Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adipocyte ; 6(2): 154-160, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28425845

RESUMO

The regulation of adipose tissue expansion by adipocyte hypertrophy and/or hyperplasia is the topic of extensive investigations given the potential differential contribution of the 2 processes to the development of numerous chronic diseases associated with obesity. We recently discovered that the loss-of-function of the Src homology domain-containing protein Nck2 in mice promotes adiposity accompanied with adipocyte hypertrophy and impaired function, and enhanced adipocyte differentiation in vitro. Moreover, in severely-obese human's adipose tissue, we found that Nck2 expression is markedly downregulated. In this commentary, our goal is to expand upon additional findings providing further evidence for a unique Nck2-dependent mechanism regulating adipogenesis. We propose that Nck2 should be further investigated as a regulator of the reliance of white adipose tissue on hyperplasia versus hypertrophy during adipose tissue expansion, and hence, as a potential novel molecular target in obesity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adipogenia/fisiologia , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Adipogenia/genética , Tecido Adiposo/metabolismo , Tecido Adiposo Branco/metabolismo , Adiposidade , Animais , Regulação para Baixo , Hipertrofia/metabolismo , Camundongos , Obesidade/metabolismo
2.
Int J Obes (Lond) ; 40(6): 912-20, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26786352

RESUMO

BACKGROUND/OBJECTIVES: Adipose tissue (AT) autophagy gene expression is elevated in human obesity, correlating with increased metabolic risk, but mechanistic links between the two remain unclear. Thus, the objective of this study was to assess whether elevated autophagy may cause AT endocrine dysfunction, emphasizing the putative role of adiponectin in fat-liver endocrine communication. SUBJECTS/METHODS: We utilized a large (N=186) human AT biobank to assess clinical associations between human visceral AT autophagy genes, adiponectin and leptin, by multivariate models. A broader view of adipocytokines association with elevated autophagy was assessed using adipocytokine array. Finally, to establish causality, ex vivo studies utilizing a murine AT-hepatocyte cell line co-culture system was used. RESULTS: Circulating high-molecular-weight adiponectin and leptin levels were associated with human omental-AT expression of ATG5 mRNA, associations that remained significant (ß=-0.197, P=0.011; ß=0.267, P<0.001, respectively) in a multivariate model adjusted for age, sex, body mass index and interleukin-6 (IL-6). A similar association was observed with omental-AT LC3A mRNA levels. Bafilomycin-A1 (Baf A) pretreatment of AT explants from high-fat-fed (HFF) mice had no effect on the secretion of some AT-derived endocrine factors, but partially or fully reversed obesity-related changes in secretion of a subset of adipocytokines by >30%, including the obesity-associated upregulation of IL-6, vascular endothelial growth factor, tumor necrosis factor alpha (TNFα) and certain insulin-like growth factor-binding proteins, and the HFF-induced downregulated secretion of IL-10 and adiponectin. Similarly, decreased adiponectin and increased leptin secretion from cultured adipocytes stimulated with TNFα+IL-1ß was partially reversed by small interfering RNA-mediated knockdown of ATG7. AT explants from HFF mice co-cultured with Hepa1c hepatoma cells impaired insulin-induced Akt and GSK3 phosphorylation. This effect was significantly reversed by pretreating explants with Baf A, but not if adiponectin was immunodepleted from the conditioned media. CONCLUSIONS: Reduced secretion of adiponectin may link obesity-associated elevated AT autophagy/lysosomal activity with adipose endocrine dysfunction.


Assuntos
Adipócitos/metabolismo , Adiponectina/metabolismo , Tecido Adiposo/metabolismo , Autofagia , Glândulas Endócrinas/patologia , Doenças do Sistema Endócrino/patologia , Obesidade/fisiopatologia , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Técnicas de Cocultura , Modelos Animais de Doenças , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/patologia , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
3.
Mol Cell Endocrinol ; 409: 21-32, 2015 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-25818883

RESUMO

BACKGROUND: Pathophysiology of obesity is closely associated with enhanced autophagy in adipose tissue (AT). Autophagic process can promote survival or activate cell death. Therefore, we examine the occurrence of autophagy in AT of type 2 diabetes (T2D) patients in comparison to obese and lean individuals without diabetes. METHODOLOGY/PRINCIPAL FINDINGS: Numerous autophagosomes accumulated within adipocytes were visualized by electron transmission microscopy and by immunofluorescence staining for autophagy marker LC3 in obese and T2D patients. Increased autophagy was demonstrated by higher LC3-II/LC3-I ratio, up-regulated expression of LC3 and Atg5 mRNA, along with decreased p62 and mTOR protein levels. Increased autophagy occurred together with AT inflammation. CONCLUSIONS: Our data suggest fat depot-related differences in autophagy regulation. In subcutaneous AT, increased autophagy is accompanied by increased markers of apoptosis in patients with obesity independently of T2D. In contrast, in visceral AT only in T2D patients increased autophagy was related to higher markers of apoptosis.


Assuntos
Tecido Adiposo/metabolismo , Autofagia , Diabetes Mellitus Tipo 2/fisiopatologia , Gordura Intra-Abdominal/patologia , Obesidade/fisiopatologia , Gordura Subcutânea/patologia , Tecido Adiposo/ultraestrutura , Apoptose , Proteína 5 Relacionada à Autofagia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Regulação da Expressão Gênica , Humanos , Gordura Intra-Abdominal/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Gordura Subcutânea/metabolismo , Serina-Treonina Quinases TOR/metabolismo
4.
Obes Rev ; 15(4): 322-37, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24251825

RESUMO

While obesity is clearly accepted as a major risk factor for cardio-metabolic morbidity, it is also apparent that some obese patients largely escape this association, forming a unique obese subphenotype(s). Current approaches to define such subphenotypes include clinical biomarkers that largely reflect already manifested comorbidities, such as markers of dyslipidaemia, hyperglycaemia and impaired regulation of vascular tone, and anthropometric or imaging-based assessment of adipose tissue distribution. Low-grade inflammation, evident both systemically and within adipose tissue (particularly intra-abdominal fat depots), seems to characterize the more cardio-metabolically morbid forms of obesity. Indeed, several systemic inflammatory markers (C-reactive protein), adipokines (retinol-binding protein 4, adiponectin) and cytokines have been shown to correlate in humans with adipose tissue inflammation and with obesity-associated health risks. Circulating leucocytes constitute a diverse group of cells that form a major arm of the immune system. They are both major sources of cytokines and likely also of infiltrating adipose tissue immune cells in obesity. In the present review, we summarize currently available literature on 'classical' blood white cell classes and on more specific leucocyte subclasses present in the circulation in human obesity. We critically raise the possibility that leucocytes may constitute clinically available markers for the more morbidity-associated obesity subphenotype(s), and when available, for intra-abdominal adipose tissue inflammation.


Assuntos
Tecido Adiposo/metabolismo , Inflamação/metabolismo , Leucócitos/metabolismo , Obesidade/sangue , Tecido Adiposo/imunologia , Linfócitos B , Biomarcadores/metabolismo , Distribuição da Gordura Corporal , Humanos , Inflamação/imunologia , Células Matadoras Naturais , Subpopulações de Linfócitos , Obesidade/imunologia , Fenótipo , Valor Preditivo dos Testes , Prognóstico , Linfócitos T
5.
Int J Obes (Lond) ; 35(6): 785-792, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21042325

RESUMO

AIMS: Over 75% of obese subjects fail to maintain their weight following weight loss interventions. We aimed to identify phenotypic and genetic markers associated with weight maintenance/regain following a dietary intervention. SUBJECTS AND METHODS: In the 2-year Dietary Intervention Randomized Controlled Trial, we assessed potential predictors for weight changes during the 'weight loss phase' (0-6 months) and the 'weight maintenance/regain phase' (7-24 months). Genetic variation between study participants was studied using single-nucleotide polymorphisms in the leptin gene (LEP). RESULTS: Mean weight reduction was -5.5% after 6 months, with a mean weight regain of 1.2% of baseline weight during the subsequent 7-24 months. In a multivariate regression model, higher baseline high-molecular-weight adiponectin was the only biomarker predictor of greater success in 0- to 6-month weight loss (ß = -0.222, P-value = 0.044). In a multivariate regression model adjusted for 6-month changes in weight and various biomarkers, 6-month plasma leptin reduction exhibited the strongest positive association with 6-month weight loss (ß = 0.505, P-value < 0.001). Conversely, 6-month plasma leptin reduction independently predicted weight regain during the following 18 months (ß = -0.131, P-value < 0.013). Weight regain was higher among participants who had a greater (top tertiles) 6-month decrease in both weight and leptin (+3.4% (95% confidence interval 2.1-4.8)) as compared with those in the lowest combined tertiles (+0.2% (95% confidence interval -1.1 to 1.4)); P-value < 0.001. Weight regain was further significantly and independently associated with genetic variations in LEP (P = 0.006 for both rs4731426 and rs2071045). Adding genetic data to the phenotypic multivariate model increased its predictive value for weight regain by 34%. CONCLUSION: Although greater reduction in leptin concentrations during the initial phase of a dietary intervention is associated with greater weight loss in the short term, plasma leptin reduction, combined with the degree of initial weight loss and with genetic variations in the LEP gene, constitutes a significant predictor of subsequent long-term weight regain.


Assuntos
Leptina/genética , Obesidade/genética , Aumento de Peso/genética , Biomarcadores/metabolismo , Índice de Massa Corporal , Dieta Redutora/métodos , Feminino , Variação Genética , Humanos , Leptina/sangue , Masculino , Pessoa de Meia-Idade , Obesidade/metabolismo , Fenótipo , Aumento de Peso/fisiologia
6.
Diabetologia ; 50(4): 833-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17334653

RESUMO

AIMS/HYPOTHESIS: The association between increased (visceral) fat mass, insulin resistance and type 2 diabetes mellitus is well known. Yet, it is unclear whether the mere increase in intra-abdominal fat mass, or rather functional alterations in fat tissue in obesity contribute to the development of insulin resistance in obese patients. Here we attempted to isolate the metabolic effect of increased fat mass by fat tissue transplantation. METHODS: Epididymal fat pads were removed from male C57Bl6/J mice and transplanted intraperitoneally into male littermates (recipients), increasing the combined perigonadal fat mass by 50% (p < 0.005). At 4 and 8 weeks post-transplantation, glucose and insulin tolerance tests were performed, and insulin, NEFA and adipokines measured. RESULTS: Circulating levels of NEFA, adiponectin and leptin were not significantly different between transplanted and sham-operated control mice, while results of the postprandial insulin tolerance test were similar between the two groups. In contrast, under fasting conditions, the mere increase in intra-abdominal fat mass resulted in decreased plasma glucose levels (6.9 +/- 0.4 vs 8.1 +/- 0.3 mmol/l, p = 0.03) and a approximately 20% lower AUC in the glucose tolerance test (p = 0.02) in transplanted mice. Homeostasis model assessment of insulin resistance (HOMA-IR) was 4.1 +/- 0.4 in transplanted mice (vs 6.2 +/- 0.7 in sham-operated controls) (p = 0.02), suggesting improved insulin sensitivity. Linear regression modelling revealed that while total body weight positively correlated, as expected, with HOMA-IR (beta: 0.728, p = 0.006), higher transplanted fat mass correlated with lower HOMA-IR (beta: -0.505, p = 0.031). CONCLUSIONS/INTERPRETATION: Increasing intra-abdominal fat mass by transplantation of fat from normal mice improved, rather than impaired, fasting glucose tolerance and insulin sensitivity, achieving an effect opposite to the expected metabolic consequence of increased visceral fat in obesity.


Assuntos
Tecido Adiposo/metabolismo , Epididimo/metabolismo , Glucose/metabolismo , Tecido Adiposo/patologia , Animais , Área Sob a Curva , Teste de Tolerância a Glucose , Insulina/metabolismo , Resistência à Insulina , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade , Fatores de Tempo , Transplante de Tecidos
7.
Diabetologia ; 49(10): 2463-73, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16896943

RESUMO

AIMS/HYPOTHESIS: Induction of stress kinases leading to serine hyperphosphorylation of IRS1 may link oxidative stress to insulin resistance. The aim of this study was to investigate the roles of the phosphorylated serine residues Ser307 and Ser632, two sites implicated in the inhibition of IRS1 function in insulin signalling. MATERIALS AND METHODS: Fao hepatoma cells were exposed to an H(2)O(2)-generating system, and antibodies against the two phosphorylated serine residues were used for immunoprecipitation, immunoblot and immunofluorescence analyses. RESULTS: Exposure to approximately 50 mumol/l H(2)O(2) for 2 h resulted in IRS1 phosphorylation on both Ser307 and Ser632, concomitant with activation of inhibitor kappa kinase beta (IKKbeta) and c-Jun kinase (JNK). Immunoprecipitation studies revealed that the maximum overlap between phospho (p) Ser307-IRS1 and pSer632-IRS1 was 20%, and confocal microscopy suggested distinct localisations of IRS1 molecules phosphorylated on either site. Although pSer307-IRS1 showed decreased insulin-induced tyrosine phosphorylation and interaction with phosphatidylinositol 3-kinase (PI3K) in response to insulin, pSer632-IRS1 molecules were normally tyrosine-phosphorylated and exhibited typical associated PI3K activity. Salicylic acid and SP600125 partially inhibited IKKbeta and JNK, respectively, which indicated distinct roles for these two kinases in the phosphorylation of IRS1 at the two serine sites. Protection against oxidation-mediated impairment in insulin-induced phosphorylation of protein kinase B/Akt and in glycogen synthesis was achieved only by combining salicylic acid and SP600125. CONCLUSIONS/INTERPRETATION: These results suggest that pSer307-IRS1 and pSer632-IRS1 may define two minimally overlapping pools of IRS1 in response to oxidative stress, contributing differentially to insulin resistance. A combination of stress kinase inhibitors is required to protect against insulin resistance and IRS1 hyperphosphorylation induced by oxidative stress.


Assuntos
Resistência à Insulina/fisiologia , Estresse Oxidativo/fisiologia , Fosfoproteínas/metabolismo , Fosfosserina/metabolismo , Animais , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/farmacologia , Proteínas Substratos do Receptor de Insulina , Neoplasias Hepáticas , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
8.
Diabetologia ; 48(5): 954-66, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15834551

RESUMO

AIMS/HYPOTHESIS: Troglitazone was the first thiazolidinedione (TZD) approved for clinical use, exerting hypoglycaemic effects related to its action as a ligand of the peroxisome proliferator-activated receptor gamma receptor in adipocytes. However, emerging evidence suggests that mitochondrial function may be affected by troglitazone, and that skeletal muscle cells acutely respond to troglitazone by enhancing glucose uptake. The aim of the present study was to determine the cellular mechanisms by which troglitazone acutely stimulates glucose utilisation in skeletal muscle cells. METHODS: L6 cells overexpressing GLUT4myc were incubated with troglitazone. Glucose uptake, transport and phosphorylation as well as AMP-activated protein kinase (AMPK) signalling and insulin signalling were examined. Changes in mitochondrial membrane potential were measured using the J-aggregate-forming dye JC-1. AMPK signalling was interfered with using AMPK alpha1/alpha2 siRNA. RESULTS: Troglitazone acutely (in 10 min) reduced the mitochondrial membrane potential in L6GLUT4myc myotubes and robustly stimulated AMPK activity. Following 30 min of incubation with troglitazone or insulin, 2-deoxyglucose uptake was stimulated 1.5- and 2.1-fold respectively, and in cells treated with troglitazone, a 1.8-fold increase in the 2-deoxyglucose-6-phosphate:2-deoxyglucose ratio was observed. Moreover, contrary to insulin, troglitazone did not significantly stimulate 3-O-methylglucose uptake. Unlike insulin, troglitazone did not increase surface GLUT4myc content and did not increase IRS1-associated phosphatidylinositol 3-kinase activity or Akt phosphorylation on T308 and S473. Interestingly, interfering with troglitazone-induced activation of AMPK by decreasing the expression of the enzyme using siRNA inhibited the stimulation of 2-deoxyglucose uptake by the TZD. CONCLUSIONS/INTERPRETATION: We propose that troglitazone acutely increases glucose flux in muscle via an AMPK-mediated increase in glucose phosphorylation.


Assuntos
Cromanos/farmacologia , Glucose/metabolismo , Hipoglicemiantes/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias Musculares/fisiologia , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Tiazolidinedionas/farmacologia , Proteínas Quinases Ativadas por AMP , Animais , Diferenciação Celular , Células Cultivadas , Insulina/farmacologia , Células L , Potenciais da Membrana/fisiologia , Camundongos , Mitocôndrias Musculares/efeitos dos fármacos , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Troglitazona
9.
Acta Physiol Scand ; 183(1): 75-88, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15654921

RESUMO

Accumulating clinical evidence now links HIV protease inhibitors (HPIs) to the pathogenesis of insulin resistance, dyslipidaemia, lipodystrophy and atherosclerosis associated with highly active anti-retroviral therapy. Here we briefly describe the evidence for a distinct causative role for HPIs, and explore the cellular mechanisms proposed to underlie these side-effects. Acute inhibition of GLUT4-mediated glucose transport, and defective insulin signalling induced by chronic exposure to nelfinavir, are described as cellular mechanisms of insulin resistance. Interference with adipogenesis and adipocyte apoptosis and nelfinavir-induced activation of lipolysis are discussed as potential mechanisms of HPI-induced lipodystrophy. HPI-induced free radical production, apoptosis and increased glucose utilization in vascular smooth muscle cells are presented as possible novel mechanisms for atherosclerosis. Common pathways and cause-effect relationships between the various cellular mechanisms presented are then discussed, with emphasis on the role of insulin resistance, free radical production and enhanced lipolysis. Understanding the cellular mechanisms of HPI-induced side-effects will enhance the search for improved anti-retroviral therapy, and may also shed light on the pathogenesis of common forms of insulin resistance, dyslipidaemia and atherosclerosis.


Assuntos
Arteriosclerose/induzido quimicamente , Inibidores da Protease de HIV/efeitos adversos , Resistência à Insulina/fisiologia , Lipodistrofia/induzido quimicamente , Adipócitos/metabolismo , Terapia Antirretroviral de Alta Atividade/efeitos adversos , Apoptose/fisiologia , Ácidos Graxos não Esterificados/metabolismo , Transportador de Glucose Tipo 4 , Humanos , Insulina/metabolismo , Lipólise/fisiologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia
10.
Diabetologia ; 47(6): 1107-17, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15168016

RESUMO

AIMS/HYPOTHESIS: Chronic exposure of 3T3-L1 adipocytes to the HIV protease inhibitor nelfinavir induces insulin resistance, recapitulating key metabolic alterations of adipose tissue in the lipodystrophy syndrome induced by these agents. Our goal was to identify the defect in the insulin signal transduction cascade leading to nelfinavir-induced insulin resistance. METHODS: Fully differentiated 3T3-L1 adipocytes were exposed to 30 micro mol/l nelfinavir for 18 h, after which the amount, the phosphorylation and the localisation of key proteins in the insulin signalling cascade were evaluated. RESULTS: Insulin-induced interaction of phosphatidylinositol 3'-kinase (PI 3-kinase) with IRS proteins was normal in cells treated with nelfinavir, as was IRS-1-associated PI 3-kinase activity. Yet insulin-induced phosphorylation of Akt/protein kinase B (PKB), p70S6 kinase and extracellular signal-regulated kinase 1/2 was significantly impaired. This could not be attributed to increased protein phosphatase 2A activity or to increased expression of phosphoinositide phosphatases (SHIP2 or PTEN). However, insulin failed to induce translocation of the PI 3-kinase effectors Akt/PKB and protein kinase C-zeta (PKC-zeta) to plasma membrane fractions of nelfinavir-treated adipocytes. CONCLUSIONS/INTERPRETATION: We therefore conclude that nelfinavir induces a defect in the insulin signalling cascade downstream of the activation of PI 3-kinase. This defect manifests itself by impaired insulin-mediated recruitment of Akt/PKB and PKC-zeta to the plasma membrane.


Assuntos
Membrana Celular/metabolismo , Resistência à Insulina , Nelfinavir/efeitos adversos , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Células 3T3-L1 , Animais , Membrana Celular/patologia , Desoxiglucose/antagonistas & inibidores , Desoxiglucose/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Glucose/metabolismo , Japão , Camundongos , Fosfatidilinositóis/química , Fosfatidilinositóis/genética , Fosfatidilinositóis/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosfoproteínas Fosfatases/química , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Fosfatase 2 , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases S6 Ribossômicas 70-kDa/química , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
11.
Acta Physiol Scand ; 178(4): 297-308, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12864734

RESUMO

AIMS: Understanding the mechanisms by which insulin regulates glucose transporter 4 (GLUT4) traffic in skeletal muscle has been a major challenge since the discoveries of glucose transporter's translocation and the cloning of GLUT4. Here we summarize our work of the past 5 years on the regulation of GLUT4 traffic in skeletal muscle cells. METHODS: L6 cells overexpressing GLUT4 harbouring an exofacial myc epitope gave us the opportunity to perform dynamic assessments of GLUT4 exocytosis, endocytosis, as well as a means to follow GLUT4 molecules along their journey through intracellular compartments. RESULTS: We found that insulin stimulation, which results in the expected gain in surface GLUT4, is mostly attributed to enhanced GLUT4 exocytosis, and does not significantly affect the initial rate of internalization. Two mechanisms by which insulin enhances GLUT4 exocytosis are described: 'Pull' relates to actin remodelling-based segregation of the insulin signalling molecules and the directed recruitment of GLUT4/VAMP2 containing vesicles. 'Push' is the accelerated inter-endosomal transit of endocytosed GLUT4 molecules through the recycling endosome. The interface between the two types of regulatory input by insulin is suggested to be the budding of GLUT4 from the transferrin receptor (TfR)-containing, recycling endosome. CONCLUSIONS: We propose a model on the identity of the GLUT4 pools responsible for GLUT4 recruitment to the plasma membrane in the basal state, or following insulin or hyperosmolarity stimuli.


Assuntos
Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Glucose/metabolismo , Transportador de Glucose Tipo 4 , Humanos , Hipoglicemiantes/farmacologia , Indinavir/farmacologia , Insulina/farmacologia , Proteínas de Membrana/metabolismo , Camundongos , Microscopia Confocal , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efeitos dos fármacos , Proteínas R-SNARE
12.
Diabetologia ; 46(5): 639-48, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12750770

RESUMO

AIM/HYPOTHESIS: Oxidative stress was shown to selectively induce impaired metabolic response to insulin, raising the possible involvement of alterations in Insulin-Receptor-Substrate (IRS) proteins. This study was conducted to assess whether oxidative stress induced IRS protein degradation and enhanced serine phosphorylation, and to assess their functional importance. METHODS: 3T3-L1 adipocytes and rat hepatoma cells (FAO) were exposed to micro-molar H(2)O(2) by adding glucose oxidase to the culture medium, and IRS1 content, its serine phosphorylation and downstream metabolic insulin effects were measured. RESULTS: Cells exposed to oxidative stress exhibited decreased IRS1 (but not IRS2) content, and increased serine phosphorylation of both proteins. Total protein ubiquitination was increased in oxidized cells, but not in cells exposed to prolonged insulin treatment. Yet, lactacystin and MG132, two unrelated proteasome inhibitors, prevented IRS1 degradation induced by prolonged insulin but not by oxidative stress. The PI 3-kinase inhibitor LY294002 and the mTOR inhibitor rapamycin, but not the MEK1 inhibitor PD98059, could prevent IRS1 changes in oxidized cells. Rapamycin, which protected against IRS1 degradation and serine phosphorylation was not associated with improved response to acute insulin stimulation. Moreover, the antioxidant alpha lipoic acid, while protecting against oxidative stress-induced insulin resistance in 3T3-L1 adipocytes, could not prevent IRS1 degradation and serine phosphorylation. CONCLUSION/INTERPRETATION: Oxidative stress induces serine phosphorylation of IRS1 and increases its degradation by a proteasome-independent pathway; yet, these changes do not correlate with the induction of impaired metabolic response to insulin.


Assuntos
Peróxido de Hidrogênio/farmacologia , Resistência à Insulina/fisiologia , Estresse Oxidativo/fisiologia , Fosfoproteínas/metabolismo , Fosfosserina/metabolismo , Células 3T3 , Animais , Antioxidantes/farmacologia , Biomarcadores , Cicloeximida/farmacologia , Cisteína Endopeptidases/metabolismo , Glucose Oxidase/metabolismo , Proteínas Substratos do Receptor de Insulina , Camundongos , Complexos Multienzimáticos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Complexo de Endopeptidases do Proteassoma , Proteínas/metabolismo , Serina/metabolismo , Ácido Tióctico/farmacologia , Ubiquitina/metabolismo
13.
Diabetologia ; 46(5): 649-58, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12712244

RESUMO

AIMS/HYPOTHESIS: Insulin-dependent glucose influx in skeletal muscle and adipocytes is believed to rely largely on GLUT4, but this has not been confirmed directly. We assessed the relative functional contribution of GLUT4 in experimental models of skeletal muscle and adipocytes using the HIV-1 protease inhibitor indinavir. METHODS: Indinavir (up to 100 micro mol/l) was added to the glucose transport solution after insulin stimulation of wild-type L6 muscle cells, L6 cells over-expressing either GLUT4myc or GLUT1myc, 3T3-L1 adipocytes, isolated mouse brown or white adipocytes, and isolated mouse muscle preparations. RESULTS: 100 micro mol/l indinavir inhibited 80% of both basal and insulin-stimulated 2-deoxyglucose uptake in L6GLUT4myc myotubes and myoblasts, but only 25% in L6GLUT1myc cells. Cell-surface density of glucose transporters was not affected. In isolated soleus and extensor digitorum longus muscles, primary white and brown adipocytes, insulin-stimulated glucose uptake was inhibited 70 to 80% by indinavir. The effect of indinavir on glucose uptake was variable in 3T3-L1 adipocytes, averaging 45% and 67% inhibition of basal and maximally insulin-stimulated glucose uptake, respectively. In this cell, fractional inhibition of glucose uptake by indinavir correlated positively with the fold-stimulation of glucose uptake by insulin, and was higher with sub-maximal insulin concentrations. The latter finding coincided with an increase only in GLUT4, but not GLUT1, in plasma membrane lawns. CONCLUSION/INTERPRETATION: Indinavir is a useful tool to assess different functional contributions of GLUT4 to glucose uptake in common models of skeletal muscle and adipocytes.


Assuntos
Adipócitos/metabolismo , Glucose/metabolismo , Indinavir/farmacologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , Células 3T3 , Adipócitos/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Membrana Celular/metabolismo , Transportador de Glucose Tipo 1 , Transportador de Glucose Tipo 4 , Inibidores da Protease de HIV/farmacologia , Humanos , Insulina/farmacologia , Camundongos , Proteínas de Transporte de Monossacarídeos/efeitos dos fármacos , Proteínas de Transporte de Monossacarídeos/genética , Músculo Esquelético/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo
15.
Acad Med ; 76(10): 1072-5, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11597853

RESUMO

Integrating clinical and basic sciences throughout the medical school curriculum has become a major objective of various innovations in medical education. While early clinical exposure has evolved as an efficient means of introducing clinical studies in the preclinical years, interdisciplinary integration of basic sciences during the clinical years remains a challenge. The authors describe their three years of experience with an interdisciplinary course designed to demonstrate the continuum of medical information from the clinic to the basic sciences. In this course, sixth-year medical students are required to choose one of three to four different one-week programs, each of which requires them to conduct an in-depth investigation of a defined clinical topic. Program coordinators are encouraged to work in clinician-basic scientist teams and to use a variety of teaching methods, with an emphasis on tutored individual and group learning based on critical readings of original papers. Coordinators are also encouraged to enable graduate research students to participate. From 1998 to 2000, students participated in nine programs, seven of which were coordinated by interdisciplinary teams. Several clinical and basic science disciplines were represented in each program, and various teaching methods were used. Graduate students participated in two of the programs. Evaluation of the programs (a debriefing discussion as well as short written evaluations) indicated moderate to good achievement of the course objectives.


Assuntos
Currículo , Educação de Pós-Graduação , Educação Médica , Ciência/educação , Avaliação Educacional , Israel , Seleção de Pessoal , Ensino/métodos
16.
Diabetes ; 50(6): 1425-31, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11375344

RESUMO

HIV protease inhibitors (HPIs) are potent antiretroviral agents clinically used in the management of HIV infection. Recently, HPI therapy has been linked to the development of a metabolic syndrome in which adipocyte insulin resistance appears to play a major role. In this study, we assessed the effect of nelfinavir on glucose uptake and lipolysis in differentiated 3T3-L1 adipocytes. An 18-h exposure to nelfinavir resulted in an impaired insulin-stimulated glucose uptake and activation of basal lipolysis. Impaired insulin stimulation of glucose up take occurred at nelfinavir concentrations >10 micromol/l (EC(50) = 20 micromol/l) and could be attributed to impaired GLUT4 translocation. Basal glycerol and free fatty acid (FFA) release were significantly enhanced with as low as 5 micromol/l nelfinavir, displaying fivefold stimulation of FFA release at 10 micromol/l. Yet, the antilipolytic action of insulin was preserved at this concentration. Potential underlying mechanisms for these metabolic effects included both impaired insulin stimulation of protein kinase B Ser 473 phosphorylation with preserved insulin receptor substrate tyrosine phosphorylation and decreased expression of the lipolysis regulator perilipin. Troglitazone pre- and cotreatment with nelfinavir partly protected the cells from the increase in basal lipolysis, but it had no effect on the impairment in insulin-stimulated glucose uptake induced by this HPI. This study demonstrates that nelfinavir induces insulin resistance and activates basal lipolysis in differentiated 3T3-L1 adipocytes, providing potential cellular mechanisms that may contribute to altered adipocyte metabolism in treated HIV patients.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/fisiologia , Inibidores da Protease de HIV/farmacologia , Resistência à Insulina , Lipólise/efeitos dos fármacos , Proteínas Musculares , Nelfinavir/farmacologia , Proteínas Serina-Treonina Quinases , Células 3T3 , Animais , Transporte Biológico/efeitos dos fármacos , Glucose/metabolismo , Transportador de Glucose Tipo 4 , Camundongos , Proteínas de Transporte de Monossacarídeos/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt
17.
Biochem J ; 355(Pt 3): 757-63, 2001 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11311139

RESUMO

Activation of phosphatidylinositol 3-kinase (PI 3-kinase) is a common event in both insulin and platelet-derived growth factor (PDGF) signalling, but only insulin activates this enzyme in the high-speed pellet (HSP), and induces GLUT4 translocation. Recently, we have demonstrated that exposure of 3T3-L1 adipocytes to oxidative stress impairs insulin-stimulated GLUT4 translocation and glucose transport, associated with impaired PI 3-kinase translocation and activation in the HSP [Tirosh, Potashnik, Bashan and Rudich (1999) J. Biol. Chem. 274, 10595-10602]. In this study the effect of a 2 h exposure to approximately 30 microM H(2)O(2) on insulin versus PDGF-BB signalling and metabolic effects was compared. PDGF-stimulated p85-associated PI 3-kinase activity in total cell lysates, as well as co-precipitation of the PDGF receptor, were unaffected by oxidative stress. Additionally, the increase in p85 association with the plasma-membrane lawns by PDGF remained intact following oxidation, whereas the insulin effect was decreased. PDGF significantly increased protein kinase B (PKB) activity in early differentiated cells, and that of p70 S6-kinase in both early and fully differentiated 3T3-L1 adipocytes. Following oxidation the effect of PDGF on PKB and p70 S6-kinase activation remained intact, whereas significant inhibition of insulin-stimulated activation of those enzymes was observed. In accordance, in both early and fully differentiated cells, oxidative stress completely blunted insulin- but not PDGF-stimulated protein synthesis. In conclusion, oxidative stress impairs insulin, but not PDGF, signalling and metabolic actions in both early and fully differentiated 3T3-L1 adipocytes. This emphasizes compartment-specific activation of PI 3-kinase as an oxidation-sensitive step specifically leading to insulin resistance.


Assuntos
Adipócitos/metabolismo , Insulina/metabolismo , Estresse Oxidativo/fisiologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Serina-Treonina Quinases , Transdução de Sinais/fisiologia , Células 3T3 , Adipócitos/enzimologia , Animais , Ativação Enzimática , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases S6 Ribossômicas/metabolismo , Frações Subcelulares/metabolismo
18.
Diabetologia ; 44(1): 55-62, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11206412

RESUMO

AIMS/HYPOTHESIS: Adipose tissue lipolysis plays a central part in total body fuel metabolism. Our study was to assess the long-term regulation of glycerol and non-esterified fatty acid (NEFA) release by insulin or TNF-alpha. METHODS: Fully differentiated 3T3-L1 adipocytes were exposed for up to 22 h to insulin or TNF-alpha. RESULTS: Long-term insulin treatment resulted in increased basal glycerol release, reaching sixfold at 22 h with 1 nmol/l insulin. Partial inhibition was observed by pharmacologically inhibiting phosphatidylinositol 3-kinase or the mitogen-activated kinase kinase--extracellular signal-regulated kinase cascades. This represented 50-60% of the response induced by 1 nmol/l TNF-alpha and approximately 40 % of the glycerol release maximally stimulated by isoproterenol (1 micromol/l, 30 min). The cellular mechanism seemed to be distinct from that of TNF-alpha: First, glycerol release in response to long-term insulin was progressive with time and did not display a lag-time characteristic of the effect of TNF-alpha. Second, pretreatment and co-treatment of the cells with troglitazone greatly inhibited TNF-alpha-induced glycerol release (128.5 +/- 10.2 to 35.4 +/- 2.1 nmol/mg protein per h) but not the effect of insulin, which was exaggerated. Third, hormone-sensitive lipase protein content was decreased (45 %) by TNF-alpha but not following long-term insulin. Finally, TNF-alpha was associated with NEFA release to the medium, whereas long-term insulin treatment was not. Moreover, glycerol release during isoproterenol-stimulated lipolysis was additive to the effect of long-term insulin, whereas NEFA release was inhibited by nearly 90 %. CONCLUSIONS INTERPRETATION: Contradictory to its short-term inhibitory effect, long-term insulin stimulates glycerol release with concomitant stimulation of NEFA re-esterification.


Assuntos
Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Glicerol/metabolismo , Insulina/farmacologia , Proteínas Musculares , Fator de Necrose Tumoral alfa/farmacologia , Células 3T3 , Agonistas Adrenérgicos beta/farmacologia , Animais , Glicemia/metabolismo , Membrana Celular/química , Cromonas/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Transportador de Glucose Tipo 4 , Insulina/administração & dosagem , Proteínas Substratos do Receptor de Insulina , Cinética , Lipólise/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas de Transporte de Monossacarídeos/análise , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosfoproteínas/análise , Tiazóis/farmacologia , Fator de Necrose Tumoral alfa/administração & dosagem
19.
Diabetologia ; 44(12): 2156-64, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11793016

RESUMO

AIMS/HYPOTHESIS: Substantial evidence suggests an important role for the expression of GLUT4 in adipocytes, in the pathogenesis of insulin resistance and Type II (non-insulin-dependent) diabetes mellitus. We investigated whether oxidative stress decreases GLUT4 expression by impairing DNA binding of nuclear proteins to the insulin responsive element in the GLUT4 promoter. METHODS: 3T3-L1 adipocytes were exposed to micromolar H2O2 concentrations and GLUT4 expression and binding of nuclear proteins to defined DNA sequences were assessed. RESULTS: GLUT4 mRNA was decreased after at least 4 h exposure to H2O2, without a major change in the stability of GLUT4 transcripts. Nuclear protein extracts prepared from oxidized cells showed decreased binding to the insulin responsive element of the GLUT4 promoter but not to other DNA sequences. The direct effect of oxidation on the binding to the insulin response element was shown by the observation that in vitro oxidation of nuclear extracts with H2O2, n-ethylmaleimide or diamide decreased protein-DNA complex formation. This, and decreased binding capacity observed in nuclear extracts from oxidized cells, were partly reversible by subsequent treatment with a reducing agent. Protein binding to a consensus DNA sequence for nuclear factor 1 transcription factors was decreased 16 % by oxidation, whereas no change was observed in the protein content of several isoforms of these proteins. CONCLUSION/INTERPRETATION: Oxidative stress causes decreased GLUT4 expression, associated with impaired binding of nuclear proteins to the insulin responsive element in the GLUT4 promoter.


Assuntos
DNA/metabolismo , Insulina/fisiologia , Proteínas de Transporte de Monossacarídeos/genética , Proteínas Musculares , Proteínas Nucleares/metabolismo , Estresse Oxidativo/fisiologia , Regiões Promotoras Genéticas/genética , Elementos de Resposta/genética , Células 3T3/efeitos dos fármacos , Animais , Sequência Consenso , DNA/genética , Diamida/farmacologia , Etilmaleimida/farmacologia , Transportador de Glucose Tipo 4 , Peróxido de Hidrogênio/farmacologia , Camundongos , Proteínas de Transporte de Monossacarídeos/metabolismo , Oxidantes/farmacologia , RNA Mensageiro/metabolismo
20.
Biochem J ; 349(Pt 2): 579-86, 2000 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-10880357

RESUMO

Decreased cellular GSH content is a common finding in experimental and human diabetes, in which increased oxidative stress appears to occur. Oxidative stress has been suggested to play a causative role in the development of impaired insulin action on adipose tissue and skeletal muscle. In this study we undertook to investigate the potential of GSH depletion to induce insulin resistance, by utilizing the GSH synthesis inhibitor, L-buthionine-[S,R]-sulfoximine (BSO). GSH depletion (20-80% in various tissues), was achieved in vivo by treating rats for 20 days with BSO, and in vitro (80%) by treating 3T3-L1 adipocytes with BSO for 18 h. No demonstrable change in the GSH/GSSG ratio was observed following BSO treatment. GSH depletion was progressively associated with abnormal glucose tolerance test, which could not be attributed to impaired insulin secretion. Skeletal muscle insulin responsiveness was unaffected by GSH depletion, based on normal glucose response to exogenous insulin, 2-deoxyglucose uptake measurements in isolated soleus muscle, and on normal skeletal muscle expression of GLUT4 protein. Adipocyte insulin responsiveness in vitro was assessed in 3T3-L1 adipocytes, which displayed decreased insulin-stimulated tyrosine phosphorylation of insulin-receptor-substrate proteins and of the insulin receptor, but exaggerated protein kinase B phosphorylation. However, insulin-stimulated glucose uptake was unaffected by GSH depletion. In accordance, normal adipose tissue insulin sensitivity was observed in BSO-treated rats in vivo, as demonstrated by normal inhibition of circulating non-esterified fatty acid levels by endogenous insulin secretion. In conclusion, GSH depletion by BSO results in impaired glucose tolerance, but preserved adipocyte and skeletal muscle insulin responsiveness. This suggests that alternative oxidation-borne factors mediate the induction of peripheral insulin resistance by oxidative stress.


Assuntos
Adipócitos/efeitos dos fármacos , Butionina Sulfoximina/farmacologia , Glutationa/antagonistas & inibidores , Insulina/fisiologia , Músculo Esquelético/efeitos dos fármacos , Células 3T3 , Adipócitos/metabolismo , Animais , Inibidores Enzimáticos/farmacologia , Masculino , Camundongos , Músculo Esquelético/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...