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











Base de dados
Intervalo de ano de publicação
1.
Ann Oncol ; 27(7): 1286-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27056998

RESUMO

BACKGROUND: RET rearrangements are targetable, oncogenic lung cancer drivers. While previous series have shown durable clinical benefit with pemetrexed-based therapies in ALK- and ROS1-rearranged lung cancers, the benefits of pemetrexed-based treatments in patients with RET-rearranged lung cancers relative to other genomic subsets have not previously been explored. PATIENTS AND METHODS: A retrospective review of patients with pathologically confirmed stage IIIB/IV lung adenocarcinomas and evidence of a RET, ROS1, or ALK rearrangement, or a KRAS mutation was conducted. Patients were eligible if they received treatment with pemetrexed alone or in combination. The primary outcome of progression-free survival (PFS), and secondary outcomes of overall response rate (ORR, RECIST v1.1), time to progression (TTP), and time to treatment discontinuation were compared between RET-rearranged and groups of ROS1-rearranged, ALK-rearranged, and KRAS-mutant lung cancers. RESULTS: We evaluated 104 patients. Patients with RET-rearranged lung cancers (n = 18) had a median PFS of 19 months [95% confidence interval (CI) 12-not reached (NR)] that was comparable with patients with ROS1- (23 months, 95% CI 14-NR, n = 10) and ALK-rearranged (19 months, 95% CI 15-36, n = 36) lung cancers, and significantly improved compared with patients with KRAS-mutant lung cancers (6 months, 95% CI 5-9, P < 0.001, n = 40). ORR (45%), median TTP (20 months, 95% CI 17-NR), and median time to treatment discontinuation (21 months, 95% CI 6-NR) in patients with RET-rearranged lung cancers were not significantly different compared with patients with ALK- and ROS1-rearranged lung cancers, and improved compared with patients with KRAS-mutant lung cancers. CONCLUSION: Durable benefits with pemetrexed-based therapies in RET-rearranged lung cancers are comparable with ALK- and ROS1-rearranged lung cancers. When selecting therapies for patients with RET-rearranged lung cancers, pemetrexed-containing regimens should be considered.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Idoso , Quinase do Linfoma Anaplásico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Intervalo Livre de Doença , Feminino , Rearranjo Gênico , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Pemetrexede/administração & dosagem , Pemetrexede/efeitos adversos , Proteínas Proto-Oncogênicas p21(ras)/genética
2.
Oncogene ; 28(31): 2773-83, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19525976

RESUMO

To address the biological heterogeneity of lung cancer, we studied 199 lung adenocarcinomas by integrating genome-wide data on copy number alterations and gene expression with full annotation for major known somatic mutations in this cancer. This showed non-random patterns of copy number alterations significantly linked to EGFR and KRAS mutation status and to distinct clinical outcomes, and led to the discovery of a striking association of EGFR mutations with underexpression of DUSP4, a gene within a broad region of frequent single-copy loss on 8p. DUSP4 is involved in negative feedback control of EGFR signaling, and we provide functional validation for its role as a growth suppressor in EGFR-mutant lung adenocarcinoma. DUSP4 loss also associates with p16/CDKN2A deletion and defines a distinct clinical subset of lung cancer patients. Another novel observation is that of a reciprocal relationship between EGFR and LKB1 mutations. These results highlight the power of integrated genomics to identify candidate driver genes within recurrent broad regions of copy number alteration and to delineate distinct oncogenetic pathways in genetically complex common epithelial cancers.


Assuntos
Adenocarcinoma/genética , Fosfatases de Especificidade Dupla/genética , Receptores ErbB/genética , Perfilação da Expressão Gênica , Neoplasias Pulmonares/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Mutação , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células , Aberrações Cromossômicas , Análise por Conglomerados , Inibidor p16 de Quinase Dependente de Ciclina/genética , Feminino , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Genes ras/genética , Estudo de Associação Genômica Ampla , Humanos , Hibridização in Situ Fluorescente , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/patologia , Masculino , Hibridização de Ácido Nucleico , Interferência de RNA
3.
Biochem J ; 359(Pt 3): 639-49, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11672439

RESUMO

We previously reported that SB203580, an inhibitor of p38 mitogen-activated protein kinase (p38 MAPK), attenuates insulin-stimulated glucose uptake without altering GLUT4 translocation. These results suggested that insulin might activate GLUT4 via a p38 MAPK-dependent pathway. Here we explore this hypothesis by temporal and kinetic analyses of the stimulation of GLUT4 translocation, glucose uptake and activation of p38 MAPK isoforms by insulin. In L6 myotubes stably expressing GLUT4 with an exofacial Myc epitope, we found that GLUT4 translocation (t(1/2)=2.5 min) preceded the stimulation of 2-deoxyglucose uptake (t(1/2)=6 min). This segregation of glucose uptake from GLUT4 translocation became more apparent when the two parameters were measured at 22 degrees C. Preincubation with the p38 MAPK inhibitors SB202190 and SB203580 reduced insulin-stimulated transport of either 2-deoxyglucose or 3-O-methylglucose by 40-60%. Pretreatment with SB203580 lowered the apparent transport V(max) of insulin-mediated 2-deoxyglucose and 3-O-methylglucose without any significant change in the apparent K(m) for either hexose. The IC(50) values for the partial inhibition of 2-deoxyglucose uptake by SB202190 and SB203580 were 1 and 2 microM respectively, and correlated with the IC(50) for full inhibition of p38 MAPK by the two inhibitors in myotubes (2 and 1.4 microM, respectively). Insulin caused a dose- (EC(50)=15 nM) and time- (t(1/2)=3 min) dependent increase in p38 MAPK phosphorylation, which peaked at 10 min (2.3+/-0.3-fold). In vitro kinase assay of immunoprecipitates from insulin-stimulated myotubes showed activation of p38 alpha (2.6+/-0.3-fold) and p38 beta (2.3+/-0.2-fold) MAPK. These results suggest that activation of GLUT4 follows GLUT4 translocation and that both mechanisms contribute to the full stimulation of glucose uptake by insulin. Furthermore, activation of GLUT4 may occur via an SB203580-sensitive pathway, possibly involving p38 MAPK.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , Proteínas Serina-Treonina Quinases , Animais , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Glucose/análogos & derivados , Transportador de Glucose Tipo 4 , Humanos , Isoenzimas , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Músculo Esquelético/citologia , Músculo Esquelético/efeitos dos fármacos , Fosforilação , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
4.
Endocrinology ; 142(11): 4806-12, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11606447

RESUMO

Obesity is a major risk factor for the development of insulin resistance, characterized by impaired stimulation of glucose disposal into muscle. The mechanisms underlying insulin resistance are unknown. Here we examine the direct effect of leptin, the product of the obesity gene, on insulin-stimulated glucose uptake in cultured rat skeletal muscle cells. Preincubation of L6 myotubes with leptin (2 or 100 nM, 30 min) had no effect on basal glucose uptake but reduced insulin-stimulated glucose uptake. However, leptin had no effect on the insulin-induced gain in myc-tagged glucose transporter 4 (GLUT4) appearance at the cell surface of L6 myotubes. Preincubation of cells with leptin also had no effect on insulin-stimulated tyrosine phosphorylation of insulin receptor, IRS-1 and IRS-2, phosphatidylinositol 3-kinase activity, or Akt phosphorylation. We have previously shown that insulin regulates glucose uptake via a signaling pathway sensitive to inhibitors of p38 MAP kinase. Here, leptin pretreatment reduced the extent of insulin-stimulated p38 MAP kinase phosphorylation and phosphorylation of cAMP response element binder, a downstream effector of p38 MAP kinase. These results show that high leptin levels can directly reduce insulin-stimulated glucose uptake in L6 muscle cells despite normal GLUT4 translocation. The mechanism of this effect could involve inhibition of insulin-stimulated p38 MAP kinase and GLUT4 activation.


Assuntos
Glucose/antagonistas & inibidores , Glucose/metabolismo , Insulina/farmacologia , Leptina/farmacologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , Proteínas Serina-Treonina Quinases , Animais , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Transportador de Glucose Tipo 4 , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Esquelético/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Tirosina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
5.
Diabetologia ; 43(3): 294-303, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10768090

RESUMO

AIMS/HYPOTHESIS: A natural cofactor of mitochondrial dehydrogenase complexes and a potent antioxidant, alpha-lipoic acid improves glucose metabolism in people with Type II (non-insulin-dependent) diabetes mellitus and in animal models of diabetes. In this study we investigated the cellular mechanism of action of alpha-lipoic acid in 3T3-L1 adipocytes. METHODS: We treated 3T3-L1 adipocytes with 2.5 mmol/l R (+) alpha-lipoic acid for 2 to 60 min, followed by assays of: 2-deoxyglucose uptake; glucose transporter 1 and 4 (GLUT1 and GLUT4) subcellular localization; tyrosine phosphorylation of the insulin receptor or of the insulin receptor substrate-1 in cell lysates; association of phosphatidylinositol 3-kinase activity with immunoprecipitates of proteins containing phosphotyrosine or of insulin receptor substrate-1 using a in vitro kinase assay; association of the p85 subunit of phosphatidylinositol 3-kinase with phosphotyrosine proteins or with insulin receptor substrate-1; and in vitro activity of immunoprecipitated Akt1. The effect of R (+) alpha-lipoic acid was also compared with that of S(-) alpha-lipoic acid. RESULTS: Short-term treatment of 3T3-L1 adipocytes with R (+) alpha-lipoic acid rapidly stimulated glucose uptake in a wortmannin-sensitive manner, induced a redistribution of GLUT1 and GLUT4 to the plasma membrane, caused tyrosine phosphorylation of insulin receptor substrate-1 and of the insulin receptor, increased the antiphosphotyrosine-associated and insulin receptor substrate-1 associated phosphatidylinositol 3-kinase activity and stimulated Akt activity. CONCLUSION/INTERPRETATION: These results indicate that R (+) alpha-lipoic acid directly activates lipid, tyrosine and serine/threonine kinases in target cells, which could lead to the stimulation of glucose uptake induced by this natural cofactor. These properties are unique among all agents currently used to lower glycaemia in animals and humans with diabetes.


Assuntos
Adipócitos/metabolismo , Glucose/metabolismo , Insulina/farmacologia , Proteínas Musculares , Proteínas Serina-Treonina Quinases , Ácido Tióctico/farmacologia , Células 3T3 , Adipócitos/efeitos dos fármacos , Androstadienos/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Transportador de Glucose Tipo 1 , Transportador de Glucose Tipo 4 , Proteínas Substratos do Receptor de Insulina , Camundongos , Proteínas de Transporte de Monossacarídeos/metabolismo , Concentração Osmolar , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor de Insulina/metabolismo , Tirosina/metabolismo , Wortmanina
6.
J Biol Chem ; 274(43): 30459-67, 1999 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-10521425

RESUMO

Glucagon-like peptide-2 (GLP-2) promotes the expansion of the intestinal epithelium through stimulation of the GLP-2 receptor, a recently identified member of the glucagon-secretin G protein-coupled receptor superfamily. Although activation of G protein-coupled receptors may lead to stimulation of cell growth, the mechanisms transducing the GLP-2 signal to mitogenic proliferation remain unknown. We now report studies of GLP-2R signaling in baby hamster kidney (BHK) cells expressing a transfected rat GLP-2 receptor (BHK-GLP-2R cells). GLP-2, but not glucagon or GLP-1, increased the levels of cAMP and activated both cAMP-response element- and AP-1-dependent transcriptional activity in a dose-dependent manner. The activation of AP-1-luciferase activity was protein kinase A (PKA) -dependent and markedly diminished in the presence of a dominant negative inhibitor of PKA. Although GLP-2 stimulated the expression of c-fos, c-jun, junB, and zif268, and transiently increased p70 S6 kinase in quiescent BHK-GLP-2R cells, GLP-2 also inhibited extracellular signal-regulated kinase 1/2 and reduced serum-stimulated Elk-1 activity. Furthermore, no rise in intracellular calcium was observed following GLP-2 exposure in BHK-GLP-2R cells. Although GLP-2 stimulated both cAMP accumulation and cell proliferation, 8-bromo-cyclic AMP alone did not promote cell proliferation. These findings suggest that the GLP-2R may be coupled to activation of mitogenic signaling in heterologous cell types independent of PKA via as yet unidentified downstream mediators of GLP-2 action in vivo.


Assuntos
Peptídeos/farmacologia , Proteínas Serina-Treonina Quinases , Receptores de Glucagon/fisiologia , Transdução de Sinais/fisiologia , Animais , Divisão Celular , Linhagem Celular , Cricetinae , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fibroblastos , Hormônios Gastrointestinais/farmacologia , Hormônios Gastrointestinais/fisiologia , Glucagon/farmacologia , Peptídeo 1 Semelhante ao Glucagon , Peptídeo 2 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Rim , Luciferases/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/fisiologia , Fosforilação , Precursores de Proteínas/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Receptores de Glucagon/genética , Proteínas Recombinantes/metabolismo , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica , Transfecção
7.
Mol Cell Biol ; 19(6): 4008-18, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10330141

RESUMO

L6 myoblasts stably transfected with a GLUT4 cDNA harboring an exofacial myc epitope tag (L6-GLUT4myc myoblasts) were used to study the role of protein kinase B alpha (PKBalpha)/Akt1 in the insulin-induced translocation of GLUT4 to the cell surface. Surface GLUT4myc was detected by immunofluorescent labeling of the myc epitope in nonpermeabilized cells. Insulin induced a marked translocation of GLUT4myc to the plasma membrane within 20 min. This was prevented by transient transfection of a dominant inhibitory construct of phosphatidylinositol (PI) 3-kinase (Deltap85alpha). Transiently transfected cells were identified by cotransfection of green fluorescent protein. A constitutively active PKBalpha, created by fusion of a viral Gag protein at its N terminus (GagPKB), increased the cell surface density of GLUT4myc compared to that of neighboring nontransfected cells. A kinase-inactive, phosphorylation-deficient PKBalpha/Akt1 construct with the mutations K179A (substitution of alanine for the lysine at position 179), T308A, and S473A (AAA-PKB) behaved as a dominant-negative inhibitor of insulin-dependent activation of cotransfected wild-type hemagglutinin (HA)-tagged PKB. Furthermore, AAA-PKB markedly inhibited the insulin-induced phosphorylation of cotransfected BAD, demonstrating inhibition of the endogenous PKB/Akt. Under the same conditions, AAA-PKB almost entirely blocked the insulin-dependent increase in surface GLUT4myc. PKBalpha with alanine substitutions T308A and S473A (AA-PKB) or K179A (A-PKB) alone was a less potent inhibitor of insulin-dependent activation of wild-type HA-PKB or GLUT4myc translocation than was AAA-PKB. Cotransfection of AAA-PKB with a fourfold DNA excess of HA-PKB rescued insulin-stimulated GLUT4myc translocation. AAA-PKB did not prevent actin bundling (membrane ruffling), though this response was PI 3-kinase dependent. Therefore, it is unlikely that AAA-PKB acted by inhibiting PI 3-kinase signaling. These results outline an important role for PKBalpha/Akt1 in the stimulation of glucose transport by insulin in muscle cells in culture.


Assuntos
Insulina/metabolismo , Proteínas de Transporte de Monossacarídeos/fisiologia , Proteínas Musculares , Miocárdio/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Células Cultivadas , Imunofluorescência , Transportador de Glucose Tipo 4 , Humanos , Immunoblotting , Mutagênese , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Plasmídeos , Testes de Precipitina , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt , Sequências Reguladoras de Ácido Nucleico , Transfecção , Translocação Genética
8.
J Biol Chem ; 274(15): 10071-8, 1999 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-10187787

RESUMO

The precise mechanisms underlying insulin-stimulated glucose transport still require investigation. Here we assessed the effect of SB203580, an inhibitor of the p38 MAP kinase family, on insulin-stimulated glucose transport in 3T3-L1 adipocytes and L6 myotubes. We found that SB203580, but not its inactive analogue (SB202474), prevented insulin-stimulated glucose transport in both cell types with an IC50 similar to that for inhibition of p38 MAP kinase (0.6 microM). Basal glucose uptake was not affected. Moreover, SB203580 added only during the transport assay did not inhibit basal or insulin-stimulated transport. SB203580 did not inhibit insulin-stimulated translocation of the glucose transporters GLUT1 or GLUT4 in 3T3-L1 adipocytes as assessed by immunoblotting of subcellular fractions or by immunofluorescence of membrane lawns. L6 muscle cells expressing GLUT4 tagged on an extracellular domain with a Myc epitope (GLUT4myc) were used to assess the functional insertion of GLUT4 into the plasma membrane. SB203580 did not affect the insulin-induced gain in GLUT4myc exposure at the cell surface but largely reduced the stimulation of glucose uptake. SB203580 had no effect on insulin-dependent insulin receptor substrate-1 phosphorylation, association of the p85 subunit of phosphatidylinositol 3-kinase with insulin receptor substrate-1, nor on phosphatidylinositol 3-kinase, Akt1, Akt2, or Akt3 activities in 3T3-L1 adipocytes. In conclusion, in the presence of SB203580, insulin caused normal translocation and cell surface membrane insertion of glucose transporters without stimulating glucose transport. We propose that insulin stimulates two independent signals contributing to stimulation of glucose transport: phosphatidylinositol 3-kinase leads to glucose transporter translocation and a pathway involving p38 MAP kinase leads to activation of the recruited glucose transporter at the membrane.


Assuntos
Adipócitos/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Imidazóis/farmacologia , Insulina/farmacologia , Proteínas Quinases Ativadas por Mitógeno , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Músculos/metabolismo , Proteínas do Tecido Nervoso , Proteínas Proto-Oncogênicas , Piridinas/farmacologia , 3-O-Metilglucose/metabolismo , Células 3T3 , Adipócitos/efeitos dos fármacos , Animais , Transporte Biológico/efeitos dos fármacos , Desoxiglucose/metabolismo , Inibidores Enzimáticos/química , Transportador de Glucose Tipo 1 , Transportador de Glucose Tipo 3 , Transportador de Glucose Tipo 4 , Imidazóis/química , Camundongos , Músculos/efeitos dos fármacos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt , Piridinas/química , Proteínas Quinases p38 Ativadas por Mitógeno
9.
J Biol Chem ; 273(43): 28322-31, 1998 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-9774456

RESUMO

The exact mechanism of the spatial organization of the insulin signaling pathway leading to nuclear events remains unknown. Here, we investigated the involvement of the actin cytoskeleton in propagation of insulin signaling events leading to DNA synthesis and expression of the immediate early genes c-fos and c-jun in L6 muscle cells. Insulin reorganized the cellular actin network and increased the rate of DNA synthesis and the levels of c-fos mRNA, but not those of c-jun mRNA, in undifferentiated L6 myoblasts. Similarly, insulin markedly elevated the levels of c-fos mRNA but not of c-jun mRNA in differentiated L6 myotubes. Disassembly of the actin filaments by cytochalasin D, latrunculin B, or botulinum C2 toxin significantly inhibited insulin-mediated DNA synthesis in myoblasts and abolished stimulation of c-fos expression by the hormone in myoblasts and myotubes. Actin disassembly abolished insulin-induced phosphorylation and activation of extracellulor signal-regulated kinases, activation of a 65-kda member of the p21-activated kinases, and phosphorylation of p38 mitogen-activated protein kinases but did not prevent activation of phosphatidylinositol 3-kinase and p70(S6k). Under these conditions, insulin-induced Ras activation was also abolished, and Grb2 association with the Src and collogen homologous (Shc) molecule was inhibited without inhibition of the tyrosine phosphorylation of Shc. We conclude that the actin filament network plays an essential role in insulin regulation of Shc-dependent signaling events governing gene expression by facilitating the interaction of Shc with Grb2.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Insulina/farmacologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Toxinas Botulínicas/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular , Células Cultivadas , Citocalasina D/farmacologia , DNA/biossíntese , Ativação Enzimática , Proteína Adaptadora GRB2 , Modelos Biológicos , Músculos/citologia , Músculos/efeitos dos fármacos , Músculos/metabolismo , Fosforilação , Ligação Proteica , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-jun/biossíntese , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Tiazóis/farmacologia , Tiazolidinas , Proteínas ras/metabolismo , Quinases da Família src/metabolismo
10.
Diabetologia ; 41(10): 1199-204, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9794108

RESUMO

Despite the important physiological role of insulin in the regulation of ionic homeostasis, primarily mediated by the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter, the intracellular signalling molecules mediating this effect of insulin have not been elucidated. Treatment of 3T3-L1 fibroblasts with insulin increased total 86Rb+ (K+) uptake from 0.8 +/- 0.04 to 1.02 +/- 0.05 nmol.mg-1.protein-1.min-1 (p < 0.005). These changes in K+ flux, though small, can alter the membrane potential. Uptake occurred through both the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter and both were stimulated by insulin. Interestingly, when bumetanide was used to inhibit the Na+/K+/2Cl- cotransporter prior to insulin action, no increase in 86Rb+ uptake via the Na+/K(+)-ATPase was observed. The structurally distinct phosphatidylinositol 3-kinase inhibitors wortmannin (50-200 nmol/l) and LY294002 (50 mumol/l) attenuated both total insulin-stimulated 86Rb+ uptake as well as uptake via the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter. Neither the inhibitor of p70.S6 kinase activation, rapamycin (30 ng/ml) nor the mitogen activated protein kinase kinase inhibitor, PD098059 (50 mumol/l), had any effect on insulin's stimulation of K+ influx. A 10 mumol/l concentration of the protein kinase C (PKC) inhibitor bisindolylmaleimide attenuated insulin action but at 1 mumol/l it was ineffective, suggesting involvement of the atypical PKC-zeta isoform. We conclude that insulin-stimulated K+ uptake in 3T3-L1 fibroblasts appears to involve concerted regulation of both the Na+/K(+)-ATPase and Na+/K+/2Cl- cotransporter and we show for the first time that this process is signalled via a pathway involving phosphatidylinositol 3-kinase and PKC-zeta.


Assuntos
Células 3T3/efeitos dos fármacos , Insulina/farmacologia , Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Potássio/metabolismo , Proteína Quinase C/metabolismo , Células 3T3/metabolismo , Androstadienos/farmacologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Isoenzimas/antagonistas & inibidores , Maleimidas/farmacologia , Camundongos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Proteína Quinase C/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas/antagonistas & inibidores , Radioisótopos de Rubídio , Sirolimo/farmacologia , Simportadores de Cloreto de Sódio-Potássio , ATPase Trocadora de Sódio-Potássio/metabolismo , Wortmanina
11.
Am J Physiol ; 275(4): E618-25, 1998 10.
Artigo em Inglês | MEDLINE | ID: mdl-9755080

RESUMO

Several studies have suggested that activation of p70 ribosomal S6 kinase (p70 S6 kinase) by insulin may be mediated by the phosphatidylinositol 3-kinase (PI 3-kinase)-Akt pathway. However, by temporal analysis of the activation of each kinase in L6 muscle cells, we report that the activation of the two serine/threonine kinases (Akt and p70 S6 kinase) can be dissociated. Insulin stimulated p70 S6 kinase in intact cells in two phases. The first phase (5 min) of stimulation was fully inhibited by wortmannin (IC50 = 20 nM) and LY-294002 (full inhibition at 5 microM). After this early inhibition, p70 S6 kinase was gradually stimulated by insulin in the presence of 100 nM wortmannin. After 30 min, the stimulation was 65% of the maximum attained in the absence of wortmannin. The IC50 of wortmannin for inhibition of this second phase was approximately 150 nM. In contrast, activation of Akt1 by insulin was completely inhibited by 100 nM wortmannin at all time points investigated. Inhibition of mitogen-activated protein kinase/extracellular signal-regulated protein kinase kinase with PD-098059 (10 microM) or treatment with the protein kinase C inhibitor bisindolylmaleimide (10 microM) had no effect on the late phase of insulin stimulation of p70 S6 kinase. We have previously shown that GLUT-1 protein synthesis in these cells is stimulated by insulin via the mTOR-p70 S6 kinase pathway, based on its sensitivity to rapamycin. We therefore investigated whether the signals leading to GLUT-1 synthesis correlated with the early or late phase of stimulation of p70 S6 kinase. GLUT-1 synthesis was not inhibited by wortmannin (100 nM). In summary, insulin activates p70 ribosomal S6 kinase in L6 muscle cells by two mechanisms, one dependent on and one independent of the activation of PI 3-kinase. In addition, activation of Akt1 is fully inhibited by wortmannin, suggesting that Akt1 does not participate in the late activation of p70 S6 kinase. Wortmannin-sensitive PI 3-kinases and Akt1 are not required for insulin stimulation of GLUT-1 protein biosynthesis.


Assuntos
Insulina/farmacologia , Músculo Esquelético/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Quinases S6 Ribossômicas/metabolismo , Androstadienos/farmacologia , Animais , Linhagem Celular , Cromonas/farmacologia , Células Clonais , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Homeostase , Indóis/farmacologia , Cinética , Maleimidas/farmacologia , Modelos Biológicos , Morfolinas/farmacologia , Proteínas Proto-Oncogênicas c-akt , Ratos , Sirolimo/farmacologia , Fatores de Tempo , Wortmanina
12.
Endocrinology ; 138(3): 1029-34, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9048605

RESUMO

vp165, a recently described member of the family of zinc-dependent membrane aminopeptidases, is a major constituent of glucose transporter-4 (GLUT4)-containing vesicles in adipocytes and skeletal muscle. Here we show that vp165 is expressed in L6 myoblasts and increases by 4.3-fold during differentiation into myotubes. The localization of vp165 in L6 myotubes was assessed by immunoblotting subcellular fractions from basal and insulin-stimulated cells and was compared to the distribution of GLUT4. vp165 and GLUT4 were mainly concentrated in the low density microsomal membranes under basal conditions. Upon stimulation with insulin, vp165 and GLUT4 were redistributed from the low density microsomes to the plasma membrane. The majority of vp165 was found in immunoisolated GLUT4-containing vesicles, and vice versa, the majority of GLUT4 was detected in immunoisolated vp165-containing vesicles. In contrast, the two other glucose transporter isoforms expressed in L6, GLUT1 and GLUT3, were excluded from GLUT4- and vp165-containing vesicles. These results suggest that in rat skeletal muscle cells, vp165 and GLUT4 cosegregate to the same intracellular compartment and that this is distinct from the compartment containing GLUT1 and GLUT3.


Assuntos
Aminopeptidases/metabolismo , Insulina/fisiologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Cistinil Aminopeptidase , Transportador de Glucose Tipo 4 , Microssomos/metabolismo , Músculo Esquelético/citologia , Ratos , Frações Subcelulares/metabolismo
13.
Diabetes ; 45(11): 1516-23, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8866555

RESUMO

Insulin-stimulated glucose uptake in skeletal muscle is mediated through the GLUT4 glucose transporter. Transgenic (TG) mice overexpressing human GLUT4 in skeletal muscle show an increased ability to handle a glucose load. Here, the participation of the overexpressed GLUT4 in the response to insulin was examined. In TG mouse muscle, the GLUT4 protein content was 10-fold higher in crude membrane (CM), sevenfold higher in internal membrane (IM), and 15-fold higher in a plasma membrane (PM)-rich fraction, relative to non-TG littermates. This suggested partial saturation of the normal sorting mechanisms. The distribution and abundance of the GLUT1 glucose transporter was not affected. Insulin injection (4.3 U/kg body wt) increased GLUT4 in the PM-rich fraction; the increase was threefold higher in TG than in non-TG mice. Insulin decreased the GLUT4 content of the IM in both animal groups and of a second, heavier intracellular membrane fraction only in TG mice. The net content of Na+-K+-pump subunits was 40-65% lower in CM from TG compared with non-TG littermates. In spite of this, insulin caused a three- to sixfold higher translocation of the alpha2 and beta1 subunits of the Na+-K+-pump in TG compared with non-TG animals. The results suggest that overexpression of GLUT4 confers to the muscle increased ability to translocate subunits of the Na+-K+-pump either as a direct consequence of the recruitment of glucose transporters or as an adaptation to the more demanding metabolic state.


Assuntos
Insulina/farmacologia , Proteínas de Transporte de Monossacarídeos/biossíntese , Proteínas Musculares , Músculo Esquelético/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Glicemia/metabolismo , Membrana Celular/metabolismo , Expressão Gênica/efeitos dos fármacos , Técnica Clamp de Glucose , Transportador de Glucose Tipo 4 , Humanos , Substâncias Macromoleculares , Camundongos , Camundongos Transgênicos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Músculo Esquelético/efeitos dos fármacos , Valores de Referência , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA