Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetes ; 60(11): 2883-91, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21933986

RESUMO

OBJECTIVE: Appropriate regulation of insulin receptor substrate 2 (IRS-2) expression in pancreatic ß-cells is essential to adequately compensate for insulin resistance. In liver, basal IRS-2 expression is controlled via a temporal negative feedback of sterol regulatory element-binding protein 1 (SREBP-1) to antagonize transcription factors forkhead box class O (FoxO)1/FoxO3a at an insulin response element (IRE) on the IRS-2 promoter. The purpose of the study was to examine if a similar mechanism controlled IRS-2 expression in ß-cells. RESEARCH DESIGN AND METHODS: IRS-2 mRNA and protein expression, as well as IRS-2 gene promoter activity, were examined in isolated rat islets. Specific transcription factor association with the IRE on the IRS-2 promoter was examined by chromatin immunoprecipitation (ChIP) assay, and their nuclear translocation was examined by immunofluorescence. A direct in vivo effect of insulin on control of IRS-2 expression in liver and pancreatic islets was also investigated. RESULTS: In IRS-2 promoter-reporter assays conducted in isolated islets, removal of the IRE decreased basal IRS-2 promoter activity in ß-cells up to 80%. Activation of IRS signaling in isolated rat islets by insulin/IGF-I (used as an experimental in vitro tool) or downstream constitutive activation of protein kinase B (PKB) significantly decreased IRS-2 expression. In contrast, inhibition of phosphatidylinositol 3-kinase (PI3K) or PKB significantly increased IRS-2 levels in ß-cells. ChIP assays indicated that transcription factors FoxO1 and FoxO3a associated with the IRE on the IRS-2 promoter in ß-cells in a PI3K/PKB-dependent manner, whereas others, such as SREBP-1, the transcription factor binding to immunoglobulin heavy chain enhancer 3', and the aryl hydrocarbon receptor nuclear translocator (ARNT), did not. However, only FoxO3a, not FoxO1, was capable of driving IRS-2 promoter activity via the IRE in ß-cells. In vivo studies showed insulin was able to suppress IRS-2 expression via activation of SREBP-1 in the liver, but this mechanism was not apparent in pancreatic islets from the same animal. CONCLUSIONS: The molecular mechanism for feedback control of IRS signaling to decrease IRS-2 expression in liver and ß-cells is quite distinct, with a predominant role played by FoxO3a in ß-cells.


Assuntos
Retroalimentação Fisiológica , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/genética , Humanos , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Células Secretoras de Insulina/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Regiões Promotoras Genéticas , Transporte Proteico , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Elementos de Resposta , Técnicas de Cultura de Tecidos
2.
Diabetes ; 60(11): 2892-902, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21940781

RESUMO

OBJECTIVE: Insulin receptor substrate-2 (IRS-2) plays an essential role in pancreatic islet ß-cells by promoting growth and survival. IRS-2 turnover is rapid in primary ß-cells, but its expression is highly regulated at the transcriptional level, especially by glucose. The aim was to investigate the molecular mechanism on how glucose regulates IRS-2 gene expression in ß-cells. RESEARCH DESIGN AND METHODS: Rat islets were exposed to inhibitors or subjected to adenoviral vector-mediated gene manipulations and then to glucose-induced IRS-2 expression analyzed by real-time PCR and immunoblotting. Transcription factor nuclear factor of activated T cells (NFAT) interaction with IRS-2 promoter was analyzed by chromatin immunoprecipitation assay and glucose-induced NFAT translocation by immunohistochemistry. RESULTS: Glucose-induced IRS-2 expression occurred in pancreatic islet ß-cells in vivo but not in liver. Modulating rat islet ß-cell Ca(2+) influx with nifedipine or depolarization demonstrated that glucose-induced IRS-2 gene expression was dependent on a rise in intracellular calcium concentration derived from extracellular sources. Calcineurin inhibitors (FK506, cyclosporin A, and a peptide calcineurin inhibitor [CAIN]) abolished glucose-induced IRS-2 mRNA and protein levels, whereas expression of a constitutively active calcineurin increased them. Specific inhibition of NFAT with the peptide inhibitor VIVIT prevented a glucose-induced IRS-2 transcription. NFATc1 translocation to the nucleus in response to glucose and association of NFATc1 to conserved NFAT binding sites in the IRS-2 promoter were demonstrated. CONCLUSIONS: The mechanism behind glucose-induced transcriptional control of IRS-2 gene expression specific to the islet ß-cell is mediated by the Ca(2+)/calcineurin/NFAT pathway. This insight into the IRS-2 regulation could provide novel therapeutic means in type 2 diabetes to maintain an adequate functional mass.


Assuntos
Calcineurina/metabolismo , Sinalização do Cálcio , Regulação da Expressão Gênica , Hiperglicemia/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição NFATC/metabolismo , Animais , Inibidores de Calcineurina , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Substratos do Receptor de Insulina/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/ultraestrutura , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/antagonistas & inibidores , Especificidade de Órgãos , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Técnicas de Cultura de Tecidos
3.
J Biol Chem ; 281(23): 15884-92, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16574657

RESUMO

Insulin receptor substrate 2 (IRS-2) plays a critical role in pancreatic beta-cells. Increased IRS-2 expression promotes beta-cell growth and survival, whereas decreased IRS-2 levels lead to apoptosis. It was found that IRS-2 turnover in rat islet beta-cells was rapid, with mRNA and protein half-lives of approximately 90 min and approximately 2 h, respectively. However, this was countered by specific glucose-regulated IRS-2 expression mediated at the transcriptional level. Glucose (> or = 6 mM) increased IRS-2 mRNA and protein levels in a dose-dependent manner, reaching a maximum 4-fold increase in IRS-2 mRNA and a 5-6-fold increase in IRS-2 protein levels at > or = 12 mM glucose (p < or = 0.01). Glucose (15 mM) regulation of islet beta-cell IRS-2 gene expression was rapid, with a significant increase in IRS-2 mRNA levels within 2 h that reached a maximum 4-fold increase by 4 h. IRS-2 protein expression in beta-cells followed that of IRS-2 mRNA. Glucose metabolism was necessary for increased IRS-2 expression in beta-cells. Moreover, inhibition of a glucose-induced rise in islet beta-cell cytosolic [Ca2+]i prevented an increase in IRS-2 expression, indicating this was Ca2+-dependent. The glucose-induced rise in IRS-2 levels correlated with increased IRS-2 tyrosine phosphorylation and downstream activation of protein kinase B. These data indicate that fluctuations of glucose in the normal physiological range (5-15 mM) promote beta-cell survival via regulation of IRS-2 expression and a subsequent parallel protein kinase B activation. Given that the onset of type-2 diabetes is marked by loss of beta-cells, these data further the idea that controlled IRS-2 expression in beta-cells could be a therapeutic means to promote beta-cell survival and delay the onset of the disease.


Assuntos
Glucose/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Imunofluorescência , Imunoprecipitação , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Fosfoproteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Transcrição Gênica
4.
J Biol Chem ; 280(3): 2282-93, 2005 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-15537654

RESUMO

Regulation of insulin receptor substrate (IRS)-2 expression is critical to beta-cell survival, but the mechanisms that control this are complex and undefined. Here in pancreatic beta-cells (INS-1), chronic exposure (>8 h) to 15 mm glucose and/or 5 nm IGF-1, increased Ser/Thr phosphorylation of IRS-2, which correlated with decreased IRS-2 levels. This glucose/IGF-1-induced decrease in IRS-2 levels was prevented by the proteasomal inhibitor, lactacystin. In addition, the glucose/IGF-1-induced increase in Ser/Thr phosphorylation of IRS-2 and the subsequent decrease in INS-1 cell IRS-2 protein levels was thwarted by the mammalian target of rapamycin(mTOR) inhibitor, rapamycin. Moreover, adenoviral-mediated expression of constitutively active mTOR (mTORDelta) further increased glucose/IGF-1-induced Ser/Thr phosphorylation of IRS-2 and decreased IRS-2 protein levels, whereas adenoviral-mediated expression of "kinase-dead" mTOR (mTOR-KD) conversely reduced Ser/Thr phosphorylation of IRS-2 and maintained IRS-2 protein levels. In adenoviral-infected beta-cells expressing mTORDelta, the decrease in IRS-2 protein levels was also prevented by rapamycin or lactacystin, further indicating a proteasomal mediated degradation of IRS-2 mediated via mTOR-induced Ser/Thr phosphorylation of IRS-2. Finally, we found that chronic activation of mTOR leading to decreased levels of IRS-2 in INS-1 cells led to a significant decrease in PKB activation and consequently increased beta-cell apoptosis. Thus, chronic activation of mTOR by glucose (and/or IGF-1) in beta-cells leads to increased Ser/Thr phosphorylation of IRS-2 that targets it for proteasomal degradation, resulting in decreased IRS-2 expression and increased beta-cell apoptosis. This may be a contributing mechanism as to how beta-cell mass is decreased by chronic hyperglycemia in the pathogenesis of type-2 diabetes.


Assuntos
Fosfoproteínas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/patologia , Ensaio de Desvio de Mobilidade Eletroforética , Imunofluorescência , Glucose/administração & dosagem , Hidrólise , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/administração & dosagem , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/metabolismo , Mamíferos , Fosforilação
5.
Mol Cell Endocrinol ; 209(1-2): 17-31, 2003 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-14604813

RESUMO

IRS-2 plays a pivotal role in the control of pancreatic beta-cell growth. Here, the effect of altering IRS-2 expression levels in the pancreatic beta-cell line, INS-1, was examined. Adenoviral-mediated increased in IRS-2 protein levels protected against fatty acid (FFA)-induced apoptosis, associated with increased activation of PKB and decreased levels of activated caspase-9. Conversely, decreasing endogenous IRS-2 in INS-1 cells, using adenoviral-mediated expression of IRS-2 antisense, caused a three-fold increase in baseline apoptosis that was further enhanced in the presence of FFA. This was associated with decreased activation of PKB and increased caspase-9 activation. Although IRS-4 is not normally expressed in beta-cells, it was found that adenoviral-mediated introduction of IRS-4 into INS-1 cells enhanced glucose/IGF-1 induced mitogenesis, and protected against FFA-induced apoptosis, similarly to IRS-2. Moreover, expression of IRS-4 in INS-1 cells depleted of IRS-2 levels by IRS-2 antisense, was able to compensate for the lack of IRS-2 and reduce apoptosis in these cells back to normal. Thus, in beta-cells IRS-4 and -2 have similar biological functions. Also, this study further emphasizes the importance of IRS-2 signaling in control of beta-cell survival.


Assuntos
Apoptose , Ilhotas Pancreáticas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Adenoviridae/genética , Animais , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Ativação Enzimática , Ácidos Graxos não Esterificados/toxicidade , Vetores Genéticos , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Ilhotas Pancreáticas/citologia , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Transdução de Sinais
6.
J Biol Chem ; 278(43): 42080-90, 2003 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-12928442

RESUMO

Proinsulin biosynthesis is regulated in response to nutrients, most notably glucose. In the short term (/=10-fold). Importantly, neither exogenously added nor secreted insulin were found to play any role in regulating insulin secretion, proinsulin translation, preproinsulin mRNA levels, or total protein synthesis. The results presented here indicate that long term nutritional state sets the preproinsulin mRNA level in the beta-cell at which translation control regulates short term changes in rates of proinsulin biosynthesis in response to glucose, but this is not mediated by any autocrine effect of insulin.


Assuntos
Retroalimentação Fisiológica , Glucose/farmacologia , Insulina/farmacologia , Ilhotas Pancreáticas/metabolismo , Proinsulina/biossíntese , Proinsulina/genética , Biossíntese de Proteínas/efeitos dos fármacos , Precursores de Proteínas/genética , Animais , Insulina/metabolismo , Secreção de Insulina , Masculino , Estado Nutricional/fisiologia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
7.
Diabetes ; 52(4): 974-83, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12663469

RESUMO

Glucose can activate the mitogen-activated kinases, Erk-1/2, and the ribosomal-S6 kinase, p70(S6K), in beta-cells, contributing to an increase in mitogenesis. However, the signaling mechanism by which glucose induces Erk-1/2 and p70(S6K) phosphorylation activation is undefined. Increased glucose metabolism increases [Ca(2+)](i) and [cAMP], and it was investigated if these secondary signals were linked to glucose-induced Erk-1/2 and p70(S6K) activation in pancreatic beta-cells. Blocking Ca(2+) influx with verapamil, or inhibiting protein kinase A (PKA) with H89, prevented glucose-induced Erk-1/2 phosphorylation. Increasing cAMP levels by GLP-1 potentiated glucose-induced Erk-1/2 phosphorylation via PKA activation. Elevation of [Ca(2+)](i) by glyburide potentiated Erk-1/2 phosphorylation, which was also inhibited by H89, suggesting increased [Ca(2+)](i) preceded PKA for glucose-induced Erk-1/2 activation. Adenoviral-mediated expression of dominant negative Ras in INS-1 cells decreased IGF-1-induced Erk-1/2 phosphorylation but had no effect on that by glucose. Collectively, our study indicates that a glucose-induced rise in [Ca(2+)](i) leads to cAMP-induced activation of PKA that acts downstream of Ras and upstream of the MAP/Erk kinase, MEK, to mediate Erk-1/2 phosphorylation via phosphorylation activation of Raf-1. In contrast, glucose-induced p70(S6K) activation, in the same beta-cells, was mediated by a distinct signaling pathway independent of Ca(2+)/cAMP, most likely via mTOR-kinase acting as an "ATP-sensor."


Assuntos
Ativação Enzimática/efeitos dos fármacos , Glucose/farmacologia , Ilhotas Pancreáticas/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sulfonamidas , Animais , Cálcio/metabolismo , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Glucagon , Peptídeo 1 Semelhante ao Glucagon , Peptídeos Semelhantes ao Glucagon , Glibureto/farmacologia , Humanos , Isoquinolinas/farmacologia , Masculino , Proteína Quinase 3 Ativada por Mitógeno , Fragmentos de Peptídeos/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-raf/fisiologia , Ratos , Ratos Sprague-Dawley , Verapamil/farmacologia
8.
J Biol Chem ; 277(51): 49676-84, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12393870

RESUMO

Free fatty acids (FFA) have been reported to reduce pancreatic beta-cell mitogenesis and to increase apoptosis. Here we show that the FFA, oleic acid, increased apoptosis 16-fold in the pancreatic beta-cell line, INS-1, over a 18-h period as assessed by Hoechst 33342/propidium iodide staining and caspase-3 and -9 activation, with negligible necrosis. A parallel analysis of the phosphorylation activation of protein kinase B (PKB) showed this was reduced in the presence of FFA that correlated with the incidence of apoptosis. At stimulatory 15 mm glucose and/or in the added presence of insulin-like growth factor 1, FFA-induced beta-cell apoptosis was lessened compared with that at a basal 5 mm glucose. However, most strikingly, adenoviral mediated expression of a constitutively active PKB, but not a "kinase-dead" PKB variant, essentially prevented FFA-induced beta-cell apoptosis under all glucose/insulin-like growth factor 1 conditions. Further analysis of pro-apoptotic downstream targets of PKB, implicated a role for PKB-mediated phosphorylation inhibition of glycogen synthase kinase-3alpha/beta and the forkhead transcription factor, FoxO1, in protection of FFA-induced beta-cell apoptosis. In addition, down-regulation of the pro-apoptotic tumor suppressor protein, p53, via PKB-mediated phosphorylation of MDM2 might also play a role in partially protecting beta-cells from FFA-induced apoptosis. Adenoviral mediated expression of wild type p53 potentiated FFA-induced beta-cell apoptosis, whereas expression of a dominant negative p53 partly inhibited beta-cell apoptosis by approximately 50%. Hence, these data demonstrate that PKB activation plays an important role in promoting pancreatic beta-cell survival in part via inhibition of the pro-apoptotic proteins glycogen synthase kinase-3alpha/beta, FoxO1, and p53. This, in turn, provides novel insight into the mechanisms involved in FFA-induced beta-cell apoptosis.


Assuntos
Apoptose , Ácidos Graxos/farmacologia , Ilhotas Pancreáticas/patologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Adenoviridae/genética , Animais , Caspase 3 , Caspase 9 , Caspases/metabolismo , Sobrevivência Celular , Relação Dose-Resposta a Droga , Regulação para Baixo , Ativação Enzimática , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead , Glucose/metabolismo , Glucose/farmacologia , Immunoblotting , Fator de Crescimento Insulin-Like I/metabolismo , Necrose , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt , Ratos , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo
9.
J Biol Chem ; 277(27): 24232-42, 2002 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-11978799

RESUMO

The specific biochemical steps required for glucose-regulated insulin exocytosis from beta-cells are not well defined. Elevation of glucose leads to increases in cytosolic [Ca2+]i and biphasic release of insulin from both a readily releasable and a storage pool of beta-granules. The effect of elevated [Ca2+]i on phosphorylation of isolated beta-granule membrane proteins was evaluated, and the phosphorylation of four proteins was found to be altered by [Ca2+]i. One (a 18/20-kDa doublet) was a Ca2+-dependent increase in phosphorylation, and, surprisingly, three others (138, 42, and 36 kDa) were Ca2+-dependent dephosphorylations. The 138-kDa beta-granule phosphoprotein was found to be kinesin heavy chain (KHC). At low levels of [Ca2+]i KHC was phosphorylated by casein kinase 2, but KHC was rapidly dephosphorylated by protein phosphatase 2B beta (PP2Bbeta) as [Ca2+]i increased. Inhibitors of PP2B specifically reduced the second, microtubule-dependent, phase of insulin secretion, suggesting that dephosphorylation of KHC was required for transport of beta-granules from the storage pool to replenish the readily releasable pool of beta-granules. This is distinct from synaptic vesicle exocytosis, because neurotransmitter release from synaptosomes did not require a Ca2+-dependent KHC dephosphorylation. These results suggest a novel mechanism for regulating KHC function and beta-granule transport in beta-cells that is mediated by casein kinase 2 and PP2B. They also implicate a novel regulatory role for PP2B/calcineurin in the control of insulin secretion downstream of a rise in [Ca2+]i.


Assuntos
Cálcio/farmacologia , Grânulos Citoplasmáticos/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Cinesinas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/fisiologia , Caseína Quinase II , Linhagem Celular , Ácido Egtázico/farmacologia , Exocitose , Secreção de Insulina , Insulinoma , Cinética , Neoplasias Pancreáticas , Fosfatos/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Subunidades Proteicas , Transporte Proteico , Ratos , Células Tumorais Cultivadas
10.
Diabetes ; 51(3): 662-8, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11872664

RESUMO

Chronic elevations in plasma levels of fatty acids (FAs) adversely affect pancreatic beta-cell function in type 2 diabetes. In vitro, we have previously shown that deleterious effects of prolonged exposure of isolated islets to FAs were dependent on the presence of elevated glucose concentration. This led us to hypothesize that both hyperlipidemia and hyperglycemia must be present simultaneously for FAs to affect beta-cell function. To test this hypothesis in vivo, we administered a high-fat diet for 6 weeks to Goto-Kakizaki (GK) rats. High-fat feeding had no effect on insulin secretion, insulin content, or insulin mRNA levels in islets from normoglycemic Wistar rats. In contrast, high-fat feeding markedly impaired glucose-induced insulin secretion in islets from GK rats. High-fat feeding did not affect triglyceride (TG) content or the rate of glucose oxidation in islets. It was, however, accompanied by a twofold increase in uncoupling protein (UCP)-2 levels in GK rat islets. Insulin treatment completely normalized glucose-induced insulin secretion and prevented the increase in UCP-2 expression in islets from high-fat-fed GK rats. We conclude that hyperlipidemia induced by high-fat feeding affects insulin secretion in islets from hyperglycemic GK rats only, by a mechanism which may involve, at least in part, modulation of UCP-2 expression.


Assuntos
Glicemia/análise , Hiperglicemia/fisiopatologia , Hiperlipidemias/fisiopatologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas de Membrana Transportadoras , Proteínas Mitocondriais , Animais , Gorduras na Dieta/administração & dosagem , Ácidos Graxos/sangue , Glucose/metabolismo , Glucose/farmacologia , Gliceraldeído/farmacologia , Hiperlipidemias/etiologia , Insulina/administração & dosagem , Insulina/genética , Secreção de Insulina , Canais Iônicos , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/efeitos dos fármacos , Oxirredução , Cloreto de Potássio/farmacologia , Proteínas/análise , RNA Mensageiro/análise , Ratos , Ratos Wistar , Succinatos/farmacologia , Triglicerídeos/análise , Proteína Desacopladora 2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...