Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
FEBS J ; 291(3): 566-583, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37985375

RESUMO

Butyrate, a gut microbial metabolite, has beneficial effects on glucose homeostasis and has become an attractive drug candidate for type 2 diabetes (T2D). Recently, we showed that butyrate protects pancreatic beta cells against cytokine-induced dysfunction. In this study, we explored the underlying mechanisms of butyrate action. Pancreatic mouse islets were exposed to a non-cytotoxic concentration of interleukin-1ß (IL-1ß) for 10 days to mimic low-grade inflammation in T2D. Similar to the effect of butyrate, an isoform-selective histone deacetylase 3 (HDAC3) inhibitor normalized IL-1ß-reduced glucose-stimulated insulin secretion and insulin content. In contrast, free fatty acid receptor 2 and 3 (FFAR2/3) agonists failed to normalize IL-1ß-induced beta cell dysfunction. Furthermore, butyrate inhibited HDAC activity and increased the acetylation of histone H3 and H4 by 3- and 10-fold, respectively. Genome-wide analysis of histone H3 lysine 27 acetylation (H3K27ac) revealed that butyrate mainly increased H3K27ac at promoter regions (74%), while H3K27ac peaks regulated by IL-1ß were more equally distributed at promoters (38%), introns (23%) and intergenic regions (23%). Gene ontology analysis showed that butyrate increased IL-1ß-reduced H3K27ac levels near several genes related to hormone secretion and reduced IL-1ß-increased H3K27ac levels near genes associated with inflammatory responses. Butyrate alone increased H3K27ac near many genes related to MAPK signaling, hormone secretion, and differentiation, and decreased H3K27ac at genes involved in cell replication. Together, these results suggest that butyrate prevents IL-1ß-induced pancreatic islet dysfunction by inhibition of HDACs resulting in changes in H3K27ac levels at genes relevant for beta cell function and inflammatory responses.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Camundongos , Animais , Inibidores de Histona Desacetilases/farmacologia , Interleucina-1beta/metabolismo , Histonas/genética , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Butiratos/farmacologia , Butiratos/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Hormônios/metabolismo
2.
Cell Death Dis ; 14(7): 399, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37407581

RESUMO

Insufficient insulin secretion is a hallmark of type 2 diabetes and has been attributed to beta cell identity loss characterized by decreased expression of several key beta cell genes. The pro-inflammatory factor BMP-2 is upregulated in islets of Langerhans from individuals with diabetes and acts as an inhibitor of beta cell function and proliferation. Exposure to BMP-2 induces expression of Id1-4, Hes-1, and Hey-1 which are transcriptional regulators associated with loss of differentiation. The aim of this study was to investigate the mechanism by which BMP-2 induces beta cell dysfunction and loss of cell maturity. Mouse islets exposed to BMP-2 for 10 days showed impaired glucose-stimulated insulin secretion and beta cell proliferation. BMP-2-induced beta cell dysfunction was associated with decreased expression of cell maturity and proliferation markers specific to the beta cell such as Ins1, Ucn3, and Ki67 and increased expression of Id1-4, Hes-1, and Hey-1. The top 30 most regulated proteins significantly correlated with corresponding mRNA expression. BMP-2-induced gene expression changes were associated with a predominant reduction in acetylation of H3K27 and a decrease in NeuroD1 chromatin binding activity. These results show that BMP-2 induces loss of beta cell maturity and suggest that remodeling of H3K27ac and decreased NeuroD1 DNA binding activity participate in the effect of BMP-2 on beta cell dysfunction.


Assuntos
Diabetes Mellitus Tipo 2 , Código das Histonas , Animais , Camundongos , Proteína Morfogenética Óssea 2/metabolismo , Cromatina , Diabetes Mellitus Tipo 2/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais
3.
Int J Mol Sci ; 24(5)2023 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-36901964

RESUMO

Butyrate produced by the gut microbiota has beneficial effects on metabolism and inflammation. Butyrate-producing bacteria are supported by diets with a high fiber content, such as high-amylose maize starch (HAMS). We investigated the effects of HAMS- and butyrylated HAMS (HAMSB)-supplemented diets on glucose metabolism and inflammation in diabetic db/db mice. Mice fed HAMSB had 8-fold higher fecal butyrate concentration compared to control diet-fed mice. Weekly analysis of fasting blood glucose showed a significant reduction in HAMSB-fed mice when the area under the curve for all five weeks was analyzed. Following treatment, fasting glucose and insulin analysis showed increased homeostatic model assessment (HOMA) insulin sensitivity in the HAMSB-fed mice. Glucose-stimulated insulin release from isolated islets did not differ between the groups, while insulin content was increased by 36% in islets of the HAMSB-fed mice. Expression of insulin 2 was also significantly increased in islets of the HAMSB-fed mice, while no difference in expression of insulin 1, pancreatic and duodenal homeobox 1, MAF bZIP transcription factor A and urocortin 3 between the groups was observed. Hepatic triglycerides in the livers of the HAMSB-fed mice were significantly reduced. Finally, mRNA markers of inflammation in liver and adipose tissue were reduced in mice fed HAMSB. These findings suggest that HAMSB-supplemented diet improves glucose metabolism in the db/db mice, and reduces inflammation in insulin-sensitive tissues.


Assuntos
Butiratos , Amido , Ratos , Camundongos , Animais , Ratos Sprague-Dawley , Amilose/metabolismo , Inflamação , Fígado/metabolismo , Camundongos Endogâmicos , Insulina , Homeostase , Glucose , Camundongos Endogâmicos C57BL , Glicemia/metabolismo
4.
J Biol Chem ; 298(9): 102312, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35921894

RESUMO

Cytokine-induced beta cell dysfunction is a hallmark of type 2 diabetes (T2D). Chronic exposure of beta cells to inflammatory cytokines affects gene expression and impairs insulin secretion. Thus, identification of anti-inflammatory factors that preserve beta cell function represents an opportunity to prevent or treat T2D. Butyrate is a gut microbial metabolite with anti-inflammatory properties for which we recently showed a role in preventing interleukin-1ß (IL-1ß)-induced beta cell dysfunction, but how prevention is accomplished is unclear. Here, we investigated the mechanisms by which butyrate exerts anti-inflammatory activity in beta cells. We exposed mouse islets and INS-1E cells to a low dose of IL-1ß and/or butyrate and measured expression of inflammatory genes and nitric oxide (NO) production. Additionally, we explored the molecular mechanisms underlying butyrate activity by dissecting the activation of the nuclear factor-κB (NF-κB) pathway. We found that butyrate suppressed IL-1ß-induced expression of inflammatory genes, such as Nos2, Cxcl1, and Ptgs2, and reduced NO production. Butyrate did not inhibit IκBα degradation nor NF-κB p65 nuclear translocation. Furthermore, butyrate did not affect binding of NF-κB p65 to target sequences in synthetic DNA but inhibited NF-κB p65 binding and RNA polymerase II recruitment to inflammatory gene promoters in the context of native DNA. We found this was concurrent with increased acetylation of NF-κB p65 and histone H4, suggesting butyrate affects NF-κB activity via inhibition of histone deacetylases. Together, our results show butyrate inhibits IL-1ß-induced inflammatory gene expression and NO production through suppression of NF-κB activation and thereby possibly preserves beta cell function.


Assuntos
Anti-Inflamatórios não Esteroides , Butiratos , Diabetes Mellitus Tipo 2 , Inibidores de Histona Desacetilases , Inflamação , Células Secretoras de Insulina , Interleucina-1beta , NF-kappa B , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Butiratos/farmacologia , Ciclo-Oxigenase 2/metabolismo , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/patologia , Regulação da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Histonas/metabolismo , Inflamação/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacologia , Camundongos , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , RNA Polimerase II/metabolismo
5.
Int J Mol Sci ; 22(19)2021 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-34638768

RESUMO

Pancreatic beta cell dysfunction caused by metabolic and inflammatory stress contributes to the development of type 2 diabetes (T2D). Butyrate, produced by the gut microbiota, has shown beneficial effects on glucose metabolism in animals and humans and may directly affect beta cell function, but the mechanisms are poorly described. The aim of this study was to investigate the effect of butyrate on cytokine-induced beta cell dysfunction in vitro. Mouse islets, rat INS-1E, and human EndoC-ßH1 beta cells were exposed long-term to non-cytotoxic concentrations of cytokines and/or butyrate to resemble the slow onset of inflammation in T2D. Beta cell function was assessed by glucose-stimulated insulin secretion (GSIS), gene expression by qPCR and RNA-sequencing, and proliferation by incorporation of EdU into newly synthesized DNA. Butyrate protected beta cells from cytokine-induced impairment of GSIS and insulin content in the three beta cell models. Beta cell proliferation was reduced by both cytokines and butyrate. Expressions of the beta cell specific genes Ins, MafA, and Ucn3 reduced by the cytokine IL-1ß were not affected by butyrate. In contrast, butyrate upregulated the expression of secretion/transport-related genes and downregulated inflammatory genes induced by IL-1ß in mouse islets. In summary, butyrate prevents pro-inflammatory cytokine-induced beta cell dysfunction.


Assuntos
Butiratos/farmacologia , Diabetes Mellitus Tipo 2/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Secretoras de Insulina/imunologia , Interleucina-1beta/imunologia , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/patologia , Humanos , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Ratos
6.
Diabetes ; 70(2): 464-476, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33203694

RESUMO

The single nucleotide polymorphism rs7804356 located in the Src kinase-associated phosphoprotein 2 (SKAP2) gene is associated with type 1 diabetes (T1D), suggesting SKAP2 as a causal candidate gene. The objective of the study was to investigate if SKAP2 has a functional role in the ß-cells in relation to T1D. In a cohort of children with newly diagnosed T1D, rs7804356 predicted glycemic control and residual ß-cell function during the 1st year after diagnosis. In INS-1E cells and rat and human islets, proinflammatory cytokines reduced the content of SKAP2. Functional studies revealed that knockdown of SKAP2 aggravated cytokine-induced apoptosis in INS-1E cells and primary rat ß-cells, suggesting an antiapoptotic function of SKAP2. In support of this, overexpression of SKAP2 afforded protection against cytokine-induced apoptosis, which correlated with reduced nuclear content of S536-phosphorylated nuclear factor-κB (NF-κB) subunit p65, lower nitric oxide production, and diminished CHOP expression indicative of decreased endoplasmic reticulum stress. Knockdown of CHOP partially counteracted the increase in cytokine-induced apoptosis caused by SKAP2 knockdown. In conclusion, our results suggest that SKAP2 controls ß-cell sensitivity to cytokines possibly by affecting the NF-κB-inducible nitric oxide synthase-endoplasmic reticulum stress pathway.


Assuntos
Apoptose/genética , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Polimorfismo de Nucleotídeo Único , Adolescente , Animais , Glicemia/genética , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/genética , Feminino , Técnicas de Silenciamento de Genes , Genótipo , Controle Glicêmico , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ilhotas Pancreáticas/metabolismo , Masculino , Ratos
7.
PLoS One ; 15(2): e0222432, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32053590

RESUMO

A central and still open question regarding the pathogenesis of autoimmune diseases, such as type 1 diabetes, concerns the processes that underlie the generation of MHC-presented autoantigenic epitopes that become targets of autoimmune attack. Proteasomal degradation is a key step in processing of proteins for MHC class I presentation. Different types of proteasomes can be expressed in cells dictating the repertoire of peptides presented by the MHC class I complex. Of particular interest for type 1 diabetes is the proteasomal configuration of pancreatic ß cells, as this might facilitate autoantigen presentation by ß cells and thereby their T-cell mediated destruction. Here we investigated whether so-called inducible subunits of the proteasome are constitutively expressed in ß cells, regulated by inflammatory signals and participate in the formation of active intermediate or immuno-proteasomes. We show that inducible proteasomal subunits are constitutively expressed in human and rodent islets and an insulin-secreting cell-line. Moreover, the ß5i subunit is incorporated into active intermediate proteasomes that are bound to 19S or 11S regulatory particles. Finally, inducible subunit expression along with increase in total proteasome activities are further upregulated by low concentrations of IL-1ß stimulating proinsulin biosynthesis. These findings suggest that the ß cell proteasomal repertoire is more diverse than assumed previously and may be highly responsive to a local inflammatory islet environment.


Assuntos
Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Autoantígenos/imunologia , Autoantígenos/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Interleucina-1beta/imunologia , Células Jurkat , Camundongos , Cultura Primária de Células , Proinsulina/biossíntese , Complexo de Endopeptidases do Proteassoma/imunologia , Proteólise , RNA-Seq , Regulação para Cima/imunologia
8.
Mol Cell Endocrinol ; 496: 110524, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31362031

RESUMO

Decreased insulin secretory capacity in Type 2 diabetes mellitus is associated with beta-cell dedifferentiation and inflammation. We hypothesize that prolonged exposure of beta-cells to low concentrations of IL-1ß induce beta-cell dedifferentiation characterized by impaired glucose-stimulated insulin secretion, reduced expression of key beta-cell genes and changes in histone modifications at gene loci known to affect beta-cell function. Ten days exposure to IL-1ß at non-cytotoxic concentrations reduced insulin secretion and beta-cell proliferation and decreased expression of key beta-cell identity genes, including MafA and Ucn3 and decreased H3K27ac at the gene loci, suggesting that inflammatory cytokines directly affects the epigenome. Following removal of IL-1ß, beta-cell function was normalized and mRNA expression of beta-cell identity genes, such as insulin and Ucn3 returned to pre-stimulation levels. Our findings indicate that prolonged exposure to low concentrations of IL-1ß induces epigenetic changes associated with loss of beta-cell identity as observed in Type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Regulação da Expressão Gênica , Histonas/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Diabetes Mellitus Tipo 2/patologia , Epigênese Genética , Células Secretoras de Insulina/patologia , Fatores de Transcrição Maf Maior/biossíntese , Masculino , Camundongos , Urocortinas/biossíntese
9.
Mol Cell Endocrinol ; 478: 106-114, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30121202

RESUMO

ß-Cells may be a source of IL-1ß that is produced as inactive pro-IL-1ß and processed into biologically-active IL-1ß by enzymatic cleavage mediated by the NLRP1-, NLRP3- and NLRC4-inflammasomes. Little is known about the ß-cell inflammasomes. NLRP1-expression was upregulated in islet-cells from T2D-patients and by IL-1ß+IFNγ in INS-1 cells in a histone-deacetylase dependent manner. NLRP3 was downregulated by cytokines in INS-1 cells. NLRC4 was barely expressed and not regulated by cytokines. High extracellular K+ reduced cytokine-induced apoptosis and NO production and restored cytokine-inhibited accumulated insulin-secretion. Basal inflammasome expression was JNK1-3 dependent. Knock-down of the ASC interaction domain common for NLRP1 and 3 improved insulin secretion and ameliorated IL-1ß and/or glucolipotoxicity-induced cell death and reduced cytokine-induced NO-production. Broad inflammasome-inhibition, but not NLRP3-selective inhibition, protected against IL-1ß-induced INS-1 cell-toxicity. We suggest that IL-1ß causes ß-cell toxicity in part by NLRP1 mediated caspase-1-activation and maturation of IL-1ß leading to an autocrine potentiation loop.


Assuntos
Apoptose , Inflamassomos/metabolismo , Células Secretoras de Insulina/metabolismo , Estresse Fisiológico , Animais , Apoptose/efeitos dos fármacos , Proteínas Adaptadoras de Sinalização CARD , Morte Celular/efeitos dos fármacos , Linhagem Celular , Citocinas/farmacologia , Citoproteção/efeitos dos fármacos , Feminino , Glucose/toxicidade , Histona Desacetilases/metabolismo , Humanos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Interleucina-1beta/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipídeos/toxicidade , Pessoa de Meia-Idade , Potássio/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores Purinérgicos P2X7/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Adulto Jovem
10.
J Diabetes Res ; 2016: 1312705, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26962537

RESUMO

The relative contributions of the JNK subtypes in inflammatory ß-cell failure and apoptosis are unclear. The JNK protein family consists of JNK1, JNK2, and JNK3 subtypes, encompassing many different isoforms. INS-1 cells express JNK1α1, JNK1α2, JNK1ß1, JNK1ß2, JNK2α1, JNK2α2, JNK3α1, and JNK3α2 mRNA isoform transcripts translating into 46 and 54 kDa isoform JNK proteins. Utilizing Lentiviral mediated expression of shRNAs against JNK1, JNK2, or JNK3 in insulin-producing INS-1 cells, we investigated the role of individual JNK subtypes in IL-1ß-induced ß-cell apoptosis. JNK1 knockdown prevented IL-1ß-induced INS-1 cell apoptosis associated with decreased 46 kDa isoform JNK protein phosphorylation and attenuated Myc expression. Transient knockdown of Myc also prevented IL-1ß-induced apoptosis as well as caspase 3 cleavage. JNK2 shRNA potentiated IL-1ß-induced apoptosis and caspase 3 cleavage, whereas JNK3 shRNA did not affect IL-1ß-induced ß-cell death compared to nonsense shRNA expressing INS-1 cells. In conclusion, JNK1 mediates INS-1 cell death associated with increased Myc expression. These findings underline the importance of differentiated targeting of JNK subtypes in the development of inflammatory ß-cell failure and destruction.


Assuntos
Apoptose/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Interleucina-1beta/farmacologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , Ratos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
11.
Mol Cell Endocrinol ; 420: 24-36, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26610752

RESUMO

Interleukin-1ß (IL-1ß) and interferon-γ (IFNγ) contribute to type 1 diabetes (T1D) by inducing ß-cell death. Tumor necrosis factor (TNF) receptor-associated factor (TRAF) proteins are adaptors that transduce signaling from a variety of membrane receptors including cytokine receptors. We show here that IL-1ß and IFNγ upregulate the expression of TRAF2 in insulin-producing INS-1E cells and isolated rat pancreatic islets. siRNA-mediated knockdown (KD) of TRAF2 in INS-1E cells reduced IL-1ß-induced phosphorylation of JNK1/2, but not of p38 or ERK1/2 mitogen-activated protein kinases. TRAF2 KD did not modulate NFκB activation by cytokines, but reduced cytokine-induced inducible nitric oxide synthase (iNOS) promotor activity and expression. We further observed that IFNγ-stimulated phosphorylation of STAT3 required TRAF2. KD of TRAF2 or STAT3 reduced cytokine-induced caspase 3/7 activation, but, intriguingly, potentiated cytokine-mediated loss of plasma membrane integrity and augmented the number of propidium iodide-positive cells. Finally, we found that TRAF2 KD increased cytokine-induced production of reactive oxygen species (ROS). In summary, our data suggest that TRAF2 is an important mediator of IL-1ß and IFNγ signaling in pancreatic ß-cells.


Assuntos
Células Secretoras de Insulina/citologia , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Mediadores da Inflamação/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Necrose , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação/efeitos dos fármacos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/efeitos dos fármacos
12.
PLoS One ; 9(1): e87067, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475223

RESUMO

Pancreatic ß-cell dysfunction is central to type 2 diabetes pathogenesis. Prolonged elevated levels of circulating free-fatty acids and hyperglycemia, also termed glucolipotoxicity, mediate ß-cell dysfunction and apoptosis associated with increased c-Jun N-terminal Kinase (JNK) activity. Endoplasmic reticulum (ER) and oxidative stress are elicited by palmitate and high glucose concentrations further potentiating JNK activity. Our aim was to determine the role of the JNK subtypes JNK1, JNK2 and JNK3 in palmitate and high glucose-induced ß-cell apoptosis. We established insulin-producing INS1 cell lines stably expressing JNK subtype specific shRNAs to understand the differential roles of the individual JNK isoforms. JNK activity was increased after 3 h of palmitate and high glucose exposure associated with increased expression of ER and mitochondrial stress markers. JNK1 shRNA expressing INS1 cells showed increased apoptosis and cleaved caspase 9 and 3 compared to non-sense shRNA expressing control INS1 cells when exposed to palmitate and high glucose associated with increased CHOP expression, ROS formation and Puma mRNA expression. JNK2 shRNA expressing INS1 cells did not affect palmitate and high glucose induced apoptosis or ER stress markers, but increased Puma mRNA expression compared to non-sense shRNA expressing INS1 cells. Finally, JNK3 shRNA expressing INS1 cells did not induce apoptosis compared to non-sense shRNA expressing INS1 cells when exposed to palmitate and high glucose but showed increased caspase 9 and 3 cleavage associated with increased DP5 and Puma mRNA expression. These data suggest that JNK1 protects against palmitate and high glucose-induced ß-cell apoptosis associated with reduced ER and mitochondrial stress.


Assuntos
Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Proteína Quinase 10 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/genética , Ácido Palmítico/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Linhagem Celular , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Regulação da Expressão Gênica , Humanos , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Proteína Quinase 10 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo
13.
Endocrinology ; 152(8): 3040-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21693679

RESUMO

Apolipoprotein CIII (ApoCIII) is mainly synthesized in the liver and is important for triglyceride metabolism. The plasma concentration of ApoCIII is elevated in patients with type 1 diabetes (T1D), and in vitro ApoCIII causes apoptosis in pancreatic ß-cells in the absence of inflammatory stress. Here, we investigated the effects of ApoCIII on function, signaling, and viability in intact rat pancreatic islets exposed to proinflammatory cytokines to model the intraislet inflammatory milieu in T1D. In contrast to earlier observations in mouse ß-cells, exposure of rat islets to ApoCIII alone (50 µg/ml) did not cause apoptosis. In the presence of the islet-cytotoxic cytokines IL-1ß + interferon-γ, ApoCIII reduced cytokine-mediated islet cell death and impairment of ß-cell function. ApoCIII had no effects on mitogen-activated protein kinases (c-Jun N-terminal kinase, p38, and ERK) and had no impact on IL-1ß-induced c-Jun N-terminal kinase activation. However, ApoCIII augmented cytokine-mediated nitric oxide (NO) production and inducible NO synthase expression. Further, ApoCIII caused degradation of the nuclear factor κB-inhibitor inhibitor of κB and stimulated Ser473-phosphorylation of the survival serine-threonine kinase Akt. Inhibition of the Akt signaling pathway by the phosphatidylinositol 3 kinase inhibitor LY294002 counteracted the antiapoptotic effect of ApoCIII on cytokine-induced apoptosis. We conclude that ApoCIII in the presence of T1D-relevant proinflammatory cytokines reduces rat pancreatic islet cell apoptosis via Akt.


Assuntos
Apolipoproteína C-III/farmacologia , Apoptose/efeitos dos fármacos , Citocinas/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Cálcio/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Fosforilação , Ratos , Ratos Wistar , Transdução de Sinais
14.
Invest Ophthalmol Vis Sci ; 48(1): 355-60, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17197554

RESUMO

PURPOSE: To study topographical differences in porcine retinal pigment epithelial (RPE) cell proliferation (1) in vivo, after experimental central surgical subretinal injury, and (2) in vitro. METHODS: Domestic pigs underwent either experimental RPE debridement (n = 5), subretinal amniotic membrane transplantation (n= 4), or both (n= 1) in the left eye. RPE cell proliferation was assayed by injection of the thymidine analogue 5-bromodeoxyuridine (5-BrdU) at postoperative day 0 and 1. RPE cells in S-phase were identified by their incorporation of 5-BrdU, as detected by immunohistochemistry. The in vitro proliferation of primary RPE isolates from the peripheral and central retina was assayed by a colorimetric assay and by [(3)H]thymidine incorporation. RESULTS: After subretinal surgery, in vivo incorporation of 5-BrdU was seen in peripheral RPE cells in 8 of 10 surgically treated eyes, but never in central RPE cells. This observation was true of both types of experimental surgery performed. In vitro, RPE isolates from the pre-equatorial region consistently yielded higher cell densities than did RPE cell isolates from more central parts of the epithelium. This was apparent through the three first passages of porcine RPE cells in culture. After 1 and 4 days in culture, pre-equatorial RPE cells had incorporated significantly more [(3)H]thymidine than had the more central RPE cells. CONCLUSIONS: Experimental subretinal surgical injury of the RPE below the central retina is followed within 48 hours by a peripheral, but not a central, proliferation of RPE cells. In vitro, peripheral RPE cells have a higher proliferative capacity than do central RPE cells.


Assuntos
Proliferação de Células , Ferimentos Oculares Penetrantes/complicações , Epitélio Pigmentado Ocular/patologia , Retina/lesões , Âmnio/transplante , Animais , Bromodesoxiuridina , Contagem de Células , Células Cultivadas , Replicação do DNA , Desbridamento , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA