Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Int J Mol Sci ; 21(11)2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32492936

RESUMO

Chronic exposure of pancreatic ß-cells to elevated nutrient levels impairs their function and potentially induces apoptosis. Like in other cell types, AMPK is activated in ß-cells under conditions of nutrient deprivation, while little is known on AMPK responses to metabolic stresses. Here, we first reviewed recent studies on the role of AMPK activation in ß-cells. Then, we investigated the expression profile of AMPK pathways in ß-cells following metabolic stresses. INS-1E ß-cells and human islets were exposed for 3 days to glucose (5.5-25 mM), palmitate or oleate (0.4 mM), and fructose (5.5 mM). Following these treatments, we analyzed transcript levels of INS-1E ß-cells by qRT-PCR and of human islets by RNA-Seq; with a special focus on AMPK-associated genes, such as the AMPK catalytic subunits α1 (Prkaa1) and α2 (Prkaa2). AMPKα and pAMPKα were also evaluated at the protein level by immunoblotting. Chronic exposure to the different metabolic stresses, known to alter glucose-stimulated insulin secretion, did not change AMPK expression, either in insulinoma cells or in human islets. Expression profile of the six AMPK subunits was marginally modified by the different diabetogenic conditions. However, the expression of some upstream kinases and downstream AMPK targets, including K-ATP channel subunits, exhibited stress-specific signatures. Interestingly, at the protein level, chronic fructose treatment favored fasting-like phenotype in human islets, as witnessed by AMPK activation. Collectively, previously published and present data indicate that, in the ß-cell, AMPK activation might be implicated in the pre-diabetic state, potentially as a protective mechanism.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Apoptose , Regulação Enzimológica da Expressão Gênica , Ilhotas Pancreáticas/enzimologia , Adulto , Animais , Glicemia/análise , Feminino , Frutose/metabolismo , Perfilação da Expressão Gênica , Homeostase , Humanos , Insulina/metabolismo , Insulinoma/enzimologia , Masculino , Pessoa de Meia-Idade , Ácido Oleico/análise , Ácido Palmítico/análise , Fenótipo , RNA-Seq , Ratos , Estresse Fisiológico
2.
J Biol Chem ; 293(37): 14210-14223, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30012886

RESUMO

Human islet amyloid polypeptide (hIAPP) is the principal constituent of amyloid deposits and toxic oligomers in the pancreatic islets. Together with hyperglycemia, hIAPP-derived oligomers and aggregates are important culprits in type 2 diabetes mellitus (T2DM). Here, we explored the role of the cell's main proteolytic complex, the proteasome, in hIAPP turnover in normal and stressed ß-cells evoked by chronic hyperglycemia. Moderate inhibition (10-35%) of proteasome activity/function in cultured human islets by the proteasome inhibitor lactacystin enhanced intracellular accumulation of hIAPP. Unexpectedly, prolonged (>1 h) and marked (>50%) impairment of proteasome activity/function had a strong inhibitory effect on hIAPP transcription and secretion from normal and stressed ß-cells. This negative compensatory feedback mechanism for controlling IAPP turnover was also observed in the lactacystin-treated rat insulinoma ß-cell line (INS 832/13), demonstrating the presence of an evolutionarily conserved mechanism for IAPP production. In line with these in situ studies, our current ex vivo data showed that proteasome activity and hIAPP expression are also down-regulated in islets isolated from T2DM subjects. Gene expression and promoter activity studies demonstrated that the functional proteasome complex is required for efficient activation of the hIAPP promoter and for full expression of IAPP's essential transcription factor, FOXA2. ChIP studies revealed that promoter occupancy of FoxA2 at the rat IAPP promoter region is an important and limiting factor for amylin expression in proteasome-impaired murine cells. This study suggests a novel regulatory pathway in ß-cells involving proteasome, FOXA2, and IAPP, which can be possibly targeted to regulate hIAPP levels and islet amyloidosis in T2DM.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/patologia , Regulação para Baixo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Células Secretoras de Insulina/enzimologia , Insulinoma/enzimologia , Insulinoma/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Leupeptinas/farmacologia , Camundongos , Oligopeptídeos/farmacologia , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Inibidores de Proteassoma/farmacologia , Ratos
3.
J Cell Biol ; 217(4): 1287-1301, 2018 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-29507125

RESUMO

In mammalian pancreatic ß cells, the IRE1α-XBP1 pathway is constitutively and highly activated under physiological conditions. To elucidate the precise role of this pathway, we constructed ß cell-specific Ire1α conditional knockout (CKO) mice and established insulinoma cell lines in which Ire1α was deleted using the Cre-loxP system. Ire1α CKO mice showed the typical diabetic phenotype including impaired glycemic control and defects in insulin biosynthesis postnatally at 4-20 weeks. Ire1α deletion in pancreatic ß cells in mice and insulinoma cells resulted in decreased insulin secretion, decreased insulin and proinsulin contents in cells, and decreased oxidative folding of proinsulin along with decreased expression of five protein disulfide isomerases (PDIs): PDI, PDIR, P5, ERp44, and ERp46. Reconstitution of the IRE1α-XBP1 pathway restored the proinsulin and insulin contents, insulin secretion, and expression of the five PDIs, indicating that IRE1α functions as a key regulator of the induction of catalysts for the oxidative folding of proinsulin in pancreatic ß cells.


Assuntos
Endorribonucleases/metabolismo , Células Secretoras de Insulina/enzimologia , Insulina/metabolismo , Proinsulina/metabolismo , Dobramento de Proteína , Proteínas Serina-Treonina Quinases/metabolismo , Proteína 1 de Ligação a X-Box/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Animais , Sítios de Ligação , Glicemia/metabolismo , Linhagem Celular Tumoral , Diabetes Mellitus/sangue , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Endorribonucleases/deficiência , Endorribonucleases/genética , Insulina/genética , Insulinoma/enzimologia , Insulinoma/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Knockout , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Oxirredução , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Fosforilação , Proinsulina/química , Proinsulina/genética , Regiões Promotoras Genéticas , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Proteína 1 de Ligação a X-Box/genética , eIF-2 Quinase/metabolismo
4.
Cell Signal ; 38: 212-222, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28743549

RESUMO

Apoptotic death of pancreatic ß cells is a major cause of type 2 diabetes mellitus (T2D) progression. Two isoforms of pyruvate kinase, PKM1 and PKM2, have been reported to participate in cell death in several cell types; however, little is known about their causal pathways in pancreatic ß-cell death. We examined whether the suppression of PKM1 or PKM2 affects endoplasmic reticulum (ER) stress-induced apoptosis in a pancreatic ß-cell line, MIN6, and Beta-TC-6 and found that knockdown of PKM1, but not of PKM2, leads to the induction of ER stress-induced apoptosis in these cells. We also investigated the mechanism by which PKM1 inhibits ER stress-induced apoptosis. We confirmed that PKM1 interacts with A-Raf, an upstream regulator of the MEK/ERK pathway, and that this interaction contributes to MEK1 phosphorylation by A-Raf. PKM1 knockdown suppresses the phosphorylation of MEK, ERK, and caspase-9 (Thr125), which is phosphorylated by the MEK/ERK pathway, thereby inhibiting the cleavage and activation of caspase-9. Thus, PKM1 knockdown activates the caspase-9/caspase-3 pathway under ER stress conditions and leads to apoptosis.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Estresse do Retículo Endoplasmático , Insulinoma/enzimologia , Insulinoma/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas A-raf/metabolismo , Hormônios Tireóideos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Ácido Pirúvico/metabolismo , eIF-2 Quinase/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
5.
Biochem Biophys Res Commun ; 461(3): 549-54, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25912139

RESUMO

Pro-inflammatory cytokines play a crucial role in the destruction of pancreatic ß-cells, thereby triggering the development of autoimmune diabetes mellitus. We recently developed a cell-permeable fusion protein, PEP-1-heme oxygenase-1 (PEP-1-HO-1) and investigated the anti-inflammatory effects in macrophage cells. In this study, we transduced PEP-1-HO-1 into INS-1 insulinoma cells and examined its protective effect against cytokine-induced cell death. PEP-1-HO-1 was successfully delivered into INS-1 cells in time- and dose-dependent manner and was maintained within the cells for at least 48 h. Pre-treatment with PEP-1-HO-1 increased the survival of INS-1 cells exposed to cytokine mixture (IL-1ß, IFN-γ, and TNF-α) in a dose-dependent manner. PEP-1-HO-1 markedly decreased cytokine-induced production of reactive oxygen species (ROS), nitric oxide (NO), and malondialdehyde (MDA). These protective effects of PEP-1-HO-1 against cytokines were correlated with the changes in the levels of signaling mediators of inflammation (iNOS and COX-2) and cell apoptosis/survival (Bcl-2, Bax, caspase-3, PARP, JNK, and Akt). These results showed that the transduced PEP-1-HO-1 efficiently prevented cytokine-induced cell death of INS-1 cells by alleviating oxidative/nitrosative stresses and inflammation. Further, these results suggested that PEP-1-mediated HO-1 transduction may be a potential therapeutic strategy to prevent ß-cell destruction in patients with autoimmune diabetes mellitus.


Assuntos
Morte Celular/fisiologia , Citocinas/fisiologia , Heme Oxigenase-1/genética , Insulina/biossíntese , Proteínas Recombinantes de Fusão/genética , Transdução Genética , Animais , Linhagem Celular Tumoral , Insulinoma/enzimologia , Insulinoma/metabolismo , Insulinoma/patologia , Malondialdeído/metabolismo , Nitritos/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
6.
Metabolism ; 63(1): 120-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24140090

RESUMO

OBJECTIVE: Exendin-4 (Ex-4) is an anti-diabetic drug that is a potent agonist of the glucagon-like peptide-1 (GLP-1) receptor. It has already been approved for the treatment of type 2 diabetes mellitus, but its underlying mechanisms of action are not fully understood. Calcium/calmodulin-dependent serine protein kinase (CASK), which plays a vital role in the transport and release of neurotransmitters in neurons, is expressed in pancreatic islet cells and ß-cells. This study aimed to investigate whether CASK is involved in the insulin secretagogue action induced by Ex-4 in INS-1 cells. MATERIAL/METHODS: A glucose-stimulated insulin secretion (GSIS) assay was performed with or without siRNA treatment against CASK. The expression level and location of CASK were evaluated by real-time PCR, western blotting and immunofluorescence. With the use of a protein kinase A (PKA) inhibitor or an exchange protein directly activated by cAMP-2 (Epac2) agonist, immunoblotting was performed to establish the signaling pathway through which Ex-4 alters CASK expression. RESULTS: Knock-down of CASK significantly attenuated the Ex-4-enhanced insulin release, and we showed that Ex-4 could increase transcription of CASK mRNA and expression of CASK protein but did not change the cellular location of CASK. A PKA inhibitor reduced the ability of Ex-4 to stimulate CASK expression, but an Epac2 agonist had no effect suggesting that regulation was mediated by the cAMP/PKA pathway. CONCLUSION: Our study suggests that the stimulation of ß-cell insulin secretion by Ex-4 is mediated, at least in part, by CASK via a novel signaling mechanism.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Guanilato Quinases/metabolismo , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Insulinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Peptídeos/farmacologia , Peçonhas/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/tratamento farmacológico , Exenatida , Técnicas de Silenciamento de Genes , Peptídeo 1 Semelhante ao Glucagon/agonistas , Guanilato Quinases/genética , Secreção de Insulina , Insulinoma/enzimologia , Microscopia de Fluorescência , Neoplasias Pancreáticas/enzimologia , RNA Interferente Pequeno/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais
7.
Islets ; 5(3): 122-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23959334

RESUMO

AIMS/HYPOTHESIS: Protein gene product 9.5 (PGP 9.5) is a marker for neuroendocrine cells but has not been used for pancreatic islet cells and pancreatic endocrine tumors (PETs). Antibodies for PGP 9.5 are now commercially available for immunocytochemical study, with which immunostaining may be able to differentiate between benign and malignant PETs. RESULTS: All 4 kinds of normal islet cells were positively immunostained for PGP 9.5-moderately positive for ß-cells and strongly positive for δ-cells, whereas ganglion cells were immunostained more strongly than islet cells. Nine of 12 insulinomas were moderately to strongly positive for PGP 9.5. Two glucagonomas, 3 of 6 pancreatic polypeptidomas (PPomas), 3 of 9 gastrinomas, and 2 of 4 non-functioning PETs were negative for PGP 9.5. MATERIALS AND METHODS: Thirty-four PETs were immunocytochemically stained for PGP 9.5 using a rabbit polyclonal antibody together with immunostaining for 4 pancreatic hormones, chromogranin A (CgA), and gastrin. PETs consisted of 12 insulinomas, 2 glucagonomas, 1 somatostatinoma (SRIFoma), 6 PPomas, 9 gastrinomas, and 4 non-functioning PETs. CONCLUSION/INTERPRETATION: PGP 9.5 immunostaining was universally positive for 4 kinds of islet cells and was moderately to strongly positive for 9 of 12 (75%) insulinomas. All 22 non-ß-cell PETs were negative or weakly positive for PGP 9.5, and thus negative or weakly positive PGP 9.5 immunostaining may be used as a marker for potential malignancy and poor prognosis for non-ß-cell PETs.


Assuntos
Proteínas de Neoplasias/metabolismo , Células Neuroendócrinas/metabolismo , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Ubiquitina Tiolesterase/metabolismo , Biomarcadores/metabolismo , Gânglios/metabolismo , Células Secretoras de Glucagon/enzimologia , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/patologia , Humanos , Imuno-Histoquímica , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Insulinoma/enzimologia , Insulinoma/metabolismo , Insulinoma/patologia , Gradação de Tumores , Proteínas do Tecido Nervoso/metabolismo , Células Neuroendócrinas/enzimologia , Células Neuroendócrinas/patologia , Pâncreas/inervação , Pâncreas/patologia , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia
8.
Oxid Med Cell Longev ; 2012: 989372, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23213352

RESUMO

Group VIB Phospholipase A(2) (iPLA(2)γ) is distributed in membranous organelles in which ß-oxidation occurs, that is, mitochondria and peroxisomes, and is expressed by insulin-secreting pancreatic islet ß-cells and INS-1 insulinoma cells, which can be injured by inflammatory cytokines, for example, IL-1ß and IFN-γ, and by oxidants, for example, streptozotocin (STZ) or t-butyl-hydroperoxide (TBHP), via processes pertinent to mechanisms of ß-cell loss in types 1 and 2 diabetes mellitus. We find that incubating INS-1 cells with IL-1ß and IFN-γ, with STZ, or with TBHP causes increased expression of iPLA(2)γ mRNA and protein. We prepared INS-1 knockdown (KD) cell lines with reduced iPLA(2)γ expression, and they proliferate more slowly than control INS-1 cells and undergo increased membrane peroxidation in response to cytokines or oxidants. Accumulation of oxidized phospholipid molecular species in STZ-treated INS-1 cells was demonstrated by LC/MS/MS scanning, and the levels in iPLA(2)γ-KD cells exceeded those in control cells. iPLA(2)γ-KD INS-1 cells also exhibited higher levels of apoptosis than control cells when incubated with STZ or with IL-1ß and IFN-γ. These findings suggest that iPLA(2)γ promotes ß-cell proliferation and that its expression is increased during inflammation or oxidative stress as a mechanism to mitigate membrane injury that may enhance ß-cell survival.


Assuntos
Apoptose/efeitos dos fármacos , Citocinas/farmacologia , Fosfolipases A2 do Grupo VI/metabolismo , Insulinoma/enzimologia , Insulinoma/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Oxidantes/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Técnicas de Silenciamento de Genes , Mediadores da Inflamação/farmacologia , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Lipídeos/química , Oxirredução/efeitos dos fármacos , Ratos , Espectrometria de Massas por Ionização por Electrospray , Estreptozocina/farmacologia , terc-Butil Hidroperóxido/farmacologia
9.
J Surg Oncol ; 106(8): 972-80, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22711648

RESUMO

BACKGROUND: Insulinoma was a rare tumor and its pathogenesis was poorly understood. There had no study that focused on the role of mTOR signaling pathway in insulinoma tumorigenesis. MATERIALS AND METHODS: Expression of p-mTOR and its downstream p-P70S6K in insulinoma and normal pancreatic tissue was evaluated by immunohistochemical staining and Western blotting. In vitro study, an insulinoma cell line (INS-1) was treated with inhibitors of mTOR (rapamycin) or dual PI3K/mTOR inhibitor (NVP-BEZ235), RT-PCR, and Western blotting were applied to evaluate their influence on the expression of mTOR and P70S6K. Cell proliferation was evaluated by MTT test, cell cycle and apoptosis were analyzed by flow cytometry, insulin secretion level was evaluated by GSIS method. RESULTS: Positive expression of p-mTOR and p-P70S6K was much higher in insulinoma tumor specimens than the normal pancreatic islet (P < 0.05). mTOR inhibitors can induce decreased expression of mTOR and P70S6K, which resulting in inhibiting INS-1 cell proliferation, insulin secretion and inducing apoptosis. NVP-BEZ235 had better influence on inhibiting the cell proliferation and inducing apoptosis than rapamycin. CONCLUSION: mTOR/P70S6K signaling pathway is involved in tumorigenesis of insulinoma, NVP-BEZ235 and rapamycin offer a promising role as novel drugs in treatment of insulinoma.


Assuntos
Insulinoma/enzimologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Adulto , Feminino , Humanos , Insulinoma/patologia , Masculino , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia
10.
Cell Immunol ; 271(2): 379-84, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21885043

RESUMO

The pro-inflammatory cytokine IL-1ß leads to losses in functional ß-cell mass in part by inducing the expression of genes that produce soluble mediators of inflammation, such as cyclooxygenase-2 (COX2). In the current study, we sought to understand what factors control the COX2 gene in response to IL-1ß and how prostaglandins downstream of COX2 impact pro-inflammatory gene transcription in pancreatic ß-cells. We analyzed COX2 gene expression in response to different maneuvers impacting NF-κB proteins. Also, we report alterations in the expression of COX2, EP-3 and EP-4 receptor genes by PGD(2) and PGE(2). Moreover, we examined whether PGD(2) and PGE(2) regulated NF-κB and interferon-gamma activated sequence (GAS) reporter gene activity. IL-1ß-mediated induction of the COX2 gene requires the p65 and p50 subunits of NF-κB. In addition, PGD(2) and PGE(2) coordinately alter COX2 and EP receptor gene expression patterns and potentiate the cytokine-mediated transcriptional activity of promoters containing NF-κB or GAS response elements.


Assuntos
Ciclo-Oxigenase 2/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Interleucina-1beta/farmacologia , Prostaglandinas/farmacologia , Animais , Linhagem Celular Tumoral , Dinoprostona/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/farmacologia , Células Secretoras de Insulina/imunologia , Insulinoma/enzimologia , Insulinoma/genética , Insulinoma/imunologia , Interleucina-1beta/imunologia , Subunidade p50 de NF-kappa B/metabolismo , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Regiões Promotoras Genéticas , Prostaglandina D2/farmacologia , Ratos , Receptores de Prostaglandina E Subtipo EP3/genética , Receptores de Prostaglandina E Subtipo EP4/genética , Fator de Transcrição RelA/metabolismo
11.
J Biol Chem ; 284(51): 35359-67, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19858194

RESUMO

The cytosolic malic enzyme (ME1) has been suggested to augment insulin secretion via the malate-pyruvate and/or citrate-pyruvate shuttles, through the production of NADPH or other metabolites. We used selectable vectors expressing short hairpin RNA (shRNA) to stably decrease Me1 mRNA levels by 80-86% and ME1 enzyme activity by 78-86% with either of two shRNAs in the INS-1 832/13 insulinoma cell line. Contrary to published short term ME1 knockdown experiments, our long term targeted cells showed normal insulin secretion in response to glucose or to glutamine plus 2-aminobicyclo[2,2,1]heptane-2-carboxylic acid. We found no increase in the mRNAs and enzyme activities of the cytosolic isocitrate dehydrogenase or glucose-6-phosphate dehydrogenase, which also produce cytosolic NADPH. There was no compensatory induction of the mRNAs for the mitochondrial malic enzymes Me2 or Me3. Interferon pathway genes induced in preliminary small interfering RNA experiments were not induced in the long term shRNA experiments. We repeated our study with an improved vector containing Tol2 transposition sequences to produce a higher rate of stable transferents and shortened time to testing, but this did not alter the results. We similarly used stably expressed shRNA to reduce mitochondrial NAD(P)-malic enzyme (Me2) mRNA by up to 95%, with severely decreased ME2 protein and a 90% decrease in enzyme activity. Insulin release to glucose or glutamine plus 2-aminobicyclo[2,2,1]heptane-2-carboxylic acid remained normal. The maintenance of robust insulin secretion after lowering expression of either one of these malic enzymes is consistent with the redundancy of pathways of pyruvate cycling and/or cytosolic NADPH production in insulinoma cells.


Assuntos
Citosol/enzimologia , Insulina/metabolismo , Insulinoma/enzimologia , Insulinoma/metabolismo , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Aminoácidos Cíclicos/farmacologia , Animais , Linhagem Celular Tumoral , Expressão Gênica , Técnicas de Silenciamento de Genes , Glucose/farmacologia , Glucosefosfato Desidrogenase/genética , Glucosefosfato Desidrogenase/metabolismo , Insulina/genética , Secreção de Insulina , Insulinoma/genética , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Malato Desidrogenase , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Oxirredução/efeitos dos fármacos , Ratos , Edulcorantes/farmacologia
12.
Horm Metab Res ; 41(11): 805-13, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19598075

RESUMO

The objective of the present study was to examine the effects of melatonin on transcripts of isoforms of calcium/calmodulin-dependent protein kinases in rat insulinoma beta-cells INS-1. Investigations show that calcium/calmodulin-dependent kinase IV and calcium/calmodulin-dependent kinase 2d are expressed in human and rat pancreatic islets and INS-1 cells. By application of either forskolin or 3-isobutyl-1-methylxanthine for 6 hours, calcium spiking was evoked and the release of insulin was increased. The expression of the calcium/calmodulin-dependent kinase IV and calcium/calmodulin-dependent kinase 2d transcripts was significantly increased due to forskolin or 3-isobutyl-1-methylxanthine. Acute melatonin treatment (6 h) in the presence of either forskolin or 3-isobutyl-1-methylxanthine caused a significant decrease in insulin release and induced significant downregulation of calcium/calmodulin-dependent kinase IV and calcium/calmodulin-dependent kinase 2d transcripts in INS-1 batch cultures. The attenuating effect of melatonin on transcripts could be almost completely reversed by preincubation with the melatonin receptor antagonist luzindole. Thus, the insulin-inhibiting effect of melatonin in INS-1 cells is associated with significant changes in transcripts of calcium-signaling components suggesting that melatonin influences gene expression of components, which are known to be involved in insulin secretion or insulin gene expression.


Assuntos
1-Metil-3-Isobutilxantina/farmacologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Colforsina/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Insulinoma/enzimologia , Melatonina/farmacologia , Animais , Cálcio/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular Tumoral , Humanos , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Insulinoma/tratamento farmacológico , Insulinoma/genética , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Ratos , Ratos Wistar
13.
J Biol Chem ; 284(39): 26578-90, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19635791

RESUMO

Pancreatic beta-cells couple the oxidation of glucose to the secretion of insulin. Apart from the canonical K(ATP)-dependent glucose-stimulated insulin secretion (GSIS), there are important K(ATP)-independent mechanisms involving both anaplerosis and mitochondrial GTP (mtGTP). How mtGTP that is trapped within the mitochondrial matrix regulates the cytosolic calcium increases that drive GSIS remains a mystery. Here we have investigated whether the mitochondrial isoform of phosphoenolpyruvate carboxykinase (PEPCK-M) is the GTPase linking hydrolysis of mtGTP made by succinyl-CoA synthetase (SCS-GTP) to an anaplerotic pathway producing phosphoenolpyruvate (PEP). Although cytosolic PEPCK (PEPCK-C) is absent, PEPCK-M message and protein were detected in INS-1 832/13 cells, rat islets, and mouse islets. PEPCK enzymatic activity is half that of primary hepatocytes and is localized exclusively to the mitochondria. Novel (13)C-labeling strategies in INS-1 832/13 cells and islets measured substantial contribution of PEPCK-M to the synthesis of PEP. As high as 30% of PEP in INS-1 832/13 cells and 41% of PEP in rat islets came from PEPCK-M. The contribution of PEPCK-M to overall PEP synthesis more than tripled with glucose stimulation. Silencing the PEPCK-M gene completely inhibited GSIS underscoring its central role in mitochondrial metabolism-mediated insulin secretion. Given that mtGTP synthesized by SCS-GTP is an indicator of TCA flux that is crucial for GSIS, PEPCK-M is a strong candidate to link mtGTP synthesis with insulin release through anaplerotic PEP cycling.


Assuntos
Guanosina Trifosfato/metabolismo , Insulina/metabolismo , Mitocôndrias/enzimologia , Fosfoenolpiruvato Carboxilase/metabolismo , Fosfoenolpiruvato/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Ciclo do Ácido Cítrico , Secreção de Insulina , Insulinoma/enzimologia , Insulinoma/metabolismo , Insulinoma/patologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Mitocôndrias/metabolismo , Modelos Biológicos , Fosfoenolpiruvato Carboxilase/genética , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Succinato-CoA Ligases/metabolismo
14.
Methods Enzymol ; 457: 425-50, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19426882

RESUMO

Glucose homeostasis depends upon the appropriate release of insulin from pancreatic islet beta-cells. Postpandrial changes in circulating nutrient concentrations are coupled with graded release of stored insulin pools by the proportional changes in mitochondrial metabolism. The corresponding increased synthesis rates of both ATP and of anaplerotic metabolites have been shown to be mediators for nutrient-stimulated insulin secretion. Anaplerosis leads to the export of malate or citrate from the mitochondria, both of which can be recycled through metabolic pathways to reenter the Kreb's cycle. These metabolic cycles have the net effect of either transferring mitochondrial reducing equivalents to the cytosol, or of efficiently providing pyruvate to facilitate responsive changes in the Kreb's cycle flux in proportion to increased availability of glutamate and anaplerotic flux through glutamate dehydrogenase. Here, we describe siRNA knock-down and isotopic labeling strategies to evaluate the role of cytosolic and mitochondrial isoforms of malic enzyme in facilitating malate-pyruvate cycling in the context of fuel-stimulated insulin secretion.


Assuntos
Citosol/enzimologia , Insulina/metabolismo , Malato Desidrogenase/genética , Malato Desidrogenase/metabolismo , Mitocôndrias/enzimologia , Animais , Isótopos de Carbono , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico , Expressão Gênica , Técnicas de Silenciamento de Genes , Ácido Glutâmico/análise , Ácido Glutâmico/metabolismo , Insulinoma/enzimologia , Insulinoma/genética , Ilhotas Pancreáticas/enzimologia , Camundongos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Espectrometria de Massas por Ionização por Electrospray
15.
J Cell Mol Med ; 13(4): 758-70, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19438972

RESUMO

The effect of leucine on glucose-stimulated insulin secretion (GSIS) in pancreatic beta-cells is quite controversial, and mechanism involved in the effect has not been elucidated yet. Consequently, we aimed to investigate effect of leucine on GSIS and its mechanism focusing on contribution of AMP-activated protein kinase (AMPK) and pancreatic/duodenal homeobox-1 (PDX-1). Rat insulinoma beta-cells (INS-1, RIN m5F, DN-PDX-1#28 and PDX-1#6) were cultured with or without leucine, AICAR (AMPK agonist) or compound C (AMPK antagonist) for 48 hrs. In contrast to control, AICAR treatment decreased GSIS at high glucose and insulin content, also impaired protein and mRNA expression of PDX-1 and its downstream targets, glucokinase (GCK) and glucose transporter 2 (GLUT2). Compound C treatment had the opposite effects. We observed that neither AICAR nor compound C could affect expression of GCK and GLUT2 when PDX-1 expression was absent. Chronic leucine exposure inhibited GSIS at high glucose and insulin content in a dose-dependent manner, concomitant with an increase in AMPK and a decrease in PDX-1, GCK and GLUT2. The inhibitory effects of leucine was potentiated by AICAR treatment and rescued by compound C treatment. Finally, the inhibition of PDX-1 could potentiate the impaired effects induced by leucine whereas overexpression of PDX-1 could protect the cell from impairment induced by leucine. The study indicated that chronic leucine might result in an increase in AMPK and then a decrease in PDX-l, in turn to depress GCK and GLUT2 resulting in decreased GSIS at high glucose and insulin content.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/enzimologia , Insulina/metabolismo , Insulinoma/enzimologia , Leucina/farmacologia , Neoplasias Pancreáticas/enzimologia , Transativadores/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Glucoquinase/genética , Glucoquinase/metabolismo , Transportador de Glucose Tipo 2 , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Insulinoma/patologia , Neoplasias Pancreáticas/patologia , Ratos , Ribonucleotídeos/farmacologia
16.
Mol Biol Rep ; 36(7): 1703-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18839335

RESUMO

Beta-cell apoptosis induced by adipokines may result in beta-cell dysfunction in type 2 diabetes. Resistin, an adipokine-linked obesity with type 2 diabetes, impairs glucose-stimulated insulin secretion (GSIS) in beta-cells. Presently, the effects of resistin on rat insulinoma cells RINm5F were examined. Treatment of RINm5F with resistin induced cell damage. Tissue Inhibitor of Metalloproteinase-1 (TIMP-1) protected resistin-mediated cytotoxicity in RINm5F. Incubation with resistin up-regulated caspase-3 activity and induced the formation of a DNA ladder. TIMP-1 attenuated these effects. The molecular mechanism of TIMP-1 inhibition of resistin-mediated cytotoxicity appeared to involve Akt phosphorylation and activation of IkB-alpha phosphorylation. Resistin treatment suppressed Akt phosphorylation and activated IkB-alpha phosphorylation, which could be attenuated by TIMP-1. We conclude that resistin can induce beta-cell apoptosis and that resistin-related beta-cell apoptosis can be prevented by TIMP-1.


Assuntos
Apoptose/efeitos dos fármacos , Insulinoma/patologia , Resistina/farmacologia , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Proteínas I-kappa B/metabolismo , Insulinoma/enzimologia , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Inibidor Tecidual de Metaloproteinase-1/farmacologia , Proteína X Associada a bcl-2/metabolismo
17.
Endocrinology ; 149(6): 3046-53, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18308848

RESUMO

The aim of the present investigation was to characterize the role of the MAPK kinase kinase-1 (MEKK-1) in stress-induced cell death of insulin producing cells. We observed that transient overexpression of the wild type MEKK-1 protein in the insulin-producing cell lines RIN-5AH and betaTC-6 increased c-Jun N-terminal kinase (JNK) phosphorylation and augmented cell death induced by diethylenetriamine/nitroso-1-propylhydrazino)-1-propanamine (DETA/NO), streptozotocin (STZ), and hydrogen peroxide (H2O2). Furthermore, DETA/NO or STZ induced a rapid threonine phosphorylation of MEKK-1. Silencing of MEKK-1 gene expression in betaTC-6 and human dispersed islet cells, using in vitro-generated diced small interfering RNA, resulted in protection from DETA/NO, STZ, H2O2, and tunicamycin induced cell death. Moreover, in DETA/NO-treated cells diced small interfering RNA-mediated down-regulation of MEKK-1 resulted in decreased activation of JNK but not p38 and ERK. Inhibition of JNK by treatment with SP600125 partially protected against DETA/NO- or STZ-induced cell death. In summary, our results support an essential role for MEKK-1 in JNK activation and stress-induced beta-cell death. Increased understanding of the signaling pathways that augment or diminish beta-cell MEKK-1 activity may aid in the generation of novel therapeutic strategies in the treatment of type 1 diabetes.


Assuntos
Ilhotas Pancreáticas/enzimologia , MAP Quinase Quinase Quinase 1/metabolismo , Animais , Morte Celular , Sobrevivência Celular , Diabetes Mellitus Tipo 1/terapia , Citometria de Fluxo , Genes Reporter , Humanos , Insulinoma/enzimologia , Insulinoma/patologia , Ilhotas Pancreáticas/citologia , MAP Quinase Quinase Quinase 1/genética , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas
18.
FEBS Lett ; 581(30): 5885-90, 2007 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-18061583

RESUMO

Inhibition of the lipid phosphatase SH2-domain containing inositol phosphatase 2 (SHIP2) in L6-C10 muscle cells, in 3T3-L1 adipocytes and in the liver of db/db mice has been shown to ameliorate insulin signal transduction and established SHIP2 as a negative regulator of insulin action. Here we show that SHIP2 inhibition in INS1E insulinoma cells increased Akt, glycogen synthase kinase 3 and extracellular signal-regulated kinases 1 and 2 phosphorylation. SHIP2 inhibition did not prevent palmitate-induced apoptosis, but increased cell proliferation. Our data raise the interesting possibility that SHIP2 inhibition exerts proliferative effects in beta-cells and further support the attractiveness of a specific inhibition of SHIP2 for the treatment of type 2 diabetes.


Assuntos
Insulina/metabolismo , Insulinoma/enzimologia , Insulinoma/patologia , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Transdução de Sinais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina A/genética , Ciclina A/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Genes Dominantes , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Insulinoma/genética , Proteínas Mutantes/metabolismo , Ácido Palmítico/farmacologia , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos
19.
Int J Biochem Cell Biol ; 39(11): 2076-82, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17689130

RESUMO

Endoplasmic reticulum stress-mediated apoptosis plays an important role in the destruction of pancreatic beta-cell, and contributes to the development of type 1 diabetes. The chaperone molecule, glucose regulated proteins 78 (GRP78), is required to maintain ER function during toxic insults. In this study, we investigated the effect of GRP78 on the beta-cell apoptosis. We first measured GRP78 protein expression in different phase of streptozotocin-affected beta-cell by immunoblotting analysis. An insulinoma cell line, NIT-1, transfected with GRP78 was established, named NIT-GRP78, and used to study apoptosis, which was induced by streptozotocin or inflammatory cytokines. Apoptosis of NIT-1 or NIT-GRP78 cells was detected by flow cytometry, the transcription of C/EBP homologous protein (CHOP) was monitored by real-time PCR, the concentration of nitric oxide and the activity of superoxide dismutase were measured by colorimetric method. We found that, in comparison to NIT-1 cells, NIT-GRP78 cells responded to the streptozotocin or cytokines treatments with decreased concentration of nitric oxide, but increased activity of superoxide dismutase. In addition, the level of CHOP was also decreased in the NIT-GRP78 cells, which may mediate the resistance of the GRP78 overexpressed NIT-1 cells from apoptosis. Finally, we found that NIT-GRP78 cells were also more resistant than NIT-1 cells to cytotoxic T lymphocyte (CTL) specific killing detected by flow cytometry through target cells expressing green fluorescent protein cultured with effector cells and finally stained with propidium iodide. The data suggest that modulating GRP78 expression could be useful in preventing pancreatic beta-cell from the immunological destruction in type 1 diabetes individuals.


Assuntos
Citocinas/farmacologia , Proteínas de Choque Térmico/metabolismo , Insulinoma/patologia , Chaperonas Moleculares/metabolismo , Estreptozocina/farmacologia , Linfócitos T Citotóxicos/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Proteínas de Fluorescência Verde/metabolismo , Insulinoma/enzimologia , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Superóxido Dismutase/metabolismo
20.
Biochemistry ; 46(35): 10170-85, 2007 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-17685585

RESUMO

Beta-cell mass is regulated by a balance between beta-cell growth and beta-cell death, due to apoptosis. We previously reported that apoptosis of INS-1 insulinoma cells due to thapsigargin-induced ER stress was suppressed by inhibition of the group VIA Ca2+-independent phospholipase A2 (iPLA2beta), associated with an increased level of ceramide generation, and that the effects of ER stress were amplified in INS-1 cells in which iPLA2beta was overexpressed (OE INS-1 cells). These findings suggested that iPLA2beta and ceramides participate in ER stress-induced INS-1 cell apoptosis. Here, we address this possibility and also the source of the ceramides by examining the effects of ER stress in empty vector (V)-transfected and iPLA2beta-OE INS-1 cells using apoptosis assays and immunoblotting, quantitative PCR, and mass spectrometry analyses. ER stress induced expression of ER stress factors GRP78 and CHOP, cleavage of apoptotic factor PARP, and apoptosis in V and OE INS-1 cells. Accumulation of ceramide during ER stress was not associated with changes in mRNA levels of serine palmitoyltransferase (SPT), the rate-limiting enzyme in de novo synthesis of ceramides, but both message and protein levels of neutral sphingomyelinase (NSMase), which hydrolyzes sphingomyelins to generate ceramides, were temporally increased in the INS-1 cells. The increases in the level of NSMase expression in the ER-stressed INS-1 cells were associated with corresponding temporal elevations in ER-associated iPLA2beta protein and catalytic activity. Pretreatment with BEL inactivated iPLA2beta and prevented induction of NSMase message and protein in ER-stressed INS-1 cells. Relative to that in V INS-1 cells, the effects of ER stress were accelerated and/or amplified in the OE INS-1 cells. However, inhibition of iPLA2beta or NSMase (chemically or with siRNA) suppressed induction of NSMase message, ceramide generation, sphingomyelin hydrolysis, and apoptosis in both V and OE INS-1 cells during ER stress. In contrast, inhibition of SPT did not suppress ceramide generation or apoptosis in either V or OE INS-1 cells. These findings indicate that iPLA2beta activation participates in ER stress-induced INS-1 cell apoptosis by promoting ceramide generation via NSMase-catalyzed hydrolysis of sphingomyelins, raising the possibility that this pathway contributes to beta-cell apoptosis due to ER stress.


Assuntos
Apoptose , Ceramidas/metabolismo , Fosfolipases A2 do Grupo VI/metabolismo , Insulinoma/enzimologia , Neoplasias Pancreáticas/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/metabolismo , Apoptose/fisiologia , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/enzimologia , Chaperona BiP do Retículo Endoplasmático , Regulação Enzimológica da Expressão Gênica , Humanos , Hidrólise , Inibidores de Fosfodiesterase/farmacologia , RNA Interferente Pequeno , Retroviridae/genética , Espectrometria de Massas por Ionização por Electrospray , Esfingomielina Fosfodiesterase/efeitos dos fármacos , Tapsigargina/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA