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











Base de dados
Intervalo de ano de publicação
2.
Cell Death Differ ; 21(8): 1313-24, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24786832

RESUMO

Diets rich in saturated fats may contribute to the loss of pancreatic ß-cells in type 2 diabetes. JunB, a member of the activating protein 1 (AP-1) transcription factor family, promotes ß-cell survival and mediates part of the beneficial effects of GLP-1 agonists. In this study we interrogated the molecular mechanisms involved in JunB-mediated ß-cell protection from lipotoxicity. The saturated fatty acid palmitate decreased JunB expression, and this loss may contribute to ß-cell apoptosis, as overexpression of JunB protected cells from lipotoxicity. Array analysis of JunB-deficient ß-cells identified a gene expression signature of a downregulated endoplasmic reticulum (ER) stress response and inhibited AKT signaling. JunB stimulates XBP1 expression via the transcription factor c/EBPδ during ER stress, and forced expression of XBP1s rescued the viability of JunB-deficient cells, constituting an important antiapoptotic mechanism. JunB silencing inhibited AKT activation and activated the proapoptotic Bcl-2 protein BAD via its dephosphorylation. BAD knockdown reversed lipotoxic ß-cell death potentiated by JunB siRNA. Interestingly, XBP1s links JunB and AKT signaling as XBP1 knockdown also reduced AKT phosphorylation. GLP-1 agonists induced cAMP-dependent AKT phosphorylation leading to ß-cell protection against palmitate-induced apoptosis. JunB and XBP1 knockdown or IRE1 inhibition decreased AKT activation by cAMP, leading to ß-cell apoptosis. In conclusion, JunB modulates the ß-cell ER stress response and AKT signaling via the induction of XBP1s. The activation of the JunB gene network and the crosstalk between the ER stress and AKT pathway constitute a crucial defense mechanism by which GLP-1 agonists protect against lipotoxic ß-cell death. These findings elucidate novel ß-cell-protective signal transduction in type 2 diabetes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Células Secretoras de Insulina/enzimologia , Masculino , Pessoa de Meia-Idade , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Fatores de Transcrição de Fator Regulador X , Transdução de Sinais , Fatores de Transcrição/genética , Proteína 1 de Ligação a X-Box
3.
Cell Death Differ ; 20(12): 1742-52, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24096873

RESUMO

Cell-to-cell communication mediated by gap junctions made of Connexin36 (Cx36) contributes to pancreatic ß-cell function. We have recently demonstrated that Cx36 also supports ß-cell survival by a still unclear mechanism. Using specific Cx36 siRNAs or adenoviral vectors, we now show that Cx36 downregulation promotes apoptosis in INS-1E cells exposed to the pro-inflammatory cytokines (IL-1ß, TNF-α and IFN-γ) involved at the onset of type 1 diabetes, whereas Cx36 overexpression protects against this effect. Cx36 overexpression also protects INS-1E cells against endoplasmic reticulum (ER) stress-mediated apoptosis, and alleviates the cytokine-induced production of reactive oxygen species, the depletion of the ER Ca(2+) stores, the CHOP overexpression and the degradation of the anti-apoptotic protein Bcl-2 and Mcl-1. We further show that cytokines activate the AMP-dependent protein kinase (AMPK) in a NO-dependent and ER-stress-dependent manner and that AMPK inhibits Cx36 expression. Altogether, the data suggest that Cx36 is involved in Ca(2+) homeostasis within the ER and that Cx36 expression is downregulated following ER stress and subsequent AMPK activation. As a result, cytokine-induced Cx36 downregulation elicits a positive feedback loop that amplifies ER stress and AMPK activation, leading to further Cx36 downregulation. The data reveal that Cx36 plays a central role in the oxidative stress and ER stress induced by cytokines and the subsequent regulation of AMPK activity, which in turn controls Cx36 expression and mitochondria-dependent apoptosis of insulin-producing cells.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Conexinas/metabolismo , Citocinas/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Conexinas/genética , AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Retroalimentação Fisiológica/efeitos dos fármacos , Humanos , Insulina/biossíntese , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Interleucina-1beta/farmacologia , Metformina/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Ratos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Elementos de Resposta/genética , Transcrição Gênica/efeitos dos fármacos , Proteína delta-2 de Junções Comunicantes
4.
Diabetologia ; 56(2): 234-41, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23132339

RESUMO

Protein synthesis is increased by several-fold in stimulated pancreatic beta cells. Synthesis and folding of (pro)insulin takes place in the endoplasmic reticulum (ER), and beta cells trigger the unfolded protein response (UPR) to upgrade the functional capacity of the ER. Prolonged or excessive UPR activation contributes to beta cell dysfunction and death in type 2 diabetes, but there is another side of the UPR that may be of particular relevance for autoimmune type 1 diabetes, namely, the cross-talk between the UPR and innate immunity/inflammation. Recent evidence, discussed in this review, indicates that both saturated fats and inflammatory mediators such as cytokines trigger the UPR in pancreatic beta cells. The UPR potentiates activation of nuclear factor κB, a key regulator of inflammation. Two branches of the UPR, namely IRE1/XBP1s and PERK/ATF4/CHOP, mediate the UPR-induced sensitisation of pancreatic beta cells to the proinflammatory effects of cytokines. This can contribute to the upregulation of local inflammatory mechanisms and the aggravation of insulitis. The dialogue between the UPR and inflammation may provide an explanation for the parallel increase in the prevalence of childhood obesity and type 1 diabetes.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Ilhotas Pancreáticas/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Animais , Estresse do Retículo Endoplasmático/genética , Humanos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Resposta a Proteínas não Dobradas/genética
5.
Cell Death Differ ; 19(11): 1836-46, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22653339

RESUMO

Induction of the C/EBP homologous protein (CHOP) is considered a key event for endoplasmic reticulum (ER) stress-mediated apoptosis. Type 1 diabetes (T1D) is characterized by an autoimmune destruction of the pancreatic ß-cells. Pro-inflammatory cytokines are early mediators of ß-cell death in T1D. Cytokines induce ER stress and CHOP overexpression in ß-cells, but the role for CHOP overexpression in cytokine-induced ß-cell apoptosis remains controversial. We presently observed that CHOP knockdown (KD) prevents cytokine-mediated degradation of the anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2) and myeloid cell leukemia sequence 1 (Mcl-1), thereby decreasing the cleavage of executioner caspases 9 and 3, and apoptosis. Nuclear factor-κB (NF-κB) is a crucial transcription factor regulating ß-cell apoptosis and inflammation. CHOP KD resulted in reduced cytokine-induced NF-κB activity and expression of key NF-κB target genes involved in apoptosis and inflammation, including iNOS, FAS, IRF-7, IL-15, CCL5 and CXCL10. This was due to decreased IκB degradation and p65 translocation to the nucleus. The present data suggest that CHOP has a dual role in promoting ß-cell death: (1) CHOP directly contributes to cytokine-induced ß-cell apoptosis by promoting cytokine-induced mitochondrial pathways of apoptosis; and (2) by supporting the NF-κB activation and subsequent cytokine/chemokine expression, CHOP may contribute to apoptosis and the chemo attraction of mononuclear cells to the islets during insulitis.


Assuntos
Apoptose/efeitos dos fármacos , Citocinas/farmacologia , Células Secretoras de Insulina/metabolismo , Fator de Transcrição CHOP/metabolismo , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Estresse do Retículo Endoplasmático , Quinase I-kappa B/metabolismo , Células Secretoras de Insulina/citologia , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Mitocôndrias/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Fator de Transcrição CHOP/antagonistas & inibidores , Fator de Transcrição CHOP/genética , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
6.
Diabetologia ; 55(6): 1572-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22526614

RESUMO

Type 1 diabetes is a chronic autoimmune disease involving the progressive loss of beta cell mass. Cytokines released by immune cells are early contributors to beta cell apoptosis. Thus, an understanding of the signal transduction mechanisms induced by cytokines in beta cells is necessary for the rational design of novel therapies to prevent or to cure this disease. Cytokine-mediated beta cell apoptosis is a complex phenomenon that includes activation of the transcription factors signal transducer and activator of transcription 1 and nuclear factor κB (NFκB), c-Jun N-terminal kinase, endoplasmic reticulum (ER) stress and the intrinsic mitochondrial apoptotic pathway. NFκB has both a pro-inflammatory and a pro-apoptotic role in beta cells. One of the mechanisms by which NFκB contributes to beta cell apoptosis is via activation of ER stress. The role for ER stress in beta cell apoptosis is not completely clarified but involves production of C/EBP homologous protein and activation of the intrinsic mitochondrial apoptotic pathway. In this issue of Diabetologia, Roggli et al (DOI 10.1007/s00125-011-2399-7) report on a new player in this elaborate response, the RNA-binding protein ARE/poly(U)-binding factor 1. This commentary discusses these findings and their relevance to the field.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , Ilhotas Pancreáticas/metabolismo , Isoformas de Proteínas/metabolismo , Ribonucleoproteína Nuclear Heterogênea D0 , Humanos
7.
Diabetologia ; 55(4): 1167-78, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22237685

RESUMO

AIMS/HYPOTHESIS: Endoplasmic reticulum (ER) stress has been implicated in the development of type 2 diabetes, via effects on obesity, insulin resistance and pancreatic beta cell health. C/EBP homologous protein (CHOP) is induced by ER stress and has a central role in apoptotic execution pathways triggered by ER stress. The aim of this study was to characterise the role of CHOP in obesity and insulin resistance. METHODS: Metabolic studies were performed in Chop ( -/- ) and wild-type C57Bl/6 mice, and included euglycaemic-hyperinsulinaemic clamps and indirect calorimetry. The inflammatory state of liver and adipose tissue was determined by quantitative RT-PCR, immunohistology and macrophage cultures. Viability and absence of ER stress in islets of Langerhans was determined by electron microscopy, islet culture and quantitative RT-PCR. RESULTS: Systemic deletion of Chop induced abdominal obesity and hepatic steatosis. Despite marked obesity, Chop ( -/- ) mice had preserved normal glucose tolerance and insulin sensitivity. This discrepancy was accompanied by lower levels of pro-inflammatory cytokines and less infiltration of immune cells into fat and liver. CONCLUSIONS/INTERPRETATION: These observations suggest that insulin resistance is not induced by fat accumulation per se, but rather by the inflammation induced by ectopic fat. CHOP may play a key role in the crosstalk between excessive fat deposition and induction of inflammation-mediated insulin resistance.


Assuntos
Fígado Gorduroso/metabolismo , Inflamação/metabolismo , Resistência à Insulina/fisiologia , Obesidade/metabolismo , Fator de Transcrição CHOP/metabolismo , Tecido Adiposo/metabolismo , Animais , Fígado Gorduroso/genética , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Inflamação/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Obesidade/genética , Fator de Transcrição CHOP/genética
8.
Cell Death Differ ; 18(2): 328-37, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20798690

RESUMO

Pancreatic ß-cell apoptosis is a key feature of diabetes mellitus and the mitochondrial pathway of apoptosis is a major mediator of ß-cell death. We presently evaluated the role of the myeloid cell leukemia sequence 1 (Mcl-1), an antiapoptotic protein of the Bcl-2 family, in ß-cells following exposure to well-defined ß-cell death effectors, for example, pro-inflammatory cytokines, palmitate and chemical endoplasmic reticulum (ER) stressors. All cytotoxic stresses rapidly and preferentially decreased Mcl-1 protein expression as compared with the late effect observed on the other antiapoptotic proteins, Bcl-2 and Bcl-xL. This was due to ER stress-mediated inhibition of translation through eIF2α phosphorylation for palmitate and ER stressors and through the combined action of translation inhibition and JNK activation for cytokines. Knocking down Mcl-1 using small interference RNAs increased apoptosis and caspase-3 cleavage induced by cytokines, palmitate or thapsigargin, whereas Mcl-1 overexpression partly prevented Bax translocation to the mitochondria, cytochrome c release, caspase-3 cleavage and apoptosis induced by the ß-cell death effectors. Altogether, our data suggest that Mcl-1 downregulation is a crucial event leading to ß-cell apoptosis and provide new insights into the mechanisms linking ER stress and the mitochondrial intrinsic pathway of apoptosis. Mcl-1 is therefore an attractive target for the design of new strategies in the treatment of diabetes.


Assuntos
Apoptose , Citocinas/farmacologia , Células Secretoras de Insulina/metabolismo , Palmitatos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Regulação para Baixo , Retículo Endoplasmático/metabolismo , Células Secretoras de Insulina/citologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas Proto-Oncogênicas c-bcl-2/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Tapsigargina/farmacologia , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
9.
Diabetologia ; 53(6): 1120-30, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20349222

RESUMO

AIMS/HYPOTHESIS: Pro-inflammatory cytokines involved in the pathogenesis of type 1 diabetes deplete endoplasmic reticulum (ER) Ca2+ stores, leading to ER-stress and beta cell apoptosis. However, the cytokine-induced ER-stress response in beta cells is atypical and characterised by induction of the pro-apoptotic PKR-like ER kinase (PERK)-C/EBP homologous protein (CHOP) branch of the unfolded protein response, but defective X-box binding protein 1 (XBP1) splicing and activating transcription factor 6 activation. The purpose of this study was to overexpress spliced/active Xbp1 (XBP1s) to increase beta cell resistance to cytokine-induced ER-stress and apoptosis. METHODS: Xbp1s was overexpressed using adenoviruses and knocked down using small interference RNA in rat islet cells. In selected experiments, Xbp1 was also knocked down in FACS-purified rat beta cells and rat fibroblasts. Expression and production of XBP1s and key downstream genes and proteins was measured and beta cell function and viability were evaluated. RESULTS: Adenoviral-mediated overproduction of Xbp1s resulted in increased XBP1 activity and induction of several XBP1s target genes. Surprisingly, XBP1s overexpression impaired glucose-stimulated insulin secretion and increased beta cell apoptosis, whereas it protected fibroblasts against cell death induced by ER-stress. mRNA expression of Pdx1 and Mafa was inhibited in cells overproducing XBP1s, leading to decreased insulin expression. XBP1s knockdown partially restored cytokine/ER-stress-driven insulin and Pdx1 inhibition but had no effect on cytokine-induced ER-stress and apoptosis. CONCLUSIONS/INTERPRETATION: XBP1 has a distinct inhibitory role in beta cell as compared with other cell types. Prolonged XBP1s production hampers beta cell function via inhibition of insulin, Pdx1 and Mafa expression, eventually leading to beta cell apoptosis.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/metabolismo , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas de Ligação a DNA/genética , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Imunofluorescência , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Indóis/farmacologia , Insulina/genética , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-8/farmacologia , Fatores de Transcrição Maf/genética , Fatores de Transcrição Maf/metabolismo , Masculino , Interferência de RNA , RNA Interferente Pequeno , Ratos , Ratos Wistar , Fatores de Transcrição de Fator Regulador X , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Transfecção , Proteína 1 de Ligação a X-Box
10.
Cell Death Differ ; 16(11): 1539-50, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19629134

RESUMO

Chronic inflammation and pro-inflammatory cytokines are important mediators of pancreatic beta-cell destruction in type 1 diabetes (T1D). We presently show that the cytokines IL-1beta+IFN-gamma and different ER stressors activate the Bcl-2 homology 3 (BH3)-only member death protein 5 (DP5)/harakiri (Hrk) resulting in beta-cell apoptosis. Chemical ER stress-induced DP5 upregulation is JNK/c-Jun-dependent. DP5 activation by cytokines also involves JNK/c-Jun phosphorylation and is antagonized by JunB. Interestingly, cytokine-inducted DP5 expression precedes ER stress: mitochondrial release of cytochrome c and ER stress are actually a consequence of enhanced DP5 activation by cytokine-mediated nitric oxide formation. Our findings show that DP5 is central for beta-cell apoptosis after different stimuli, and that it can act up- and downstream of ER stress. These observations contribute to solve two important questions, namely the mechanism by which IL-1beta+IFN-gamma induce beta-cell death and the nature of the downstream signals by which ER stress 'convinces' beta-cells to trigger apoptosis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Retículo Endoplasmático/metabolismo , Células Secretoras de Insulina/citologia , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Neuropeptídeos/metabolismo , Animais , Citocromos c/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Regulação para Cima
11.
Cell Transplant ; 16(5): 527-37, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17708342

RESUMO

Islet transplantation is a promising treatment in type 1 diabetes, but the need for chronic immunosuppression is a major hurdle to broad applicability. Ex vivo introduction of agents by lentiviral vectors-improving beta-cell resistance against immune attack-is an attractive path to pursue. The aim of this study was to investigate whether dissociation of islets to single cells prior to viral infection and reaggregation before transplantation would improve viral transduction efficacy without cytotoxicity. This procedure improved transduction efficacy with a LV-pWPT-CMV-EGFP construct from 11.2 +/- 4.1% at MOI 50 in whole islets to 80.0 +/- 2.8% at MOI 5. Viability (as measured by Hoechst/PI) and functionality (as measured by glucose challenge) remained high. After transplantation, the transfected pseudoislet aggregates remained EGFP positive for more than 90 days and the expression of EGFP colocalized primarily with the insulin-positive beta-cells. No increased vulnerability to immune attack was observed in vitro or in vivo. These data demonstrate that dispersion of islets prior to lentiviral transfection and reaggregation prior to transplantation is a highly efficient way to introduce genes of interest into islets for transplantation purposes in vitro and in vivo, but the amount of beta-cells needed for normalization of glycemia was more than eightfold higher when using dispersed cell aggregates versus unmanipulated islets. The high price to pay to reach stable and strong transgene expression in islet cells is certainly an important cell loss.


Assuntos
Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/citologia , Lentivirus/genética , Transdução Genética , Animais , Agregação Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/farmacologia , Citometria de Fluxo , Humanos , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Ratos , Ratos Wistar , Fatores de Tempo , Transgenes
12.
Diabetologia ; 50(5): 1006-14, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17333111

RESUMO

AIMS/HYPOTHESIS: Pancreatic beta cells respond to endoplasmic reticulum (ER) stress by activating the unfolded protein response. If the stress is prolonged, or the adaptive response fails, apoptosis is triggered. We used a 'homemade' microarray specifically designed for the study of beta cell apoptosis (the APOCHIP) to uncover mechanisms regulating beta cell responses to ER stress. MATERIALS AND METHODS: A time course viability and microarray analysis was performed in insulin-producing INS-1E cells exposed to the reversible ER stress inducer cyclopiazonic acid (CPA). Modification of selected genes was confirmed by real-time RT-PCR, and the observed inhibition of expression of the insulin-1 (Ins1) and insulin-2 (Ins2) genes was further characterised in primary beta cells exposed to a diverse range of agents that induce ER stress. RESULTS: CPA-induced ER stress modified the expression of 183 genes at one or more of the time points studied. The expression of most of these genes returned to control levels after a 3 h recovery period following CPA removal, with all cells surviving. Two groups of genes were particularly affected by CPA, namely, those related to cellular responses to ER stress, which were mostly upregulated, and those related to differentiated beta cell functions, which were downregulated. Levels of Ins1 and Ins2 mRNAs were severely decreased in response to CPA treatment as a result of degradation, and there was a concomitant increase in the level of IRE1 activation. CONCLUSIONS/INTERPRETATION: In this study we provide the first global analysis of beta cell molecular responses to a severe ER stress, and identify the early degradation of mRNA transcripts of the insulin genes as an important component of this response.


Assuntos
Retículo Endoplasmático/fisiologia , Perfilação da Expressão Gênica , Células Secretoras de Insulina/fisiologia , Insulina/genética , RNA Mensageiro/metabolismo , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Indóis/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Cinética , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Estresse Mecânico
13.
Diabetologia ; 49(6): 1229-36, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16604358

RESUMO

AIMS/HYPOTHESIS: A tight control of endoplasmic reticulum homeostasis is crucial for beta cell function and survival. We recently described that IL-1beta plus IFN-gamma deplete endoplasmic reticulum Ca2+ stores in beta cells, leading to endoplasmic reticulum stress and apoptosis. IL-1beta alone induced endoplasmic reticulum stress but failed to induce beta cell death, while IFN-gamma alone neither caused endoplasmic reticulum stress nor induced beta cell death. This suggests that IFN-gamma aggravates endoplasmic reticulum stress induced by IL-1beta, eventually triggering apoptosis. Here we tested this hypothesis and the mechanisms involved, by investigating the effects of IFN-gamma on endoplasmic reticulum-stress-induced beta cell apoptosis caused by a specific blocker of the sarcoendoplasmic-reticulum pump Ca2+-ATPase (SERCA). MATERIALS AND METHODS: INS-1E cells or beta cells were pretreated with IFN-gamma and then exposed to the SERCA blocker cyclopiazonic acid (CPA) for induction of endoplasmic reticulum stress. Cell death was evaluated by Hoechst 342 and propidium iodide staining. Expression of genes related to endoplasmic reticulum stress was determined by real-time RT-PCR, while activation of the endoplasmic reticulum stress response was determined by analysing X-box binding protein-1 (Xbp1) splicing and using a reporter construct containing five copies of the unfolded protein response element (UPRE). RESULTS: CPA induces endoplasmic reticulum stress and apoptosis in insulin-producing cells. Pretreatment with IFN-gamma decreased the basal level of spliced Xbp1 mRNA, the basal and CPA-induced activity of the UPRE reporter, and the mRNA expression of several endoplasmic reticulum chaperones (Bip, Grp94 and Orp 150) and Sec61a. Furthermore, CPA-induced Chop mRNA expression and beta cell apoptosis were potentiated in cells that had been pretreated with IFN-gamma. CONCLUSIONS/INTERPRETATION: CPA-induced endoplasmic reticulum stress and apoptosis is enhanced in IFN-gamma-treated beta cells. These effects are mediated via downregulation of the expression of genes involved in beta cell defence against endoplasmic reticulum stress.


Assuntos
Morte Celular/efeitos dos fármacos , Retículo Endoplasmático/fisiologia , Células Secretoras de Insulina/fisiologia , Interferon gama/farmacologia , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/ultraestrutura , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Indóis/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Mecânico
14.
Diabetologia ; 48(3): 496-505, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15739117

RESUMO

AIMS/HYPOTHESIS: Hyperglycaemia and the pro-inflammatory cytokine IL-1beta induce similar alterations of beta cell gene expression, including up-regulation of c-Myc and haeme-oxygenase 1. These effects of hyperglycaemia may result from nuclear factor-kappa B (NFkappaB) activation by oxidative stress. To test this hypothesis, we compared the effects of IL-1beta, high glucose, and hydrogen peroxide, on NFkappaB DNA binding activity and target gene mRNA levels in cultured rat islets. METHODS: Rat islets were pre-cultured for 1 week in serum-free RPMI medium containing 10 mmol/l glucose, and further cultured in glucose concentrations of 5-30 mmol/l plus various test substances. Islet NFkappaB activity was measured by ELISA and gene mRNA expression was measured by RT-PCR. RESULTS: IL-1beta consistently increased islet NFkappaB activity and c-Myc, haeme-oxygenase 1, inducible nitric oxide synthase (iNOS), Fas, and inhibitor of NFkappaB alpha (IkappaBalpha) mRNA levels. In comparison, 1- to 7-day culture in 30 mmol/l instead of 10 mmol/l glucose stimulated islet c-Myc and haeme-oxygenase 1 expression without affecting NFkappaB activity or iNOS and IkappaBalpha mRNA levels. Fas mRNA levels only increased after 1 week in 30 mmol/l glucose. Overnight exposure to hydrogen peroxide mimicked the effects of 30 mmol/l glucose on haeme-oxygenase 1 and c-Myc mRNA levels without activating NFkappaB. On the other hand, the antioxidant N-acetyl-L-cysteine inhibited the stimulation of haeme-oxygenase 1 and c-Myc expression by 30 mmol/l glucose and/or hydrogen peroxide. CONCLUSIONS/INTERPRETATION: In contrast to IL-1beta, high glucose and hydrogen peroxide do not activate NFkappaB in cultured rat islets. It is suggested that the stimulation of islet c-Myc and haeme-oxygenase 1 expression by 30 mmol/l glucose results from activation of a distinct, probably oxidative-stress-dependent signalling pathway.


Assuntos
Glucose/farmacologia , Heme Oxigenase (Desciclizante)/genética , Peróxido de Hidrogênio/farmacologia , Ilhotas Pancreáticas/metabolismo , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , Transcrição Gênica/efeitos dos fármacos , Animais , Heme Oxigenase-1 , Ilhotas Pancreáticas/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , RNA Mensageiro/efeitos dos fármacos , Ratos
15.
Diabetologia ; 46(12): 1641-57, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14600816

RESUMO

AIMS/HYPOTHESIS: Viral infections and local production of IFN-gamma might contribute to beta-cell dysfunction/death in Type 1 Diabetes. Double stranded RNA (dsRNA) accumulates in the cytosol of viral-infected cells, and exposure of purified rat beta cells to dsRNA (tested in the form of polyinosinic-polycytidylic acid, PIC) in combination with IFN-gamma results in beta-cell dysfunction and apoptosis. To elucidate the molecular mechanisms involved in PIC + IFN-gamma-effects, we determined the global profile of genes modified by these agents in primary rat beta cells. METHODS: FACS-purified rat beta cells were cultured for 6 or 24 h in control condition or with IFN-gamma, PIC or a combination of both agents. The gene expression profile was analysed in duplicate by high-density oligonucleotide arrays representing 5000 full-length genes and 3000 EST's. Changes of greater than or equal to 2.5-fold were considered as relevant. RESULTS: Following a 6- or 24-h treatment with IFN-gamma, PIC or IFN-gamma and PIC, we observed changes in the expression of 51 to 189 genes. IFN-gamma modified the expression of MHC-related genes, and also of genes involved in beta-cell metabolism, protein processing, cytokines and signal transduction. PIC affected preferentially the expression of genes related to cell adhesion, cytokines and dsRNA signal transduction, transcription factors and MHC. PIC and/or IFN-gamma up-regulated the expression of several chemokines and cytokines that could contribute to mononuclear cell homing and activation during viral infection, while IFN-gamma induced a positive feedback on its own signal transduction. PIC + IFN-gamma inhibited insulin and GLUT-2 expression without modifying pdx-1 mRNA expression. CONCLUSION/INTERPRETATION: This study provides the first comprehensive characterization of the molecular responses of primary beta cells to dsRNA + IFN-gamma, two agents that are probably present in the beta cell milieu during the course of virally-induced insulitis and Type 1 Diabetes. Based on these findings, we propose an integrated model for the molecular mechanisms involved in dsRNA + IFN-gamma induced beta-cell dysfunction and death.


Assuntos
Perfilação da Expressão Gênica , Interferon gama/farmacologia , Ilhotas Pancreáticas/fisiologia , RNA de Cadeia Dupla/farmacologia , Animais , Células Cultivadas , Etiquetas de Sequências Expressas , Ilhotas Pancreáticas/efeitos dos fármacos , Complexo Principal de Histocompatibilidade/efeitos dos fármacos , Masculino , Poli I-C/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
16.
Diabetologia ; 46(2): 255-66, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12627325

RESUMO

AIMS/HYPOTHESIS: Cytokines and chemokines are important mediators of immune responses due to their ability to recruit and activate leukocytes. Using microarray analysis we observed that rat beta cells exposed to IL-1beta and IFN-gamma have increased mRNA levels of chemokines and IL-15. The aim of this study was to characterize the expression of IP-10, MIP-3alpha, fractalkine and IL-15 in rat beta cells, human pancreatic islets, and in islets isolated from NOD mice, both during the pre-diabetic period and following islet transplantation. METHODS: FACS-purified rat beta cells and human islets were cultured with IL-1beta, IFN-gamma and/or TNF-alpha. Islets were isolated from NOD or BALB/c mice at different ages. For syngeneic islet transplantation, 2- or 3-week-old NOD islets were grafted under the kidney capsule of spontaneously diabetic NOD recipients. Chemokine and IL-15 mRNA expression and protein release were evaluated, respectively, by RT-PCR and ELISA. RESULTS: Human islets and rat beta cells express IP-10, MIP-3alpha, fractalkine and IL-15 mRNAs upon exposure to cytokines. The expression of IL-15, IP-10 and fractalkine is regulated by IFN-gamma, while the expression of MIP-3alpha is IL-1beta-dependent. Moreover, cytokines induced IL-15, IP-10, Mig, I-TAC and MIP-3alpha protein accumulation in culture medium from human islets. In vivo, there was an age-related increase in IL-15, IP-10 and MIP-3alpha expression in islets isolated from NOD mice. Following syngeneic islet transplantation, increased expression of IL-1beta, IFN-gamma, fractalkine, IP-10, MCP-1 and MIP-3alpha mRNAs were observed in the grafts. CONCLUSION/INTERPRETATION: Cytokine-exposed islets or beta cells express chemokines and IL-15. This could contribute to the recruitment and activation of mononuclear cells and development of insulitis in early Type 1 diabetes and during graft destruction.


Assuntos
Quimiocinas/metabolismo , Interferon gama/farmacologia , Interleucina-15/metabolismo , Interleucina-1/farmacologia , Ilhotas Pancreáticas/metabolismo , Camundongos Endogâmicos NOD/metabolismo , Adulto , Animais , Quimiocina CCL20 , Quimiocina CX3CL1 , Quimiocina CXCL10 , Quimiocinas CC/genética , Quimiocinas CX3C/genética , Quimiocinas CXC/genética , Humanos , Interleucina-15/genética , Proteínas Inflamatórias de Macrófagos/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar
17.
J Biol Chem ; 276(52): 48879-86, 2001 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-11687580

RESUMO

Type 1 diabetes mellitus results from an autoimmune destruction of pancreatic beta-cells. Cytokines, such as interleukin-1 beta and interferon-gamma, are putative mediators of immune-induced beta-cell death and, under in vitro conditions, cause beta-cell apoptosis. We have recently shown that interleukin-1 beta + interferon-gamma modifies the expression of >200 genes in beta-cells. Several of these genes are putative targets for the transcription factor nuclear factor-kappa B (NF-kappa B), and in subsequent experiments we showed that NF-kappa B activation is mostly pro-apoptotic in beta-cells. To identify cytokine-induced and NF-kappa B-regulated genes in primary rat beta-cells, we presently combined two experimental approaches: 1) blocking of NF-kappa B activation in cytokine-exposed beta-cells by a recombinant adenovirus (AdI kappa B((SA)2)) containing an inhibitor of NF-kappa B alpha (I kappa Bac) super-repressor (S32A/S36A) and 2) study of gene expression by microarray analysis. We identified 66 cytokine-modified and NF-kappa B-regulated genes in beta-cells. Cytokine-induced NF-kappa B activation decreased Pdx-1 and increased c-Myc expression. This, together with NF-kappa B-dependent inhibition of Glut-2, pro-hormone convertase-1, and Isl-1 expression, probably contributes to the loss of differentiated beta-cell functions. NF-kappa B also regulates several genes encoding for chemokines and cytokines in beta-cells. The present data suggest that NF-kappa B is a key "switch regulator" of transcription factors and gene networks controlling cytokine-induced beta-cell dysfunction and death.


Assuntos
Citocinas/farmacologia , Regulação da Expressão Gênica , Ilhotas Pancreáticas/fisiologia , NF-kappa B/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Células Cultivadas , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Proteínas I-kappa B/genética , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Modelos Biológicos , NF-kappa B/antagonistas & inibidores , NF-kappa B/farmacologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
18.
Diabetes ; 50(10): 2219-24, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11574401

RESUMO

Cytokine-induced beta-cell death is an important event in the pathogenesis of type 1 diabetes. The transcription factor nuclear factor-kappaB (NF-kappaB) is activated by interleukin-1beta (IL-1beta), and its activity promotes the expression of several beta-cell genes, including pro- and anti-apoptotic genes. To elucidate the role of cytokine (IL-1beta + gamma-interferon [IFN-gamma])-induced expression of NF-kappaB in beta-cell apoptosis, rat beta-cells were infected with the recombinant adenovirus AdIkappaB((SA)2), which contained a nondegradable mutant form of inhibitory kappaB (IkappaB((SA)2), with S32A and S36A) that locks NF-kappaB in a cytosolic protein complex, preventing its nuclear action. Expression of IkappaB((SA)2) inhibited cytokine-stimulated nuclear translocation and DNA-binding of NF-kappaB. Cytokine-induced gene expression of several NF-kappaB targets, namely inducible nitric oxide synthase, Fas, and manganese superoxide dismutase, was prevented by AdIkappaB((SA)2), as established by reverse transcriptase-polymerase chain reaction, protein blot, and measurement of nitrite in the medium. Finally, beta-cell survival after IL-1beta + IFN-gamma treatment was significantly improved by IkappaB((SA)2) expression, mostly through inhibition of the apoptotic pathway. Based on these findings, we conclude that NF-kappaB activation, under in vitro conditions, has primarily a pro-apoptotic function in beta-cells.


Assuntos
Apoptose/fisiologia , Interferon gama/farmacologia , Interleucina-1/farmacologia , Ilhotas Pancreáticas/fisiologia , NF-kappa B/antagonistas & inibidores , Adenoviridae/genética , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Proteínas I-kappa B/genética , Proteínas I-kappa B/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar
19.
Diabetes ; 50(5): 909-20, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11334433

RESUMO

Type 1 diabetes is an autoimmune disease resulting from the selective destruction of insulin-producing beta-cells. Cytokines may contribute to pancreatic beta-cell death in type 1 diabetes. beta-cell exposure to interleukin (IL)-1beta induces functional impairment, whereas beta-cell culture for 6-9 days in the presence of IL-1beta and interferon (INF)-gamma leads to apoptosis. To clarify the mechanisms involved in these effects of cytokines, we studied the general pattern of cytokine-induced gene expression in beta-cells. Primary rat beta-cells were fluorescence-activated cell sorter-purified and exposed for 6 or 24 h to control condition, IL-1beta + INF-gamma, or IL-1beta alone (24 h only). Gene expression profile was analyzed in duplicate by oligonucleotide arrays. Nearly 3,000 transcripts were detected in controls and cytokine-treated beta-cells. Of these, 96 and 147 displayed changes in expression after 6 and 24 h, respectively, of exposure to IL-1beta + INF-gamma, whereas 105 transcripts were modified after a 24-h exposure to IL-1beta. The cytokine-responsive genes were clustered according to their biological functions. The major clusters observed were metabolism, signal transduction, transcription factors, protein synthesis/ processing, hormones, and related receptors. These modifications in gene expression may explain some of the cytokine effects in beta-cells, such as decreased protein biosynthesis and insulin release. In addition, there was induction of diverse cytokines and chemokines; this suggests that beta-cells may contribute to mononuclear cell homing during insulitis. Several of the cytokine-induced genes are potentially regulated by the transcription factor NF-kappaB. Clarification of the function of the identified cytokine-induced gene patterns may unveil some of the mechanisms involved in beta-cell damage and repair in type 1 diabetes.


Assuntos
Citocinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1/farmacologia , Ilhotas Pancreáticas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Transcrição Gênica/efeitos dos fármacos , Animais , Células Cultivadas , Enzimas/genética , Regulação da Expressão Gênica/fisiologia , Ilhotas Pancreáticas/efeitos dos fármacos , Cinética , Masculino , Proteínas/genética , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Pediatr Res ; 48(6): 748-53, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11102541

RESUMO

The pyruvate dehydrogenase (PDH) complex is situated at a key position in energy metabolism and is responsible for the conversion of pyruvate to acetyl CoA. In the literature, two unrelated patients with a PDH complex deficiency and splicing out of exon 6 of the PDH E1 alpha gene have been described, although intronic/exonic boundaries on either side of exon 6 were completely normal. Analysis of exon 6 in genomic DNA of these patients revealed two exonic mutations, a silent and a missense mutation. Although not experimentally demonstrated, the authors in both publications suggested that the exonic mutations were responsible for the exon skipping. In this work, we were able to demonstrate, by performing splicing experiments, that the two exonic mutations described in the PDH E1 alpha gene lead to aberrant splicing. We observed a disruption of the predicted wild-type pre-mRNA secondary structure of exon 6 by the mutated sequences described. However, when we constructed mutations that either reverted or disrupted the wild-type predicted pre-mRNA secondary structure of exon 6, we were unable to establish a correlation between the aberrant splicing and disruption of the predicted structure. The mutagenic experiments described here and the silent mutation found in one of the patients suggest the presence of an exonic splicing enhancer in the middle region of exon 6 of the PDH E1alpha gene.


Assuntos
Éxons/genética , Piruvato Desidrogenase (Lipoamida) , Doença da Deficiência do Complexo de Piruvato Desidrogenase/genética , Complexo Piruvato Desidrogenase/genética , Splicing de RNA/genética , Substituição de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Análise Mutacional de DNA , Humanos , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Doença da Deficiência do Complexo de Piruvato Desidrogenase/enzimologia , Precursores de RNA/química , Precursores de RNA/genética , Proteínas Recombinantes de Fusão/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Spliceossomos/metabolismo , Transfecção , Cromossomo X/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA