF8 in aging. To determine the metabolic effects of these two ligands, we administered rGDF11 or rGDF8 protein to young or aged mice fed a standard chow diet, short-term high-fat diet (HFD), or long-term HFD. Under nearly all of these diet conditions, administration of exogenous rGDF11 reduced body weight by 3-17% and significantly improved glucose tolerance in aged mice fed a chow (~30% vs. saline) or HF (~50% vs. saline) diet and young mice fed a HFD (~30%). On the other hand, exogenous rGDF8 showed signifcantly lesser effect or no effect at all on glucose tolerance compared to rGDF11, consistent with data demonstrating that GFD11 is a more potent signaling ligand than GDF8. Collectively, our results show that administration of exogenous rGDF11, but not rGDF8, can reduce diet-induced weight gain and improve metabolic homeostasis.


Assuntos
Envelhecimento/metabolismo , Peso Corporal/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/administração & dosagem , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina , Miostatina/administração & dosagem , Envelhecimento/sangue , Envelhecimento/efeitos dos fármacos , Animais , Proteínas Morfogenéticas Ósseas/farmacologia , Metabolismo Energético/efeitos dos fármacos , Fatores de Diferenciação de Crescimento/administração & dosagem , Fatores de Diferenciação de Crescimento/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miostatina/farmacologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
5.
J Pharmacol Exp Ther ; 328(2): 496-503, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18974362

RESUMO

Aldose reductase (AR), an enzyme widely believed to be involved in the aberrant metabolism of glucose and development of diabetic complications, is expressed at low levels in the mouse. We studied whether expression of human AR (hAR), its inhibition with lidorestat, which is an AR inhibitor (ARI), and the presence of streptozotocin (STZ)-induced diabetes altered plasma fructose, mortality, and/or vascular lesions in low-density lipoprotein (LDL) receptor-deficient [Ldlr(-/-)] mice. Mice were made diabetic at 12 weeks of age with low-dose STZ treatment. Four weeks later, the diabetic animals (glucose > 20 mM) were blindly assigned to a 0.15% cholesterol diet with or without ARI. After 4 and 6 weeks, there were no significant differences in body weights or plasma cholesterol, triglyceride, and glucose levels between the groups. Diabetic Ldlr(-/-) mice receiving ARI had plasma fructose levels of 5.2 +/- 2.3 microg/ml; placebo-treated mice had plasma fructose levels of 12.08 +/- 7.4 microg/ml, p < 0.01, despite the induction of fructose-metabolizing enzymes, fructose kinase and adolase B. After 6 weeks, hAR/Ldlr(-/-) mice on the placebo-containing diet had greater mortality (31%, n = 9/26 versus 6%, n = 1/21, p < 0.05). The mortality rate in the ARI-treated group was similar to that in non-hAR-expressing mice. Therefore, diabetic hAR-expressing mice had increased fructose and greater mortality that was corrected by inclusion of lidorestat, an ARI, in the diet. If similar effects are found in humans, such treatment could improve clinical outcome in diabetic patients.


Assuntos
Aldeído Redutase/antagonistas & inibidores , Sangue/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Frutose/sangue , Ácidos Indolacéticos/farmacologia , Mortalidade , Tiazóis/farmacologia , Aldeído Redutase/metabolismo , Animais , Fenômenos Biológicos/efeitos dos fármacos , Sangue/metabolismo , Glicemia/fisiologia , Frutose/metabolismo , Humanos , Camundongos , Camundongos Knockout , Estreptozocina
6.
J Clin Invest ; 115(9): 2434-43, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127462

RESUMO

Direct evidence that hyperglycemia, rather than concomitant increases in known risk factors, induces atherosclerosis is lacking. Most diabetic mice do not exhibit a higher degree of atherosclerosis unless the development of diabetes is associated with more severe hyperlipidemia. We hypothesized that normal mice were deficient in a gene that accelerated atherosclerosis with diabetes. The gene encoding aldose reductase (AR), an enzyme that mediates the generation of toxic products from glucose, is expressed at low levels in murine compared with human tissues. Mice in which diabetes was induced through streptozotocin (STZ) treatment, but not nondiabetic mice, expressing human AR (hAR) crossed with LDL receptor-deficient (Ldlr-/-) C57BL/6 male mice had increased aortic atherosclerosis. Diabetic hAR-expressing heterozygous LDL receptor-knockout mice (Ldlr+/-) fed a cholesterol/cholic acid-containing diet also had increased aortic lesion size. Lesion area at the aortic root was increased by STZ treatment alone but was further increased by hAR expression. Macrophages from hAR-transgenic mice expressed more scavenger receptors and had greater accumulation of modified lipoproteins than macrophages from nontransgenic mice. Expression of genes that regulate regeneration of glutathione was reduced in the hAR-expressing aortas. Thus, hAR increases atherosclerosis in diabetic mice. Inhibitors of AR or other enzymes that mediate glucose toxicity could be useful in the treatment of diabetic atherosclerosis.


Assuntos
Aldeído Redutase/metabolismo , Aterosclerose , Complicações do Diabetes/fisiopatologia , Diabetes Mellitus Experimental , Aldeído Redutase/genética , Animais , Aorta/patologia , Aterosclerose/etiologia , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Glicemia/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Dieta Aterogênica , Humanos , Lipídeos/sangue , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Fatores de Risco
7.
Cancer Res ; 63(7): 1583-8, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12670908

RESUMO

Low or absent expression of the cyclin-dependent kinase inhibitor p27(Kip1) serves as an excellent malignant marker for prostate and other human cancers. The level of p27(Kip1) is regulated primarily by the ubiquitin E3 ligase SCF(SKP2) through ubiquitin-dependent proteolysis. Expression of the F-box protein SKP2 is inversely correlated with p27 in many cancers. To determine the role of SCF(SKP2) in proliferation and tumorigenesis, we established transgenic mouse lines that specifically expressed SKP2 in the prostate gland. Unscheduled expression of SKP2 promoted marked overproliferation, resulting in hyperplasia, dysplasia, and low-grade carcinoma in the prostate gland. Consistent with its critical role in p27 proteolysis, SKP2 expression caused significant down-regulation of p27 in prostate glands from transgenic animals. Immunohistological staining indicated that SKP2 expression is restricted to the hyperplastic/dysplastic regions and that there is an inverse relationship between SKP2 and p27 expression in the ductal epithelium in transgenic animals. The prostate glands from transgenic mice also exhibited high levels of proliferative and mitotic markers such as Ki67 and cyclin B1. Our data suggest that SKP2 acts as an oncoprotein in the mouse prostate gland, probably through its function as a limiting factor for ubiquitin-dependent degradation of p27.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Divisão Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p27 , Modelos Animais de Doenças , Regulação para Baixo , Masculino , Camundongos , Camundongos Transgênicos , Próstata/patologia , Hiperplasia Prostática/genética , Hiperplasia Prostática/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Quinases Associadas a Fase S , Proteínas Supressoras de Tumor/biossíntese
8.
J Clin Invest ; 126(4): 1401-12, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26974159

RESUMO

The phosphatidylinositol 3-kinase (PI3K) signaling pathway is central to the action of insulin and many growth factors. Heterozygous mutations in the gene encoding the p85α regulatory subunit of PI3K (PIK3R1) have been identified in patients with SHORT syndrome - a disorder characterized by short stature, partial lipodystrophy, and insulin resistance. Here, we evaluated whether SHORT syndrome-associated PIK3R1 mutations account for the pathophysiology that underlies the abnormalities by generating knockin mice that are heterozygous for the Pik3r1Arg649Trp mutation, which is homologous to the mutation found in the majority of affected individuals. Similar to the patients, mutant mice exhibited a reduction in body weight and length, partial lipodystrophy, and systemic insulin resistance. These derangements were associated with a reduced capacity of insulin and other growth factors to activate PI3K in liver, muscle, and fat; marked insulin resistance in liver and fat of mutation-harboring animals; and insulin resistance in vitro in cells derived from these mice. In addition, mutant mice displayed defective insulin secretion and GLP-1 action on islets in vivo and in vitro. These data demonstrate the ability of this heterozygous mutation to alter PI3K activity in vivo and the central role of PI3K in insulin/growth factor action, adipocyte function, and glucose metabolism.


Assuntos
Hormônio do Crescimento , Resistência à Insulina/genética , Fígado/enzimologia , Mutação de Sentido Incorreto , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Substituição de Aminoácidos , Animais , Peptídeo 1 Semelhante ao Glucagon/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Insulina/genética , Insulina/metabolismo , Secreção de Insulina , Lipodistrofia/enzimologia , Lipodistrofia/genética , Lipodistrofia/patologia , Fígado/patologia , Camundongos , Camundongos Mutantes
9.
J Clin Invest ; 126(11): 4372-4386, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27669460

RESUMO

Obese, insulin-resistant states are characterized by a paradoxical pathogenic condition in which the liver appears to be selectively insulin resistant. Specifically, insulin fails to suppress glucose production, yet successfully stimulates de novo lipogenesis. The mechanisms underlying this dysregulation remain controversial. Here, we hypothesized that carbohydrate-responsive element-binding protein (ChREBP), a transcriptional activator of glycolytic and lipogenic genes, plays a central role in this paradox. Administration of fructose increased hepatic hexose-phosphate levels, activated ChREBP, and caused glucose intolerance, hyperinsulinemia, hypertriglyceridemia, and hepatic steatosis in mice. Activation of ChREBP was required for the increased expression of glycolytic and lipogenic genes as well as glucose-6-phosphatase (G6pc) that was associated with the effects of fructose administration. We found that fructose-induced G6PC activity is a major determinant of hepatic glucose production and reduces hepatic glucose-6-phosphate levels to complete a homeostatic loop. Moreover, fructose activated ChREBP and induced G6pc in the absence of Foxo1a, indicating that carbohydrate-induced activation of ChREBP and G6PC dominates over the suppressive effects of insulin to enhance glucose production. This ChREBP/G6PC signaling axis is conserved in humans. Together, these findings support a carbohydrate-mediated, ChREBP-driven mechanism that contributes to hepatic insulin resistance.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Frutose/toxicidade , Glucose/biossíntese , Resistência à Insulina , Insulina/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Glucose/genética , Intolerância à Glucose/induzido quimicamente , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Glucose-6-Fosfatase/genética , Glucose-6-Fosfatase/metabolismo , Glicólise/efeitos dos fármacos , Glicólise/genética , Humanos , Insulina/genética , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética
10.
Nat Med ; 22(3): 312-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26808348

RESUMO

Uncoupling protein 1 (UCP1) is highly expressed in brown adipose tissue, where it generates heat by uncoupling electron transport from ATP production. UCP1 is also found outside classical brown adipose tissue depots, in adipocytes that are termed 'brite' (brown-in-white) or 'beige'. In humans, the presence of brite or beige (brite/beige) adipocytes is correlated with a lean, metabolically healthy phenotype, but whether a causal relationship exists is not clear. Here we report that human brite/beige adipocyte progenitors proliferate in response to pro-angiogenic factors, in association with expanding capillary networks. Adipocytes formed from these progenitors transform in response to adenylate cyclase activation from being UCP1 negative to being UCP1 positive, which is a defining feature of the beige/brite phenotype, while displaying uncoupled respiration. When implanted into normal chow-fed, or into high-fat diet (HFD)-fed, glucose-intolerant NOD-scid IL2rg(null) (NSG) mice, brite/beige adipocytes activated in vitro enhance systemic glucose tolerance. These adipocytes express neuroendocrine and secreted factors, including the pro-protein convertase PCSK1, which is strongly associated with human obesity. Pro-angiogenic conditions therefore drive the proliferation of human beige/brite adipocyte progenitors, and activated beige/brite adipocytes can affect systemic glucose homeostasis, potentially through a neuroendocrine mechanism.


Assuntos
Adipócitos/metabolismo , Glicemia/metabolismo , Intolerância à Glucose/metabolismo , Neovascularização Fisiológica , Consumo de Oxigênio , RNA Mensageiro/metabolismo , Adipócitos/transplante , Adipócitos Marrons/metabolismo , Adipócitos Marrons/transplante , Adipócitos Brancos/metabolismo , Adipócitos Brancos/transplante , Adulto , Idoso , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Capilares , Transplante de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dieta Hiperlipídica , Encefalinas/genética , Encefalinas/metabolismo , Feminino , Imunofluorescência , Técnica Clamp de Glucose , Teste de Tolerância a Glucose , Homeostase , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Interleucina-33/genética , Interleucina-33/metabolismo , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Obesidade/metabolismo , Reação em Cadeia da Polimerase , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 1/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Proteínas , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteína Desacopladora 1 , Iodotironina Desiodinase Tipo II
11.
Diabetes ; 52(8): 1910-7, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12882905

RESUMO

To investigate the physiological effects of modulating the abundance of Munc18c or syntaxin 4 (Syn4) proteins on the regulation of glucose homeostasis in vivo, we generated tetracycline-repressible transgenic mice that overexpress either Munc18c or Syn4 proteins in skeletal muscle, pancreas and adipose tissue seven-, five-, and threefold over endogenous protein, respectively. Munc18c transgenic mice displayed whole-body insulin resistance during hyperinsulinemic-euglycemic clamp resulting from >41% reductions in skeletal muscle and white adipose tissue glucose uptake, but without alteration of hepatic insulin action. Munc18c transgenic mice exhibited approximately 40% decreases in whole-body glycogen/lipid synthesis, skeletal muscle glycogen synthesis, and glycolysis. Glucose intolerance in Munc18c transgenic mice was reversed by repression of transgene expression using tetracycline or by simultaneous overexpression of Syn4 protein. In addition, Munc18c transgenic mice had depressed serum insulin levels, reflecting a threefold reduction in insulin secretion from islets isolated therefrom, thus uncovering roles for Munc18c and/or Syn4 in insulin granule exocytosis. Taken together, these results indicate that balance, more than absolute abundance, of Munc18c and Syn4 proteins directly affects whole-body glucose homeostasis through alterations in insulin secretion and insulin action.


Assuntos
Intolerância à Glucose/fisiopatologia , Resistência à Insulina/fisiologia , Proteínas do Tecido Nervoso , Proteínas/genética , Proteínas de Transporte Vesicular , Tecido Adiposo/metabolismo , Animais , Antibacterianos , Expressão Gênica/efeitos dos fármacos , Glucose/farmacocinética , Intolerância à Glucose/metabolismo , Homeostase/fisiologia , Insulina/metabolismo , Secreção de Insulina , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Munc18 , Músculo Esquelético/metabolismo , Proteínas/metabolismo , Proteínas Qa-SNARE , Tetraciclina
12.
Diabetes ; 53(4): 1060-7, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15047622

RESUMO

The circulating level of the inflammatory cytokine interleukin (IL)-6 is elevated in various insulin-resistant states including type 2 diabetes, obesity, cancer, and HIV-associated lipodystrophy. To determine the role of IL-6 in the development of insulin resistance, we examined the effects of IL-6 treatment on whole-body insulin action and glucose metabolism in vivo during hyperinsulinemic-euglycemic clamps in awake mice. Pretreatment of IL-6 blunted insulin's ability to suppress hepatic glucose production and insulin-stimulated insulin receptor substrate (IRS)-2-associated phosphatidylinositol (PI) 3-kinase activity in liver. Acute IL-6 treatment also reduced insulin-stimulated glucose uptake in skeletal muscle, and this was associated with defects in insulin-stimulated IRS-1-associated PI 3-kinase activity and increases in fatty acyl-CoA levels in skeletal muscle. In contrast, we found that co-treatment of IL-10, a predominantly anti-inflammatory cytokine, prevented IL-6-induced defects in hepatic insulin action and signaling activity. Additionally, IL-10 co-treatment protected skeletal muscle from IL-6 and lipid-induced defects in insulin action and signaling activity, and these effects were associated with decreases in intramuscular fatty acyl-CoA levels. This is the first study to demonstrate that inflammatory cytokines IL-6 and IL-10 alter hepatic and skeletal muscle insulin action in vivo, and the mechanism may involve cytokine-induced alteration in intracellular fat contents. These findings implicate an important role of inflammatory cytokines in the pathogenesis of insulin resistance.


Assuntos
Insulina/fisiologia , Interleucina-10/farmacologia , Interleucina-6/farmacologia , Fígado/fisiologia , Músculo Esquelético/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Técnica Clamp de Glucose , Hiperinsulinismo , Infusões Intravenosas , Lipídeos/administração & dosagem , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/efeitos dos fármacos
13.
Cell Metab ; 20(4): 593-602, 2014 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-25264246

RESUMO

Pancreatic ß cell failure in type 2 diabetes is associated with functional abnormalities of insulin secretion and deficits of ß cell mass. It's unclear how one begets the other. We have shown that loss of ß cell mass can be ascribed to impaired FoxO1 function in different models of diabetes. Here we show that ablation of the three FoxO genes (1, 3a, and 4) in mature ß cells results in early-onset, maturity-onset diabetes of the young (MODY)-like diabetes, with abnormalities of the MODY networks Hnf4α, Hnf1α, and Pdx1. FoxO-deficient ß cells are metabolically inflexible, i.e., they preferentially utilize lipids rather than carbohydrates as an energy source. This results in impaired ATP generation and reduced Ca(2+)-dependent insulin secretion. The present findings demonstrate a secretory defect caused by impaired FoxO activity that antedates dedifferentiation. We propose that defects in both pancreatic ß cell function and mass arise through FoxO-dependent mechanisms during diabetes progression.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Fatores de Transcrição Forkhead/genética , Insulina/metabolismo , Animais , Glicemia/análise , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Proteínas de Ciclo Celular , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/deficiência , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Transativadores/metabolismo
14.
Diabetes ; 57(6): 1674-82, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18346984

RESUMO

OBJECTIVE: Patients with diabetes often have dyslipidemia and increased postprandial lipidmia. Induction of diabetes in LDL receptor (Ldlr(-/-)) knockout mice also leads to marked dyslipidemia. The reasons for this are unclear. RESEARCH DESIGN AND METHODS: We placed Ldlr(-/-) and heterozygous LDL receptor knockout (Ldlr(+/-)) mice on a high-cholesterol (0.15%) diet, induced diabetes with streptozotocin (STZ), and assessed reasons for differences in plasma cholesterol. RESULTS: STZ-induced diabetic Ldlr(-/-) mice had plasma cholesterol levels more than double those of nondiabetic controls. Fast-performance liquid chromatography and ultracentrifugation showed an increase in both VLDL and LDL. Plasma VLDL became more cholesterol enriched, and both VLDL and LDL had a greater content of apolipoprotein (apo)E. In LDL the ratio of apoB48 to apoB100 was increased. ApoB production, assessed using [(35)S]methionine labeling in Triton WR1339-treated mice, was not increased in fasting STZ-induced diabetic mice. Similarly, postprandial lipoprotein production was not increased. Reduction of cholesterol in the diet to normalize the amount of cholesterol intake by the control and STZ-induced diabetic animals reduced plasma cholesterol levels in STZ-induced diabetic mice, but plasma cholesterol was still markedly elevated compared with nondiabetic controls. LDL from STZ-induced diabetic mice was cleared from the plasma and trapped more rapidly by livers of control mice. STZ treatment reduced liver expression of the proteoglycan sulfation enzyme, heparan sulfate N-deacetylase/N-sulfotrasferase-1, an effect that was reproduced in cultured hepatocytyes by a high glucose-containing medium. CONCLUSIONS: STZ-induced diabetic, cholesterol-fed mice developed hyperlipidemia due to a non-LDL receptor defect in clearance of circulating apoB-containing lipoproteins.


Assuntos
Colesterol/sangue , Diabetes Mellitus Experimental/sangue , Lipídeos/sangue , Lipoproteínas/sangue , Receptores de LDL/deficiência , Triglicerídeos/sangue , Animais , Apolipoproteínas B/sangue , Apolipoproteínas E/sangue , Glicemia/metabolismo , Colesterol na Dieta , Cruzamentos Genéticos , Diabetes Mellitus Experimental/fisiopatologia , Dislipidemias/genética , Fígado/fisiopatologia , Neoplasias Hepáticas , Neoplasias Hepáticas Experimentais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Tumorais Cultivadas
15.
J Lipid Res ; 49(10): 2101-12, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18515784

RESUMO

Ceramide is among a number of potential lipotoxic molecules that are thought to modulate cellular energy metabolism. The heart is one of the tissues thought to become dysfunctional due to excess lipid accumulation. Dilated lipotoxic cardiomyopathy, thought to be the result of diabetes and severe obesity, has been modeled in several genetically altered mice, including animals with cardiac-specific overexpression of glycosylphosphatidylinositol (GPI)-anchored human lipoprotein lipase (LpL(GPI)). To test whether excess ceramide was implicated in cardiac lipotoxicity, de novo ceramide biosynthesis was inhibited pharmacologically by myriocin and genetically by heterozygous deletion of LCB1, a subunit of serine palmitoyltransferase (SPT). Inhibition of SPT, a rate-limiting enzyme in ceramide biosynthesis, reduced fatty acid and increased glucose oxidation in isolated perfused LpL(GPI) hearts, improved systolic function, and prolonged survival rates. Our results suggest a critical role for ceramide accumulation in the pathogenesis of lipotoxic cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Cardiotoxinas/metabolismo , Ceramidas/metabolismo , Animais , Biomarcadores/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Cardiotoxinas/antagonistas & inibidores , Bovinos , Ceramidas/antagonistas & inibidores , Ácidos Graxos/metabolismo , Ácidos Graxos Monoinsaturados/farmacologia , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Glicosilfosfatidilinositóis/metabolismo , Coração/efeitos dos fármacos , Coração/fisiopatologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Lipase Lipoproteica/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oxirredução , Fosforilação/efeitos dos fármacos , Serina C-Palmitoiltransferase/antagonistas & inibidores , Serina C-Palmitoiltransferase/genética , Serina C-Palmitoiltransferase/metabolismo , Taxa de Sobrevida
16.
Cardiovasc Drugs Ther ; 20(6): 441-4, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17139480

RESUMO

INTRODUCTION: Most circulating fatty acids are contained in lipoprotein triglycerides. For the heart to acquire these lipids, they must be broken down into free fatty acids via the enzyme lipoprotein lipase (LpL). Although it has long been known that hearts primarily use esterified fatty acids as fuel, different sources of fatty acids were thought to be interchangeable. MATERIALS AND METHODS: By creating mice with neonatal and acute LpL deletion we showed that lipoprotein-derived fatty acids could not be replaced by albumin-associated free fatty acids. Loss of cardiac LpL forces the heart to increase its uptake of glucose, reduce fatty acid oxidation, and eventually leads to cardiac dysfunction. In contrast, cardiomyocyte specific overexpression of an anchored form of LpL leads to excess lipid uptake, induction of fatty acid oxidation genes, and dilated cardiomyopathy. CONCLUSION: Increasing lipid secretion from the heart or redirecting lipids to adipose tissue can alleviate this lipotoxic situation.


Assuntos
Lipólise/genética , Lipoproteínas/genética , Lipoproteínas/metabolismo , Miocárdio/metabolismo , Triglicerídeos/genética , Triglicerídeos/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Metabolismo Energético/genética , Ácidos Graxos/metabolismo , Predisposição Genética para Doença/genética , Humanos , Redes e Vias Metabólicas , Miócitos Cardíacos/metabolismo
17.
Am J Physiol Endocrinol Metab ; 291(4): E755-60, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16684851

RESUMO

The most energy-requiring organ in the body, the cardiac muscle, relies primarily on lipoprotein-derived fatty acids. Prenatal loss of cardiac lipoprotein lipase (LPL) leads to hypertriglyceridemia, but no cardiac dysfunction, in young mice. Cardiac specific loss of LPL in 8-wk-old mice was produced by a 2-wk tamoxifen treatment of MerCreMer (MCM)/Lpl(flox/flox) mice. LPL gene deletion was confirmed by PCR analysis, and LPL mRNA expression was reduced by approximately 70%. One week after tamoxifen was completed, triglyceride was increased with LPL deletion, 162 +/- 53 vs. 91 +/- 21 mg/dl, P < 0.01. Tamoxifen treatment of Lpl(flox/flox) mice did not cause a significant increase in triglyceride levels. Four weeks after tamoxifen, MCM/Lpl(flox/flox) mice had triglyceride levels of 190 +/- 27 mg/dl, similar to those of mice with prenatal LPL deletion. One week after the tamoxifen, MCM/Lpl(flox/flox), but not Lpl(flox/flox), mice had decreases in carnitine palmitoyl transferase I mRNA (18%) and pyruvate dehydrogenase kinase 4 mRNA (38%). These changes in gene expression became more robust with time. Acute loss of LPL decreased ejection fraction and increased mRNA levels for atrial natriuretic factor. Our studies show that acute loss of LPL can be produced and leads to rapid alteration in gene expression and cardiac dysfunction.


Assuntos
Dislipidemias/enzimologia , Cardiopatias/enzimologia , Lipase Lipoproteica/deficiência , Miocárdio/enzimologia , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Colesterol/sangue , Dislipidemias/sangue , Dislipidemias/metabolismo , Ecocardiografia , Antagonistas de Estrogênios/farmacologia , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Cardiopatias/sangue , Cardiopatias/genética , Cardiopatias/metabolismo , Integrases/genética , Integrases/metabolismo , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miocárdio/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Complexo Piruvato Desidrogenase/genética , Complexo Piruvato Desidrogenase/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Tamoxifeno/farmacologia , Triglicerídeos/sangue
18.
J Biol Chem ; 281(13): 8716-23, 2006 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-16410253

RESUMO

Long-chain fatty acids (FAs) are the predominant energy substrate utilized by the adult heart. The heart can utilize unesterified FA bound to albumin or FA obtained from lipolysis of lipoprotein-bound triglyceride (TG). We used heart-specific lipoprotein lipase knock-out mice (hLpL0) to test whether these two sources of FA are interchangeable and necessary for optimal heart function. Hearts unable to obtain FA from lipoprotein TG were able to compensate by increasing glucose uptake, glycolysis, and glucose oxidation. HLpL0 hearts had decreased expression of pyruvate dehydrogenase kinase 4 and increased cardiomyocyte expression of glucose transporter 4. Conversely, FA oxidation rates were reduced in isolated perfused hLpL0 hearts. Following abdominal aortic constriction expression levels of genes regulating FA and glucose metabolism were acutely up-regulated in control and hLpL0 mice, yet all hLpL0 mice died within 48 h of abdominal aortic constriction. Older hLpL0 mice developed cardiac dysfunction characterized by decreased fractional shortening and interstitial and perivascular fibrosis. HLpL0 hearts had increased expression of several genes associated with transforming growth factor-beta signaling. Thus, long term reduction of lipoprotein FA uptake is associated with impaired cardiac function despite a compensatory increase in glucose utilization.


Assuntos
Cardiomiopatias/metabolismo , Glucose/metabolismo , Lipase Lipoproteica/deficiência , Lipase Lipoproteica/fisiologia , Miocárdio/enzimologia , Trifosfato de Adenosina/análise , Animais , Glicemia/análise , Radioisótopos de Carbono , Cardiomiopatias/etiologia , Colesterol/sangue , Colesterol/metabolismo , Ecocardiografia , Jejum , Ácidos Graxos/metabolismo , Feminino , Expressão Gênica , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Glicólise , Cinética , Lipase Lipoproteica/análise , Lipase Lipoproteica/genética , Lipoproteínas VLDL/metabolismo , Masculino , Camundongos , Camundongos Knockout , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Oxirredução , Fosfocreatina/análogos & derivados , Fosfocreatina/análise , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Triglicerídeos/sangue
19.
J Autoimmun ; 19(1-2): 1-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12367554

RESUMO

Glutamic acid decarboxylase (GAD) is one major autoantigen involved in the pathogenesis of autoimmune insulin dependent diabetes mellitus (IDDM). Molecular mechanisms regulating GAD expression in pancreatic beta cell are still ill-defined. Here we investigated the effect of streptozotocin (STZ), a beta cell-specific toxin, on the expression of GAD67 in MIN6N8a mouse beta cell. A 5-6-fold increase in the expression GAD67 mRNA was found in cells treated with 1.25mM STZ for 12h. Addition of NAD+ to the incubation medium slightly reduced the STZ-induced upregulation of GAD67. STZ increased p53 levels that in turn up-modulated GAD67 expression. This effect was abolished upon addition of the antioxidant N-acetyl cysteine (NAC). STZ also activated NF-kappaB and blockade of NF-kappaB activation inhibited the STZ-mediated upregulation of GAD67 expression. As a whole these data show that low dose of STZ up-regulates GAD67 expression in mouse bate cell and that NF-kappaB activation through oxidative stress plays a key role in this phenomenon. They also suggest that various stimuli promoting NF-kappaB activation may up-regulate expression of GAD autoantigen in mouse beta cells.


Assuntos
Antibacterianos/farmacologia , Glutamato Descarboxilase/metabolismo , Ilhotas Pancreáticas/enzimologia , Isoenzimas/metabolismo , Estreptozocina/farmacologia , Animais , Antioxidantes/metabolismo , Autoantígenos/metabolismo , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/etiologia , Camundongos , NAD/metabolismo , NF-kappa B/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa