Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 7: e2065, 2016 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-26794660

RESUMO

Proinflammatory cytokines exert cytotoxic effects on ß-cells, and are involved in the pathogenesis of type I and type II diabetes and in the drastic loss of ß-cells following islet transplantation. Cytokines induce apoptosis and alter the function of differentiated ß-cells. Although the MAP3 kinase tumor progression locus 2 (Tpl2) is known to integrate signals from inflammatory stimuli in macrophages, fibroblasts and adipocytes, its role in ß-cells is unknown. We demonstrate that Tpl2 is expressed in INS-1E ß-cells, mouse and human islets, is activated and upregulated by cytokines and mediates ERK1/2, JNK and p38 activation. Tpl2 inhibition protects ß-cells, mouse and human islets from cytokine-induced apoptosis and preserves glucose-induced insulin secretion in mouse and human islets exposed to cytokines. Moreover, Tpl2 inhibition does not affect survival or positive effects of glucose (i.e., ERK1/2 phosphorylation and basal insulin secretion). The protection against cytokine-induced ß-cell apoptosis is strengthened when Tpl2 inhibition is combined with the glucagon-like peptide-1 (GLP-1) analog exendin-4 in INS-1E cells. Furthermore, when combined with exendin-4, Tpl2 inhibition prevents cytokine-induced death and dysfunction of human islets. This study proposes that Tpl2 inhibitors, used either alone or combined with a GLP-1 analog, represent potential novel and effective therapeutic strategies to protect diabetic ß-cells.


Assuntos
Diabetes Mellitus Tipo 2/etiologia , MAP Quinase Quinase Quinases/metabolismo , Peptídeos/metabolismo , Peçonhas/metabolismo , Apoptose , Doença Crônica , Citocinas , Diabetes Mellitus Tipo 2/patologia , Exenatida , Humanos , Inflamação
2.
Oncogenesis ; 5: e188, 2016 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-26779810

RESUMO

The metabolic features of tumor cells diverge from those of normal cells. Otto Warburg was the first to observe that cancer cells dramatically increase their glucose consumption to generate ATP. He also claimed that cancer cells do not have functional mitochondria or oxidative phosphorylation (OXPHOS) but simply rely on glycolysis to provide ATP to the cell, even in the presence of oxygen (aerobic glycolysis). Several studies have revisited this observation and demonstrated that most cancer cells contain metabolically efficient mitochondria. Indeed, to sustain high proliferation rates, cancer cells require functional mitochondria to provide ATP and intermediate metabolites, such as citrate and cofactors, for anabolic reactions. This difference in metabolism between normal and tumors cells causes the latter to be more sensitive to agents that can disrupt energy homeostasis. In this review, we focus on energy disruptors, such as biguanides, 2-deoxyglucose and 5-aminoimidazole-4-carboxamide ribonucleotide, that interfere with the main metabolic pathways of the cells, OXPHOS, glycolysis and glutamine metabolism. We discuss the preclinical data and the mechanisms of action of these disruptors at the cellular and molecular levels. Finally, we consider whether these drugs can reasonably contribute to the antitumoral therapeutic arsenal in the future.

3.
Cell Death Differ ; 20(4): 611-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23238567

RESUMO

The phosphoinositide-3 kinase/Akt (PI3K/Akt) pathway has a central role in cancer cell metabolism and proliferation. More importantly, it is one of the cardinal pro-survival pathways mediating resistance to apoptosis. The role of Akt in response to an energetic stress is presently unclear. Here, we show that Sestrin2 (Sesn2), also known as Hi95, a p53 target gene that protects cells against oxidative and genotoxic stresses, participates in the protective role of Akt in response to an energetic stress induced by 2-deoxyglucose (2-DG). Sesn2 is upregulated in response to an energetic stress such as 2-DG and metformin, and mediates the inhibition of mammalian target of rapamycin (mTOR), the major cellular regulator of energy metabolism. The increase of Sesn2 is independent of p53 but requires the anti-apoptotic pathway, PI3K/Akt. Inhibition of Akt, as well as loss of Sesn2, sensitizes cells to 2-DG-induced apoptosis. In addition, the rescue of Sesn2 partially reverses the pro-apoptotic effects of 2-DG. In conclusion, we identify Sesn2 as a new energetic stress sensor, which appears to be protective against energetic stress-induced apoptosis that integrates the pro-survival function of Akt and the negative regulation of mTOR.


Assuntos
Apoptose/efeitos dos fármacos , Desoxiglucose/toxicidade , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/metabolismo , Caspases/metabolismo , Linhagem Celular , Humanos , Metformina/toxicidade , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinase/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
4.
Diabetologia ; 54(1): 180-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20953578

RESUMO

AIMS/HYPOTHESIS: Extracellular signal-regulated kinase (ERK) activity is increased in adipose tissue in obesity and type 2 diabetes mellitus and strong evidences suggests that it is implicated in the downregulation of insulin signalling and action in the insulin-resistant state. To determine the role of ERK1 in obesity-associated insulin resistance in vivo, we inactivated Erk1 (also known as Mapk3) in obese leptin-deficient mice (ob/ob). METHODS: Mice of genotype ob/ob-Erk1⁻(/)⁻ were obtained by crossing Erk1⁻(/)⁻ mice with ob/ob mice. Glucose tolerance and insulin sensitivity were studied in 12-week-old mice. Tissue-specific insulin sensitivity, insulin signalling, liver steatosis and adipose tissue inflammation were determined. RESULTS: While ob/ob-Erk1⁻(/)⁻ and ob/ob mice exhibited comparable body weight and adiposity, ob/ob-Erk1⁻(/)⁻ mice did not develop hyperglycaemia and their glucose tolerance was improved. Hyperinsulinaemic-euglycaemic clamp studies demonstrated an increase in whole-body insulin sensitivity in the ob/ob-Erk1⁻(/)⁻ mice associated with an increase in both insulin-stimulated glucose disposal in skeletal muscles and adipose tissue insulin sensitivity. This occurred in parallel with improved insulin signalling in both tissues. The ob/ob-Erk1⁻(/)⁻ mice were also partially protected against hepatic steatosis with a strong reduction in acetyl-CoA carboxylase level. These metabolic improvements were associated with reduced expression of mRNA encoding inflammatory cytokine and T lymphocyte markers in the adipose tissue. CONCLUSIONS/INTERPRETATION: Our results demonstrate that the targeting of ERK1 could partially protect obese mice against insulin resistance and liver steatosis by decreasing adipose tissue inflammation and by increasing muscle glucose uptake. Our results indicate that deregulation of the ERK1 pathway could be an important component in obesity-associated metabolic disorders.


Assuntos
Resistência à Insulina/fisiologia , Leptina/deficiência , Proteína Quinase 3 Ativada por Mitógeno/deficiência , Obesidade/fisiopatologia , Animais , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/fisiopatologia , Feminino , Resistência à Insulina/genética , Leptina/genética , Masculino , Camundongos , Camundongos Knockout , Camundongos Obesos , Proteína Quinase 3 Ativada por Mitógeno/genética , Obesidade/genética
5.
Diabetologia ; 52(5): 932-40, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19219422

RESUMO

AIMS/HYPOTHESIS: Insulin resistance is caused by numerous factors including inflammation. It is characterised by defective insulin stimulation of adipocyte and muscle glucose transport, which requires the glucose transporter GLUT4 translocation towards the plasma membrane. Defects in insulin signalling can cause insulin resistance, but alterations in GLUT4 trafficking could also play a role. Our goal was to determine whether proteins controlling GLUT4 trafficking are altered in insulin resistance linked to obesity. METHODS: Using real-time RT-PCR, we searched for selected transcripts that were differentially expressed in adipose tissue and muscle in obese mice and humans. Using various adipocyte culture models and in vivo mice treatment, we searched for the involvement of TNF-alpha in these alterations in obesity. RESULTS: Sortilin mRNA and protein were downregulated in adipose tissue from obese db/db and ob/ob mice, and also in muscle. Importantly, sortilin mRNA was also decreased in morbidly obese human diabetic patients. Sortilin and TNF-alpha (also known as TNF) mRNA levels were inversely correlated in mice and human adipose tissues. TNF-alpha decreased sortilin mRNA and protein levels in cultured mouse and human adipocytes, an effect partly prevented by the peroxisome proliferator-activated receptor gamma activator rosiglitazone. TNF-alpha also inhibited adipocyte and muscle sortilin mRNA when injected to mice. CONCLUSIONS/INTERPRETATION: Sortilin, an essential player in adipocyte and muscle glucose metabolism through the control of GLUT4 localisation, is downregulated in obesity and TNF-alpha is likely to be involved in this defect. Chronic low-grade inflammation in obesity could thus contribute to insulin resistance by modulating proteins that control GLUT4 trafficking.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Adipócitos/fisiologia , Músculo Esquelético/fisiopatologia , Obesidade Mórbida/genética , Obesidade/fisiopatologia , Fator de Necrose Tumoral alfa/fisiologia , Adipócitos/patologia , Adulto , Animais , Cirurgia Bariátrica , Primers do DNA , Regulação para Baixo , Epididimo/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Obesos , Pessoa de Meia-Idade , Obesidade/genética , Obesidade Mórbida/cirurgia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
6.
Diabetologia ; 52(6): 1152-63, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19183933

RESUMO

AIMS/HYPOTHESIS: Obesity is associated with adipose tissue inflammation. The CD40 molecule, TNF receptor superfamily member 5 (CD40)/CD40 ligand (CD40L) pathway plays a role in the onset and maintenance of the inflammatory reaction, but has not been studied in human adipose tissue. Our aim was to examine CD40 expression by human adipocytes and its participation in adipose tissue inflammation. METHODS: CD40 expression was investigated in human whole adipose tissue and during adipocyte differentiation by real-time PCR, Western blot and immunohistochemistry. The CD40/CD40L pathway was studied using recombinant CD40L (rCD40L) in adipocyte culture and neutralising antibodies in lymphocyte/adipocyte co-culture. RESULTS: CD40 mRNA levels in subcutaneous adipose tissue were higher in the adipocyte than in the stromal-vascular fraction. CD40 expression was upregulated during adipocyte differentiation. Addition of rCD40L to adipocytes induced mitogen activated protein kinase (MAPK) activation, stimulated inflammatory adipocytokine production, and decreased insulin-induced glucose transport in parallel with a downregulation of IRS1 and GLUT4 (also known as SCL2A4). rCD40L decreased the expression of lipogenic genes and increased lipolysis. CD40 mRNA levels were significantly higher in subcutaneous adipose tissue than in visceral adipose tissue of obese patients and were positively correlated with BMI, and with IL6 and leptin mRNA levels. Lymphocyte/adipocyte co-culture led to an upregulation of proinflammatory adipocytokines and a downregulation of leptin and adiponectin. Physical separation of the two cell types attenuated these effects, suggesting the involvement of a cell-cell contact. Blocking the CD40/CD40L interaction with neutralising antibodies reduced IL-6 secretion from adipocytes. CONCLUSIONS/INTERPRETATION: Adipocyte CD40 may contribute to obesity-related inflammation and insulin resistance. T lymphocytes regulate adipocytokine production through both the release of soluble factor(s) and heterotypic contact with adipocytes involving CD40.


Assuntos
Adipócitos/metabolismo , Antígenos CD40/genética , Antígenos CD40/metabolismo , Linfócitos/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adiponectina/metabolismo , Adrenomedulina/metabolismo , Animais , Western Blotting , Ligante de CD40/farmacologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Linfócitos/efeitos dos fármacos , Camundongos , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Oncogene ; 27(25): 3576-86, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18212742

RESUMO

Metformin is a widely used antidiabetic agent, which regulates glucose homeostasis through inhibition of liver glucose production and an increase in muscle glucose uptake. Recent studies suggest that metformin may reduce the risk of cancer, but its mode of action in cancer remains not elucidated. We investigated the effect of metformin on human prostate cancer cell proliferation in vitro and in vivo. Metformin inhibited the proliferation of DU145, PC-3 and LNCaP cancer cells with a 50% decrease of cell viability and had a modest effect on normal prostate epithelial cell line P69. Metformin did not induce apoptosis but blocked cell cycle in G(0)/G(1). This blockade was accompanied by a strong decrease of cyclin D1 protein level, pRb phosphorylation and an increase in p27(kip) protein expression. Metformin activated the AMP kinase pathway, a fuel sensor signaling pathway. However, inhibition of the AMPK pathway using siRNA against the two catalytic subunits of AMPK did not prevent the antiproliferative effect of metformin in prostate cancer cells. Importantly, oral and intraperitoneal treatment with metformin led to a 50 and 35% reduction of tumor growth, respectively, in mice bearing xenografts of LNCaP. Similar, to the in vitro study, metformin led to a strong reduction of cyclin D1 protein level in tumors providing evidence for a mechanism that may contribute to the antineoplastic effects of metformin suggested by recent epidemiological studies.


Assuntos
Antineoplásicos/farmacologia , Ciclina D1/biossíntese , Regulação Neoplásica da Expressão Gênica , Metformina/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Proteínas Quinases Ativadas por AMP , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Humanos , Hipoglicemiantes/farmacologia , Masculino , Complexos Multienzimáticos/metabolismo , Próstata/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
8.
FEBS Lett ; 581(29): 5591-6, 2007 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-17997987

RESUMO

Little is known about the role of p38MAPK in human adipocyte differentiation. Here we showed that p38MAPK activity increases during human preadipocytes differentiation. Pharmacological inhibition of p38MAPK during adipocyte differentiation of primary human preadipocytes markedly reduced triglycerides accumulation and adipocyte markers expression. Cell cycle arrest or proliferation was not affected by p38MAPK inhibition. Although induction of C/EBPbeta was not altered by the p38MAPK inhibitor, its phosphorylation on Threonine(188) was decreased as well as PPARgamma expression. These results indicate that p38MAPK plays a positive role in human adipogenesis through regulation of C/EBPbeta and PPARgamma factors.


Assuntos
Adipócitos/citologia , Adipócitos/enzimologia , Diferenciação Celular , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adipogenia/fisiologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Ciclo Celular , Humanos , Imidazóis/farmacologia , PPAR gama/metabolismo , Fosforilação , Transdução de Sinais , Treonina/genética , Treonina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
9.
Diabetologia ; 50(6): 1267-76, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17426960

RESUMO

AIMS/HYPOTHESIS: Inflammation is associated with obesity and has been implicated in the development of diabetes and atherosclerosis. During gram-negative bacterial infection, lipopolysaccharide causes an inflammatory reaction via toll-like receptor 4 (TLR4), which has an essential function in the induction of innate and adaptative immunity. Our aim was to determine what role TLR4 plays in the development of metabolic phenotypes during high-fat feeding. MATERIALS AND METHODS: We evaluated metabolic consequences of a high-fat diet in TLR4 mutant mice (C3H/HeJ) and their respective controls. RESULTS: TLR4 inactivation reduced food intake without significant modification of body weight, but with higher epididymal adipose tissue mass and adipocyte hypertrophy. It also attenuated the inflammatory response and increased glucose transport and the expression levels of adiponectin and lipogenic markers in white adipose tissue. In addition, TLR4 inactivation blunted insulin resistance induced by lipopolysaccharide in differentiated adipocytes. Increased feeding efficiency in TLR4 mutant mice was associated with lower mass and lower expression of uncoupling protein 1 gene in brown adipose tissue. Finally, TLR4 inactivation slowed the development of hepatic steatosis, reducing the liver triacylglycerol content and also expression levels of lipogenic and fibrosis markers. CONCLUSIONS/INTERPRETATION: TLR4 influences white adipose tissue inflammation and insulin sensitivity, as well as liver fat storage, and is important in the regulation of metabolic phenotype during a fat-enriched diet.


Assuntos
Tecido Adiposo/fisiologia , Gorduras na Dieta/farmacologia , Resistência à Insulina/genética , Mutação , Receptor 4 Toll-Like/genética , Tecido Adiposo/fisiopatologia , Animais , Glicemia/metabolismo , Glicólise , Inflamação , Lipídeos/fisiologia , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Triglicerídeos/sangue
10.
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
11.
Ann Endocrinol (Paris) ; 65(1): 43-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15122091

RESUMO

Insulin resistance, when combined with impaired insulin secretion, contributes to the development of type 2 diabetes. Insulin resistance is characterised by a decrease in insulin effect on glucose transport in muscle and adipose tIssue. Tyrosine phosphorylation of insulin receptor substrate 1 (IRS-1) and its binding to phosphatidylinositol 3-kinase (PI 3-kinase) are critical events in the insulin signalling cascade leading to insulin-stimulated glucose transport. Modification of IRS-1 by serine phosphorylation could be one of the mechanisms leading to a decrease in IRS-1 tyrosine phosphorylation, PI 3-kinase activity and glucose transport. Recent findings demonstrate that "diabetogenic" factors such as FFA, TNFalpha, hyperinsulinemia and cellular stress, increase the serine phosphorylation of IRS-1 and identified Ser307/612/632 as phosphorylated sites. Moreover, several kinases able to phosphorylate these serine residues have been identified. These exciting results suggest that serine phosphorylation of IRS-1 is a possible hallmark of insulin resistance in biologically insulin responsive cells or tIssues. Identifying the pathways by which "diabetogenic" factors activate IRS-1 kinases and defining the precise role of serine phosphorylation events in IRS-1 regulation represent important goals. Such studies may enable rational drug design to selectively inhibit the activity of the relevant enzymes and generate a novel class of therapeutic agents for type 2 diabetes.


Assuntos
Resistência à Insulina/fisiologia , Insulina/fisiologia , Fosfoproteínas/metabolismo , Transdução de Sinais/fisiologia , Animais , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Homeostase , Humanos , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina , Secreção de Insulina , Fosfoproteínas/fisiologia , Fosforilação , Fosfosserina/metabolismo , Receptor de Insulina/fisiologia
12.
Biochem Soc Trans ; 31(Pt 6): 1152-6, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14641015

RESUMO

Insulin resistance, when combined with impaired insulin secretion, contributes to the development of type 2 diabetes. Insulin resistance is characterized by a decrease in the insulin effect on glucose transport in muscle and adipose tissue. Tyrosine phosphorylation of IRS-1 (insulin receptor substrate 1) and its binding to PI 3-kinase (phosphoinositide 3-kinase) are critical events in the insulin signalling cascade leading to insulin-stimulated glucose transport. Various studies have implicated lipids as a cause of insulin resistance in muscle. Elevated plasma fatty acid concentrations are associated with reduced insulin-stimulated glucose transport activity as a consequence of altered insulin signalling through PI 3-kinase. Modification of IRS-1 by serine phosphorylation could be one of the mechanisms leading to a decrease in IRS-1 tyrosine phosphorylation, PI 3-kinase activity and glucose transport. Recent findings demonstrate that non-esterified fatty acids, as well as other factors such as tumour necrosis factor alpha, hyperinsulinaemia and cellular stress, increase the serine phosphorylation of IRS-1 and identified Ser(307) as one of the phosphorylated sites. Moreover, several kinases able to phosphorylate this serine residue have been identified. These exciting results suggest that Ser(307) phosphorylation is a possible hallmark of insulin resistance in biologically insulin-responsive cells or tissues. Identification of IRS-1 kinases could enable rational drug design in order to selectively inhibit the activity of the relevant enzymes and generate a novel class of therapeutic agents for type 2 diabetes.


Assuntos
Ácidos Graxos/fisiologia , Resistência à Insulina , Insulina/metabolismo , Fosfoproteínas/metabolismo , Serina/metabolismo , Transdução de Sinais , Animais , Glucose/metabolismo , Proteínas Substratos do Receptor de Insulina , Pressão Osmótica , Fosfoproteínas/química , Fator de Necrose Tumoral alfa/metabolismo
13.
Diabetologia ; 46(11): 1532-42, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14579029

RESUMO

AIM/HYPOTHESIS: Insulin-induced IRS-1 serine phosphorylation could be physiologically important to regulate insulin action. In a hyperinsulinaemic state such as obesity or Type 2 diabetes, this phosphorylation could be modified and exacerbate insulin resistance. We aimed at identifying serine residues in IRS-1 phosphorylated in response to insulin stimulation and at determining the involved kinases. METHODS: 3T3-L1 adipocytes, muscle and adipose tissue of mice were subjected to Western Blot analysis with phosphospecific antibodies to identify phosphorylation sites in IRS-1 following insulin treatment. Pharmacological inhibitors were used to determine the serine kinases involved in this phosphorylation. RESULTS: In 3T3-L1 adipocytes, insulin promoted the phosphorylation of serine 307, 612 and 632 with Serine(612/632) more rapidly phosphorylated than Serine(307). Insulin-induced phosphorylation of Serine(307) was dependent on the activation of a PI 3-kinase/mTOR pathway. The phosphorylation of Serine(612/632) required the activation of the MAP kinase pathway following short-term insulin stimulation and activation of the PI 3-kinase/mTOR pathway following prolonged insulin stimulation. Phosphorylation of Serine(307) and Serine(632) occurred in vivo in skeletal muscle and white adipose tissue of mice injected with insulin and was dependent on the activation of mTOR. Moreover, inhibition of mTOR led to a persistent PI 3-kinase activation by insulin. CONCLUSION/INTERPRETATION: Insulin-induced IRS-1 serine phosphorylation is a complex process involving different sites and kinases. This complexity could be physiologically important to accurately regulate insulin signalling. Abnormal phosphorylation of these serine residues in hyperinsulinaemic state could participate in the down-regulation of insulin signalling.


Assuntos
Insulina/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfoproteínas/metabolismo , Fosfosserina/metabolismo , Proteínas Quinases/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Insulina/fisiologia , Proteínas Substratos do Receptor de Insulina , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/química , Fosfoproteínas/efeitos dos fármacos , Fosforilação , Transdução de Sinais , Serina-Treonina Quinases TOR
15.
Nature ; 409(6816): 92-7, 2001 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-11343120

RESUMO

Insulin is the primary hormone involved in glucose homeostasis, and impairment of insulin action and/or secretion has a critical role in the pathogenesis of diabetes mellitus. Type-II SH2-domain-containing inositol 5-phosphatase, or 'SHIP2', is a member of the inositol polyphosphate 5-phosphatase family. In vitro studies have shown that SHIP2, in response to stimulation by numerous growth factors and insulin, is closely linked to signalling events mediated by both phosphoinositide-3-OH kinase and Ras/mitogen-activated protein kinase. Here we report the generation of mice lacking the SHIP2 gene. Loss of SHIP2 leads to increased sensitivity to insulin, which is characterized by severe neonatal hypoglycaemia, deregulated expression of the genes involved in gluconeogenesis, and perinatal death. Adult mice that are heterozygous for the SHIP2 mutation have increased glucose tolerance and insulin sensitivity associated with an increased recruitment of the GLUT4 glucose transporter and increased glycogen synthesis in skeletal muscles. Our results show that SHIP2 is a potent negative regulator of insulin signalling and insulin sensitivity in vivo.


Assuntos
Insulina/fisiologia , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Feminino , Deleção de Genes , Marcação de Genes , Hipoglicemia/etiologia , Hipoglicemia/genética , Insulina/sangue , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Fosfoenolpiruvato Carboxiquinase (GTP)/metabolismo , Monoéster Fosfórico Hidrolases/genética , Transdução de Sinais , Células-Tronco
17.
Eur J Biochem ; 267(22): 6642-9, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11054117

RESUMO

Phosphoinositide-dependent protein kinase-1 (PDK1) is a recently identified kinase that phosphorylates and activates protein kinase B (PKB). Activation of PKB by insulin is linked to its translocation from the cytosol to the plasma membrane. However, no data are available yet concerning the localization of PDK1 in insulin-sensitive tissue. Using isolated adipocytes, we studied the effect of insulin and of an insulin-mimicking agent peroxovanadate on the subcellular localization of PDK1. In unstimulated adipocytes, overexpressed PDK1 was mostly cytosolic with a low amount associated to membranes. Peroxovanadate stimulation induced the redistribution of PDK1 to the membranes while insulin was without effect. This peroxovanadate effect was dependent on phosphatidylinositol 3,4,5 triphosphate [PtdIns(3,4,5)P3] production as inhibition of PtdIns 3-kinase by wortmannin or deletion of the PH domain of PDK1 prevented the peroxovanadate-induced translocation of PDK1. Further, peroxovanadate-treatment induced a tyrosine phosphorylation of PDK1 which was wortmannin insensitive and did not require the PH domain of PDK1. An inhibitor of Src kinase (PP2) decreased the peroxovanadate-induced PDK1 tyrosine phosphorylation and overexpression of v-Src stimulated this phosphorylation. Mutation of tyrosine 373 of PDK1 abolished the v-Src induced PDK1 tyrosine phosphorylation and partially reduced the effect of peroxovanadate. Our findings suggest that PDK1 could be a substrate for tyrosine kinases and identify Src kinase as one of the tyrosine kinases able to phosphorylate PDK1.


Assuntos
Adipócitos/enzimologia , Fosfotirosina/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Vanadatos/farmacologia , Quinases da Família src/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Adipócitos/citologia , Animais , Células Cultivadas , Cinética , Masculino , Fosforilação , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo , Transfecção
18.
J Recept Signal Transduct Res ; 19(1-4): 217-28, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10071760

RESUMO

Insulin resistance is commonly associated with obesity in rodents. Using mice made obese with goldthioglucose (GTG-obese mice), we have shown that insulin resistance results from defects at the level of the receptor and from intracellular alterations in insulin signalling pathway, without major alteration in the number of the Glut 4 glucose transporter. Activation of phosphatidylinositol 3-kinase (PI 3-kinase) was found to be profoundly affected in response to insulin. This defect appears very early in the development of obesity, together with a marked decrease in IRS 1 tyrosine phosphorylation. In order to better understand the abnormalities in glucose transport in insulin resistance, we have studied the pathway leading from the insulin receptor kinase stimulation to the translocation of the Glut 4 containing vesicles. This stimulation involves the activation of PI 3-kinase, which in turns activates protein kinase B. We have then focussed at the mechanism of vesicle exocytosis, and more specifically at the role of the small GTPase Rab4 in this process. We have shown that Rab4 participates, first in the intracellular retention of the Glut 4 containing vesicles, second in the insulin signalling pathway leading to glucose transporter translocation.


Assuntos
Resistência à Insulina/fisiologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Obesidade/metabolismo , Proteínas Serina-Treonina Quinases , Receptor de Insulina/metabolismo , Tecido Adiposo/metabolismo , Animais , Transporte Biológico Ativo , Ativação Enzimática/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Transportador de Glucose Tipo 4 , Insulina/farmacologia , Camundongos , Músculos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Proteínas rab4 de Ligação ao GTP
19.
J Biol Chem ; 272(32): 19814-8, 1997 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-9242642

RESUMO

Phosphatidylinositol (PI) 3-kinase is activated by various growth factors such as PDGF (platelet-derived growth factor) and insulin. The aim of the present study was to determine whether PDGF could modulate insulin activation of PI 3-kinase in 3T3-L1 adipocytes. When cells were preincubated for 5-15 min with PDGF, PI 3-kinase activity associated to insulin receptor substrate 1 (IRS 1) in response to insulin was decreased, due to reduced association of the PI 3-kinase p85 subunit with IRS 1. In addition, following this PDGF pretreatment, the tyrosine phosphorylation of IRS 1 in response to insulin and its electrophoretic mobility were diminished. The change in the mobility of IRS 1 could be attributed to PDGF-induced serine/threonine phosphorylation of the protein which was partly inhibited by PI 3-kinase inhibitors. By contrast, epidermal growth factor, which does not stimulate PI 3-kinase, had no effect on the association of PI 3-kinase with IRS 1 in response to insulin. This series of results indicates that the PDGF-induced serine/threonine phosphorylation of IRS 1 could be due to activation of PI 3-kinase pathway. Furthermore, this phosphorylation of IRS 1 is associated with a decrease in its tyrosine phosphorylation by insulin and in its association with the p85 subunit of PI 3-kinase. This study suggests that a cross-talk exists between the different pathways stimulated by PDGF and insulin in intact cells.


Assuntos
Adipócitos/metabolismo , Insulina/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Células 3T3 , Androstadienos/farmacologia , Animais , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Antagonistas da Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina , Camundongos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases , Fosfoproteínas/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Tirosina/metabolismo , Wortmanina
20.
Endocrinology ; 138(5): 2005-10, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9112399

RESUMO

Phosphatidylinositol 3-kinase (PI 3-kinase) activation promotes glucose transporter 4 (Glut 4) translocation in adipocytes. In this study, we demonstrate that protein kinase B, a serine/threonine kinase stimulated by PI 3-kinase, is activated by both insulin and okadaic acid in isolated adipocytes, in parallel with their effects on Glut 4 translocation. In 3T3-L1 adipocytes, platelet-derived growth factor activated PI 3-kinase as efficiently as insulin but was only half as potent as insulin in promoting protein kinase B (PKB) activation. To look for a potential role of PKB in Glut 4 translocation, adipocytes were transfected with a constitutively active PKB (Gag-PKB) together with an epitope tagged transporter (Glut 4 myc). Gag-PKB was associated with all membrane fractions, whereas the endogenous PKB was mostly cytosolic. Expression of Gag-PKB led to an increase in Glut 4 myc amount at the cell surface. Our results suggest that PKB could play a role in promoting Glut 4 appearance at the cell surface following exposure of adipocytes to insulin and okadaic acid stimulation.


Assuntos
Adipócitos/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Animais , Transporte Biológico , Ativação Enzimática/efeitos dos fármacos , Transportador de Glucose Tipo 4 , Immunoblotting , Técnicas de Imunoadsorção , Insulina/farmacologia , Masculino , Ácido Okadáico/farmacologia , Fosfatidilinositol 3-Quinases , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Wistar , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...