Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Diabetologia ; 57(3): 502-11, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24352375

RESUMO

AIMS/HYPOTHESIS: Cytotoxic T cells and macrophages contribute to beta cell destruction in type 1 diabetes at least in part through the production of cytokines such as IL-1ß, IFN-γ and TNF-α. We have recently shown the IL-17 pathway to be activated in circulating T cells and pancreatic islets of type 1 diabetes patients. Here, we studied whether IL-17A upregulates the production of chemokines by human pancreatic islets, thus contributing to the build-up of insulitis. METHODS: Human islets (from 18 donors), INS-1E cells and islets from wild-type and Stat1 knockout mice were studied. Dispersed islet cells were left untreated, or were treated with IL-17A alone or together with IL-1ß+IFN-γ or TNF-α+IFN-γ. RNA interference was used to knock down signal transducer and activator of transcription 1 (STAT1). Chemokine expression was assessed by quantitative RT-PCR, ELISA and histology. Cell viability was evaluated with nuclear dyes. RESULTS: IL-17A augmented IL-1ß+IFN-γ- and TNF-α+IFN-γ-induced chemokine mRNA and protein expression, and apoptosis in human islets. Beta cells were at least in part the source of chemokine production. Knockdown of STAT1 in human islets prevented cytokine- or IL-17A+cytokine-induced apoptosis and the expression of particular chemokines, e.g. chemokine (C-X-C motif) ligands 9 and 10. Similar observations were made in islets isolated from Stat1 knockout mice. CONCLUSIONS/INTERPRETATION: Our findings indicate that IL-17A exacerbates proinflammatory chemokine expression and secretion by human islets exposed to cytokines. This suggests that IL-17A contributes to the pathogenesis of type 1 diabetes by two mechanisms, namely the exacerbation of beta cell apoptosis and increased local production of chemokines, thus potentially aggravating insulitis.


Assuntos
Apoptose , Diabetes Mellitus Tipo 1/metabolismo , Inflamação/metabolismo , Interleucina-17/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Apoptose/imunologia , Western Blotting , Diabetes Mellitus Tipo 1/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Inflamação/imunologia , Ilhotas Pancreáticas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Necrose Tumoral alfa/metabolismo
2.
Ann Neurol ; 72(6): 971-82, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23280845

RESUMO

OBJECTIVE: Friedreich ataxia (FRDA) is an autosomal recessive neurodegenerative disease caused in almost all cases by homozygosity for a GAA trinucleotide repeat expansion in the frataxin gene. Frataxin is a mitochondrial protein involved in iron homeostasis. FRDA patients have a high prevalence of diabetes, the pathogenesis of which is not known. We aimed to evaluate the relative contribution of insulin resistance and ß-cell failure and the pathogenic mechanisms involved in FRDA diabetes. METHODS: Forty-one FRDA patients, 26 heterozygous carriers of a GAA expansion, and 53 controls underwent oral and intravenous glucose tolerance tests. ß-Cell proportion was quantified in postmortem pancreas sections from 9 unrelated FRDA patients. Using an in vitro disease model, we studied how frataxin deficiency affects ß-cell function and survival. RESULTS: FRDA patients had increased abdominal fat and were insulin resistant. This was not compensated for by increased insulin secretion, resulting in a markedly reduced disposition index, indicative of pancreatic ß-cell failure. Loss of glucose tolerance was driven by ß-cell dysfunction, which correlated with abdominal fatness. In postmortem pancreas sections, pancreatic islets of FRDA patients had a lower ß-cell content. RNA interference-mediated frataxin knockdown impaired glucose-stimulated insulin secretion and induced apoptosis in rat ß cells and human islets. Frataxin deficiency sensitized ß cells to oleate-induced and endoplasmic reticulum stress-induced apoptosis, which could be prevented by the incretins glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide. INTERPRETATION: Pancreatic ß-cell dysfunction is central to diabetes development in FRDA as a result of mitochondrial dysfunction and higher sensitivity to metabolic and endoplasmic reticulum stress-induced ß-cell death.


Assuntos
Diabetes Mellitus/etiologia , Diabetes Mellitus/patologia , Ataxia de Friedreich/complicações , Células Secretoras de Insulina/fisiologia , Proteínas de Ligação ao Ferro/genética , Expansão das Repetições de Trinucleotídeos/genética , Tecido Adiposo/metabolismo , Adulto , Animais , Distribuição da Gordura Corporal , Metabolismo Energético/genética , Saúde da Família , Feminino , Citometria de Fluxo , Ataxia de Friedreich/genética , Teste de Tolerância a Glucose , Humanos , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Resistência à Insulina/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Ratos , Frataxina
3.
J Biol Chem ; 286(2): 929-41, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-20980260

RESUMO

Cytokines produced by islet-infiltrating immune cells induce ß-cell apoptosis in type 1 diabetes. The IFN-γ-regulated transcription factors STAT1/IRF-1 have apparently divergent effects on ß-cells. Thus, STAT1 promotes apoptosis and inflammation, whereas IRF-1 down-regulates inflammatory mediators. To understand the molecular basis for these differential outcomes within a single signal transduction pathway, we presently characterized the gene networks regulated by STAT1 and IRF-1 in ß-cells. This was done by using siRNA approaches coupled to microarray analysis of insulin-producing cells exposed or not to IL-1ß and IFN-γ. Relevant microarray findings were further studied in INS-1E cells and primary rat ß-cells. STAT1, but not IRF-1, mediates the cytokine-induced loss of the differentiated ß-cell phenotype, as indicated by decreased insulin, Pdx1, MafA, and Glut2. Furthermore, STAT1 regulates cytokine-induced apoptosis via up-regulation of the proapoptotic protein DP5. STAT1 and IRF-1 have opposite effects on cytokine-induced chemokine production, with IRF-1 exerting negative feedback inhibition on STAT1 and downstream chemokine expression. The present study elucidates the transcriptional networks through which the IFN-γ/STAT1/IRF-1 axis controls ß-cell function/differentiation, demise, and islet inflammation.


Assuntos
Apoptose/imunologia , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Pancreatite/imunologia , Pancreatite/patologia , Fator de Transcrição STAT1/imunologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Retroalimentação Fisiológica/fisiologia , Técnicas de Silenciamento de Genes , Fator Regulador 1 de Interferon/imunologia , Fator Regulador 1 de Interferon/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-1beta/farmacologia , Masculino , Neuropeptídeos/genética , Neuropeptídeos/imunologia , RNA Interferente Pequeno , Ratos , Ratos Wistar , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transcrição Gênica/imunologia
4.
J Biol Chem ; 286(45): 39632-43, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21937453

RESUMO

Type 1 diabetes is characterized by local inflammation (insulitis) in the pancreatic islets causing ß-cell loss. The mitochondrial pathway of apoptosis is regulated by the balance and interaction between Bcl-2 members. Here we clarify the molecular mechanism of ß-cell death triggered by the pro-inflammatory cytokines tumor necrosis factor (TNF)-α and interferon (IFN)-γ. The combination of TNF-α + IFN-γ induced DP5, p53 up-regulated modulator of apoptosis (PUMA), and Bim expression in human islets and rodent ß-cells. DP5 and PUMA inactivation by RNA interference partially protected against TNF-α + IFN-γ-induced ß-cell apoptosis. DP5 knock-out mice had increased ß-cell area, and isolated islets from these mice were resistant to cytokine exposure. Bim expression was transcriptionally regulated by STAT1, and its activation triggered cleavage of caspases. Silencing of Bim protected rodent and human ß-cells to a large extent against TNF-α + IFN-γ, indicating a major role of this BH3-only activator protein in the mechanism of apoptosis. Our data support a highly regulated and context-dependent modulation of specific Bcl-2 members controlling the mitochondrial pathway of ß-cell apoptosis during insulitis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Células Secretoras de Insulina/metabolismo , Interferon gama/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Idoso , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Inativação Gênica , Humanos , Interferon gama/genética , Interferon gama/farmacologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Proteínas Proto-Oncogênicas/genética , Fator de Transcrição STAT1/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
5.
Hum Mol Genet ; 19(1): 135-46, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19825843

RESUMO

beta-Cell destruction in type 1 diabetes (T1D) is at least in part consequence of a 'dialog' between beta-cells and immune system. This dialog may be affected by the individual's genetic background. We presently evaluated whether modulation of MDA5 and PTPN2, two candidate genes for T1D, affects beta-cell responses to double-stranded RNA (dsRNA), a by-product of viral replication. These genes were selected following comparison between known candidate genes for T1D and genes expressed in pancreatic beta-cells, as identified in previous array analysis. INS-1E cells and primary fluorescence-activated cell sorting-purified rat beta-cells were transfected with small interference RNAs (siRNAs) targeting MDA5 or PTPN2 and subsequently exposed to intracellular synthetic dsRNA (polyinosinic-polycitidilic acid-PIC). Real-time RT-PCR, western blot and viability assays were performed to characterize gene/protein expression and viability. PIC increased MDA5 and PTPN2 mRNA expression, which was inhibited by the specific siRNAs. PIC triggered apoptosis in INS-1E and primary beta-cells and this was augmented by PTPN2 knockdown (KD), although inhibition of MDA5 did not modify PIC-induced apoptosis. In contrast, MDA5 silencing decreased PIC-induced cytokine and chemokine expression, although inhibition of PTPN2 induced minor or no changes in these inflammatory mediators. These findings indicate that changes in MDA5 and PTPN2 expression modify beta-cell responses to dsRNA. MDA5 regulates inflammatory signals, whereas PTPN2 may function as a defence mechanism against pro-apoptotic signals generated by dsRNA. These two candidate genes for T1D may thus modulate beta-cell apoptosis and/or local release of inflammatory mediators in the course of a viral infection by acting, at least in part, at the pancreatic beta-cell level.


Assuntos
RNA Helicases DEAD-box/metabolismo , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , RNA de Cadeia Dupla/farmacologia , Vírus/metabolismo , Animais , Apoptose/efeitos dos fármacos , Quimiocinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Interferon beta/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , NF-kappa B/metabolismo , Poli I-C/farmacologia , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar
6.
Biochim Biophys Acta ; 1793(11): 1768-75, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19747951

RESUMO

Enteroviral infections are associated with type I diabetes. The mechanisms by which viruses or viral products such as double-stranded RNA (dsRNA) affect pancreatic beta cell function and survival remain unclear. We have shown that extracellular dsRNA induces beta cell death via Toll-like receptor-3 (TLR3) signaling whereas cytosolic dsRNA triggers the production of type I interferons and apoptosis via a TLR3-independent process. We presently examined expression of the intracellular viral RNA sensors, the RNA helicases RIG-I and MDA5, and documented the functionality of RIG-I in pancreatic beta cells. FACS-purified rat beta cells and islet cells from wild-type or TLR3(-/-) mice were cultured with or without the RIG-I-specific ligand 5'-triphosphate single-stranded RNA (5'triP-ssRNA), the synthetic dsRNA polyI:C (PIC) or 5'OH-ssRNA (negative control); the RNA compounds were added in the medium or transfected in the cells using lipofectamine. RIG-I and MDA5 expression were determined by real-time RT-PCR. NF-kappaB and IFN-beta promoter activation were studied in the presence or absence of a dominant-negative form of RIG-I (DN-RIG-I). Both extracellular (PICex) and intracellular (PICin) PIC increased expression of RIG-I and MDA5 in pancreatic beta cells. TLR3 deletion abolished PICex-induced up-regulation of the helicases in beta cells but not in dendritic cells. PICin-induced NF-kappaB and IFN-beta promoter activation were prevented by the DN-RIG-I. The RIG-I-specific ligand 5'triP-ssRNA induced IFN-beta promoter activation and beta cell apoptosis. Our results suggest that the RIG-I pathway is present and active in beta cells and could contribute to the induction of insulitis by viral RNA intermediates.


Assuntos
Citosol/enzimologia , Infecções por Enterovirus/enzimologia , Enterovirus/metabolismo , Células Secretoras de Insulina/enzimologia , Proteínas de Membrana/biossíntese , Proteínas do Tecido Nervoso/biossíntese , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Citosol/virologia , RNA Helicases DEAD-box/biossíntese , Infecções por Enterovirus/genética , Deleção de Genes , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Células Secretoras de Insulina/virologia , Helicase IFIH1 Induzida por Interferon , Interferon beta/biossíntese , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Poli I-C/farmacologia , Regiões Promotoras Genéticas/genética , Ratos , Ratos Wistar , Receptores de Superfície Celular , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
7.
J Leukoc Biol ; 84(3): 713-20, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18567840

RESUMO

Overexpression of CD95 (Fas/Apo-1) ligand (CD95L) has been shown to induce T cell tolerance but also, neutrophilic inflammation and rejection of allogeneic tissue. We explored the capacity of dendritic cells (DCs) genetically engineered to overexpress CD95L to induce an antitumor response. We first found that DCs overexpressing CD95L, in addition to MHC class I-restricted OVA peptides (CD95L-OVA-DCs), induced increased antigen-specific CD8(+) T cell responses as compared with DCs overexpressing OVA peptides alone. The enhanced T cell responses were associated with improved regression of a tumor expressing OVA, allowing survival of all animals. When DCs overexpressing CD95L (CD95L-DCs) were injected with the tumor expressing OVA, in vivo tumor proliferation was strikingly inhibited. A strong cellular apoptosis and a massive neutrophilic infiltrate developed in this setting. Neutrophil depletion prevented tumor regression as well as enhanced IFN-gamma production induced by CD95L-OVA-DCs. Furthermore, the CD8(+) T cell response induced by the coadministration of tumor cells and CD95L-DCs led to rejection of a tumor implanted at a distance from the DC injection site. In summary, DCs expressing CD95L promote tumor rejection involving neutrophil-mediated innate immunity and CD8(+) T cell-dependent adaptative immune responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/fisiologia , Proteína Ligante Fas/metabolismo , Neutrófilos/imunologia , Ovalbumina/imunologia , Timoma/prevenção & controle , Animais , Apoptose/imunologia , Medula Óssea , Comunicação Celular , Células Dendríticas/citologia , Proteína Ligante Fas/imunologia , Genes MHC Classe I , Imunização , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/genética , Ovalbumina/metabolismo , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Retroviridae/genética , Transplante de Pele , Taxa de Sobrevida , Linfócitos T Citotóxicos/imunologia , Timoma/imunologia , Timoma/metabolismo , Transfecção , Vacinação , Receptor fas/metabolismo
8.
Transplantation ; 79(6): 648-54, 2005 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-15785370

RESUMO

BACKGROUND: Allografts are occasionally accepted in the absence of immunosuppression. Because naturally occurring CD4(+)CD25(+) regulatory T cells (natural CD25(+) Treg cells) have been shown to inhibit allograft rejection, we investigated their influence on the outcome of allografts in nonimmunosuppressed mouse recipients. METHODS: We compared survival times of male CBA/Ca skin grafts in female CBA/Ca recipients expressing a transgenic anti-HY T-cell receptor on a RAG-1(+/+) (A1[M]RAG+) or a RAG-1(-/-) (A1[M]RAG-) background. Depletion of natural CD25(+) Treg cells in A1[M]RAG+ mice was achieved by in vivo administration of the PC61 monoclonal antibody. The influence of natural CD25(+) Treg cells on the fate of major histocompatibility complex class II-mismatched (C57BL/6X bm12)F1 skin or bm12 heart transplants in C57BL/6 recipients was also assessed. Finally, we investigated the impact of natural CD25(+) Treg cells on the production of T-helper (Th)1 and Th2 cytokines in mixed lymphocyte cultures between C57BL/6 CD4(+) CD25(-) T cells as responders and bm12 or (C57BL/6X bm12)F1 antigen-presenting cells as stimulators. RESULTS: Male allografts were spontaneously accepted by female A1(M)RAG+ mice but readily rejected by female A1(M)RAG+ mice depleted of natural CD25(+) Treg cells by pretreatment with the PC61 monoclonal antibody. Depletion of CD25(+) Treg cells also enhanced eosinophil-determined rejection of (C57BL/6X bm12)F1 skin grafts or bm12 cardiac grafts in C57BL/6 recipients. Finally, natural CD25(+) Treg cells inhibited the production of interleukin (IL)-2, interferon-gamma, IL-5, and IL-13 in mixed lymphocyte culture in a dose-dependent manner. CONCLUSION: Natural CD25(+) Treg cells control Th1- and Th2-type allohelper T-cell responses and thereby influence the fate of allografts in nonimmunosuppressed recipients.


Assuntos
Sobrevivência de Enxerto/imunologia , Receptores de Interleucina-2/imunologia , Linfócitos T/imunologia , Animais , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Transplante de Coração/patologia , Terapia de Imunossupressão , Teste de Cultura Mista de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ratos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Interleucina-2/metabolismo , Transplante de Pele/imunologia , Transplante de Pele/patologia , Linfócitos T/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Transplante , Transplante Homólogo/imunologia
9.
Methods Mol Biol ; 820: 179-94, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22131032

RESUMO

Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by immune infiltration of the pancreatic islets resulting in an inflammatory reaction named insulitis and subsequent beta cell apoptosis. During the course of insulitis beta cell death is probably caused by direct contact with activated macrophages and T-cells, and/or exposure to soluble mediators secreted by these cells, including cytokines, nitric oxide, and free oxygen radicals. In vitro exposure of beta cells to the cytokines interleukin(IL)-1ß + interferon(IFN)-γ or to tumor necrosis factor(TNF)-α + IFN-γ induces beta cell dysfunction and ultimately apoptosis. The transcription factors NF-κB and STAT1 are key regulators of cytokine-induced beta cell death. However, little is known about the gene networks regulated by these (or other) transcription factors that trigger beta cell apoptosis. The recent development of RNA interference (RNAi) technology offers a unique opportunity to decipher the cytokine-activated molecular pathways responsible for beta cell death. Use of RNAi has been hampered by technical difficulties in transfecting primary beta cells, but in recent years we have succeeded in developing reliable and reproducible protocols for RNAi in beta cells. This chapter details the methods and settings used to achieve efficient and nontoxic transfection of small interfering RNA in immortal and primary beta cells.


Assuntos
Citocinas/metabolismo , Células Secretoras de Insulina/metabolismo , Interferência de RNA , Transdução de Sinais , Animais , Apoptose , Morte Celular , Sobrevivência Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/fisiopatologia , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Células Secretoras de Insulina/citologia , Insulinoma/metabolismo , Interleucina-1beta/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/patologia , Masculino , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Wistar , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fatores de Transcrição/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
10.
PLoS One ; 7(2): e31062, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22347430

RESUMO

In the course of Type 1 diabetes pro-inflammatory cytokines (e.g., IL-1ß, IFN-γ and TNF-α) produced by islet-infiltrating immune cells modify expression of key gene networks in ß-cells, leading to local inflammation and ß-cell apoptosis. Most known cytokine-induced transcription factors have pro-apoptotic effects, and little is known regarding "protective" transcription factors. To this end, we presently evaluated the role of the transcription factor CCAAT/enhancer binding protein delta (C/EBPδ) on ß-cell apoptosis and production of inflammatory mediators in the rat insulinoma INS-1E cells, in purified primary rat ß-cells and in human islets. C/EBPδ is expressed and up-regulated in response to the cytokines IL-1ß and IFN-γ in rat ß-cells and human islets. Small interfering RNA-mediated C/EBPδ silencing exacerbated IL-1ß+IFN-γ-induced caspase 9 and 3 cleavage and apoptosis in these cells. C/EBPδ deficiency increased the up-regulation of the transcription factor CHOP in response to cytokines, enhancing expression of the pro-apoptotic Bcl-2 family member BIM. Interfering with C/EBPδ and CHOP or C/EBPδ and BIM in double knockdown approaches abrogated the exacerbating effects of C/EBPδ deficiency on cytokine-induced ß-cell apoptosis, while C/EBPδ overexpression inhibited BIM expression and partially protected ß-cells against IL-1ß+IFN-γ-induced apoptosis. Furthermore, C/EBPδ silencing boosted cytokine-induced production of the chemokines CXCL1, 9, 10 and CCL20 in ß-cells by hampering IRF-1 up-regulation and increasing STAT1 activation in response to cytokines. These observations identify a novel function of C/EBPδ as a modulatory transcription factor that inhibits the pro-apoptotic and pro-inflammatory gene networks activated by cytokines in pancreatic ß-cells.


Assuntos
Apoptose , Proteína delta de Ligação ao Facilitador CCAAT/fisiologia , Células Secretoras de Insulina/patologia , Animais , Linhagem Celular , Citocinas/biossíntese , Humanos , Inflamação , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma , Fator Regulador 1 de Interferon , Ratos , Fator de Transcrição STAT1
11.
Diabetes ; 60(12): 3279-88, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21984578

RESUMO

OBJECTIVE: Genome-wide association studies allowed the identification of several associations between specific loci and type 1 diabetes (T1D). However, the mechanisms by which most candidate genes predispose to T1D remain unclear. We presently evaluated the mechanisms by which PTPN2, a candidate gene for T1D, modulates ß-cell apoptosis after exposure to type I and II interferons (IFNs), cytokines that contribute to ß-cell loss in early T1D. RESEARCH DESIGN AND METHODS: Small interfering RNAs were used to inhibit PTPN2, STAT1, Bim, and Jun NH(2)-terminal kinase 1 (JNK1) expression. Cell death was assessed by Hoechst and propidium iodide staining. BAX translocation, Bim phosphorylation, cytochrome c release, and caspases 9 and 3 activation were measured by Western blot or immunofluorescence. RESULTS: PTPN2 knockdown exacerbated type I IFN-induced apoptosis in INS-1E, primary rat, and human ß-cells. PTPN2 silencing and exposure to type I and II IFNs induced BAX translocation to the mitochondria, cytochrome c release, and caspase 3 activation. There was also an increase in Bim phosphorylation that was at least in part regulated by JNK1. Of note, both Bim and JNK1 knockdown protected ß-cells against IFN-induced apoptosis in PTPN2-silenced cells. CONCLUSIONS: The present findings suggest that local IFN production may interact with a genetic factor (PTPN2) to induce aberrant proapoptotic activity of the BH3-only protein Bim, resulting in increased ß-cell apoptosis via JNK activation and the intrinsic apoptotic pathway. This is the first indication of a direct interaction between a candidate gene for T1D and the activation of a specific downstream proapoptotic pathway in ß-cells.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Proteínas de Membrana/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Idoso , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteína 11 Semelhante a Bcl-2 , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Imunofluorescência , Humanos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Proteínas de Membrana/genética , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , Ratos , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo
12.
APMIS ; 119(2): 93-102, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21208276

RESUMO

Zinc is essential for the crystallization of insulin in pancreatic ß-cells and is thought to induce apoptosis in a dose-dependent manner, thereby regulating ß-cell mass. Therefore, a tight intracellular regulation of Zn²(+) is required. The zinc-transporter family SLC30A is an important factor in the regulation of zinc homeostasis. The aim of this study was to examine the effect of the zinc transporters ZnT3 and ZnT8 on insulin metabolism and apoptosis. Both these proteins are present in pancreatic ß-cells and have been linked to diabetes. The objective of our study was to perform a considerable siRNA-mediated knock-down of ZnT3 and ZnT8 in INS-1E cells, a pancreatic ß-cell model, and afterwards examine the impact on cell viability and insulin metabolism. Increased levels of apoptosis were observed after knock-down of both ZnT3 and ZnT8. Insulin secretion was significantly reduced by ZnT3 knock-down, whereas knock-down of ZnT8 resulted in increased intracellular content of insulin accompanied by a relatively lowered secretion. Both zinc transporters in this way seem to play a role in ß-cell survival and the ability of these cells to react appropriately to surrounding glucose concentrations.


Assuntos
Apoptose , Proteínas de Transporte de Cátions/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , RNA Interferente Pequeno/genética , Animais , Proteínas de Transporte de Cátions/antagonistas & inibidores , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/patologia , Ratos , Transportador 8 de Zinco
13.
Diabetes ; 60(8): 2112-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21659501

RESUMO

OBJECTIVE: CD4 T-cells secreting interleukin (IL)-17 are implicated in several human autoimmune diseases, but their role in type 1 diabetes has not been defined. To address the relevance of such cells, we examined IL-17 secretion in response to ß-cell autoantigens, IL-17A gene expression in islets, and the potential functional consequences of IL-17 release for ß-cells. RESEARCH DESIGN AND METHODS: Peripheral blood CD4 T-cell responses to ß-cell autoantigens (proinsulin, insulinoma-associated protein, and GAD65 peptides) were measured by IL-17 enzyme-linked immunospot assay in patients with new-onset type 1 diabetes (n = 50). mRNA expression of IL-17A and IFNG pathway genes was studied by qRT-PCR using islets obtained from subjects who died 5 days and 10 years after diagnosis of disease, respectively, and from matched control subjects. IL-17 effects on the function of human islets, rat ß-cells, and the rat insulinoma cell line INS-1E were examined. RESULTS: A total of 27 patients (54%) showed IL-17 reactivity to one or more ß-cell peptides versus 3 of 30 (10%) control subjects (P = 0.0001). In a single case examined close to diagnosis, islet expression of IL17A, RORC, and IL22 was detected. It is noteworthy that we show that IL-17 mediates significant and reproducible enhancement of IL-1ß/interferon (IFN)-γ-induced and tumor necrosis factor (TNF)-α/IFN-γ-induced apoptosis in human islets, rat ß-cells, and INS-1E cells, in association with significant upregulation of ß-cell IL17RA expression via activation of the transcription factors STAT1 and nuclear factor (NF)-κB. CONCLUSIONS: Circulating IL-17(+) ß-cell-specific autoreactive CD4 T-cells are a feature of type 1 diabetes diagnosis. We disclose a novel pathway to ß-cell death involving IL-17 and STAT1 and NF-κB, rendering this cytokine a novel disease biomarker and potential therapeutic target.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Células Secretoras de Insulina/patologia , Interleucina-17/fisiologia , Adolescente , Adulto , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/fisiologia , Feminino , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma/metabolismo , Interleucina-17/biossíntese , Interleucinas/biossíntese , Masculino , NF-kappa B/fisiologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Neoplasias Pancreáticas/metabolismo , Ratos , Ratos Wistar , Fator de Transcrição STAT1/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Interleucina 22
14.
Diabetes ; 59(2): 358-74, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19934004

RESUMO

OBJECTIVE: Cytokines contribute to pancreatic beta-cell death in type 1 diabetes. This effect is mediated by complex gene networks that remain to be characterized. We presently utilized array analysis to define the global expression pattern of genes, including spliced variants, modified by the cytokines interleukin (IL)-1beta + interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha + IFN-gamma in primary rat beta-cells. RESEARCH DESIGN AND METHODS: Fluorescence-activated cell sorter-purified rat beta-cells were exposed to IL-1beta + IFN-gamma or TNF-alpha + IFN-gamma for 6 or 24 h, and global gene expression was analyzed by microarray. Key results were confirmed by RT-PCR, and small-interfering RNAs were used to investigate the mechanistic role of novel and relevant transcription factors identified by pathway analysis. RESULTS Nearly 16,000 transcripts were detected as present in beta-cells, with temporal differences in the number of genes modulated by IL-1beta + IFNgamma or TNF-alpha + IFN-gamma. These cytokine combinations induced differential expression of inflammatory response genes, which is related to differential induction of IFN regulatory factor-7. Both treatments decreased the expression of genes involved in the maintenance of beta-cell phenotype and growth/regeneration. Cytokines induced hypoxia-inducible factor-alpha, which in this context has a proapoptotic role. Cytokines also modified the expression of >20 genes involved in RNA splicing, and exon array analysis showed cytokine-induced changes in alternative splicing of >50% of the cytokine-modified genes. CONCLUSIONS: The present study doubles the number of known genes expressed in primary beta-cells, modified or not by cytokines, and indicates the biological role for several novel cytokine-modified pathways in beta-cells. It also shows that cytokines modify alternative splicing in beta-cells, opening a new avenue of research for the field.


Assuntos
Processamento Alternativo/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Interleucina-1beta/farmacologia , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Ciclo do Ácido Cítrico/efeitos dos fármacos , Ciclo do Ácido Cítrico/genética , Citrulina/metabolismo , Citocinas/farmacologia , Éxons/genética , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Incretinas/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA Interferente Pequeno/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética
15.
Diabetes ; 58(6): 1283-91, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19336676

RESUMO

OBJECTIVE: The pathogenesis of type 1 diabetes has a strong genetic component. Genome-wide association scans recently identified novel susceptibility genes including the phosphatases PTPN22 and PTPN2. We hypothesized that PTPN2 plays a direct role in beta-cell demise and assessed PTPN2 expression in human islets and rat primary and clonal beta-cells, besides evaluating its role in cytokine-induced signaling and beta-cell apoptosis. RESEARCH DESIGN AND METHODS: PTPN2 mRNA and protein expression was evaluated by real-time PCR and Western blot. Small interfering (si)RNAs were used to inhibit the expression of PTPN2 and downstream STAT1 in beta-cells, allowing the assessment of cell death after cytokine treatment. RESULTS: PTPN2 mRNA and protein are expressed in human islets and rat beta-cells and upregulated by cytokines. Transfection with PTPN2 siRNAs inhibited basal- and cytokine-induced PTPN2 expression in rat beta-cells and dispersed human islets cells. Decreased PTPN2 expression exacerbated interleukin (IL)-1beta + interferon (IFN)-gamma-induced beta-cell apoptosis and turned IFN-gamma alone into a proapoptotic signal. Inhibition of PTPN2 amplified IFN-gamma-induced STAT1 phosphorylation, whereas double knockdown of both PTPN2 and STAT1 protected beta-cells against cytokine-induced apoptosis, suggesting that STAT1 hyperactivation is responsible for the aggravation of cytokine-induced beta-cell death in PTPN2-deficient cells. CONCLUSIONS: We identified a functional role for the type 1 diabetes candidate gene PTPN2 in modulating IFN-gamma signal transduction at the beta-cell level. PTPN2 regulates cytokine-induced apoptosis and may thereby contribute to the pathogenesis of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/fisiologia , Interferon gama/efeitos adversos , Proteína Tirosina Fosfatase não Receptora Tipo 2/genética , RNA Mensageiro/genética , Idoso , Animais , Apoptose , Morte Celular , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Mapeamento Cromossômico , Predisposição Genética para Doença , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1beta/farmacologia , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacologia , Doadores de Tecidos
16.
Biochem Soc Trans ; 36(Pt 3): 321-7, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18481950

RESUMO

Accumulating evidence indicates that beta-cells die by apoptosis in T1DM (Type 1 diabetes mellitus). Apoptosis is an active gene-directed process, and recent observations suggest that beta-cell apoptosis depends on the parallel and/or sequential up- and down-regulation of hundreds of genes controlled by key transcription factors such as NF-kappaB (nuclear factor kappaB) and STAT-1 (signal transducer and activator of transcription 1). Understanding the regulation of these gene networks, and how they modulate beta-cell death and the 'dialogue' between beta-cells and the immune system, will require a systems biology approach to the problem. This will hopefully allow the search for a cure for T1DM to move from a 'trial-and-error' approach to one that is really mechanistically driven.


Assuntos
Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/patologia , Biologia de Sistemas/métodos , Animais , Morte Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Redes Reguladoras de Genes , Humanos , Sistema Imunitário , Células Secretoras de Insulina/imunologia
17.
J Cell Sci ; 121(Pt 14): 2308-18, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559892

RESUMO

Free fatty acids (FFA) cause apoptosis of pancreatic beta-cells and might contribute to beta-cell loss in type 2 diabetes via the induction of endoplasmic reticulum (ER) stress. We studied here the molecular mechanisms implicated in FFA-induced ER stress initiation and apoptosis in INS-1E cells, FACS-purified primary beta-cells and human islets exposed to oleate and/or palmitate. Treatment with saturated and/or unsaturated FFA led to differential ER stress signaling. Palmitate induced more apoptosis and markedly activated the IRE1, PERK and ATF6 pathways, owing to a sustained depletion of ER Ca(2+) stores, whereas the unsaturated FFA oleate led to milder PERK and IRE1 activation and comparable ATF6 signaling. Non-metabolizable methyl-FFA analogs induced neither ER stress nor beta-cell apoptosis. The FFA-induced ER stress response was not modified by high glucose concentrations, suggesting that ER stress in primary beta-cells is primarily lipotoxic, and not glucolipotoxic. Palmitate, but not oleate, activated JNK. JNK inhibitors reduced palmitate-mediated AP-1 activation and apoptosis. Blocking the transcription factor CHOP delayed palmitate-induced beta-cell apoptosis. In conclusion, saturated FFA induce ER stress via ER Ca(2+) depletion. The IRE1 and resulting JNK activation contribute to beta-cell apoptosis. PERK activation by palmitate also contributes to beta-cell apoptosis via CHOP.


Assuntos
Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/patologia , Ácidos Graxos não Esterificados/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Fator 3 Ativador da Transcrição/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Retículo Endoplasmático/enzimologia , Ácidos Graxos não Esterificados/metabolismo , Glucose/toxicidade , Humanos , Células Secretoras de Insulina/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Pessoa de Meia-Idade , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição CHOP/metabolismo , eIF-2 Quinase/metabolismo
18.
PLoS One ; 2(5): e469, 2007 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-17520028

RESUMO

Kidney ischemia/reperfusion injury (I/R) is characterized by renal dysfunction and tubular damages resulting from an early activation of innate immunity. Recently, nicotine administration has been shown to be a powerful inhibitor of a variety of innate immune responses, including LPS-induced toxaemia. This cholinergic anti-inflammatory pathway acts via the alpha7 nicotinic acetylcholine receptor (alpha7nAChR). Herein, we tested the potential protective effect of nicotine administration in a mouse model of renal I/R injury induced by bilateral clamping of kidney arteries. Renal function, tubular damages and inflammatory response were compared between control animals and mice receiving nicotine at the time of ischemia. Nicotine pretreatment protected mice from renal dysfunction in a dose-dependent manner and through the alpha7nAChR, as attested by the absence of protection in alpha7nAChR-deficient mice. Additionally, nicotine significantly reduced tubular damages, prevented neutrophil infiltration and decreased productions of the CXC-chemokine KC, TNF-alpha and the proinflammatory high-mobility group box 1 protein. Reduced tubular damage in nicotine pre-treated mice was associated with a decrease in tubular cell apoptosis and proliferative response as attested by the reduction of caspase-3 and Ki67 positive cells, respectively. All together, these data highlight that nicotine exerts a protective anti-inflammatory effect during kidney I/R through the cholinergic alpha7nAChR pathway. In addition, this could provide an opportunity to overcome the effect of surgical cholinergic denervation during kidney transplantation.


Assuntos
Concentração de Íons de Hidrogênio , Rim/efeitos dos fármacos , Nicotina/farmacologia , Receptores Nicotínicos/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose , Western Blotting , Proliferação de Células , Mediadores da Inflamação/fisiologia , Rim/irrigação sanguínea , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nicotínicos/genética , Receptor Nicotínico de Acetilcolina alfa7
19.
J Immunol ; 178(3): 1301-11, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17237376

RESUMO

Maturation of dendritic cells (DC) is a critical step in the induction of T cell responses and depends on the activation of NF-kappaB transcription factors. Therefore, inhibition of NF-kappaB activation has been proposed as a strategy to maintain DC in an immature stage and to promote immune tolerance. Herein, we generated murine myeloid DC expressing a mutated IkappaBalpha acting as a superrepressor of the classical NF-kappaB pathway (s-rIkappaB DC) to investigate the consequences of NF-kappaB inhibition on the ability of DC to prime T cell responses. Upon in vitro LPS activation, maturation of s-rIkappaB DC was profoundly impaired as indicated by defective up-regulation of MHC class II and costimulatory molecules and reduced secretion of IL-12 p70 and TNF-alpha. In contrast, after injection, s-rIkappaB DC had the same capacity as control DC to migrate to draining lymph node and to induce Th1- and Th2-type cytokine production in a MHC class II-incompatible host mice. Likewise, s-rIkappaB DC pulsed with OVA were as efficient as control DC to induce Ag-specific T cell responses in vivo. Indeed, further in vitro experiments established that s-rIkappaB DC undergo efficient maturation upon prolonged contact with activated T cells via the alternative pathway of NF-kappaB activation triggered at least partly by lymphotoxin beta receptor ligation and involving processing of p100/RelB complexes.


Assuntos
Apresentação de Antígeno/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Proteínas I-kappa B/genética , NF-kappa B/metabolismo , Linfócitos T/imunologia , Animais , Células Cultivadas , Citocinas/biossíntese , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II , Lipopolissacarídeos/farmacologia , Camundongos , Mutação , Inibidor de NF-kappaB alfa
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa