Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1867(10): 166199, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34144091

RESUMO

During diabetes development insulin production and glucose-stimulated insulin secretion (GSIS) are defective due to inflammation-related, yet not fully understood mechanisms. MCPIP1 (monocyte chemotactic protein-induced protein-1) is a strong regulator of inflammation, and acts predominantly as a specific RNase. The impact of MCPIP1 on insulin secretory capacity is unknown. We show that the expression of the ZC3H12A gene, which encodes MCPIP1, was induced by T1DM- and by T2DM-simulating conditions, with a stronger effect of cytokines. The number of MCPIP1-positive pancreatic islet-cells, including beta-cells, was significantly higher in diabetic compared to nondiabetic individuals. In the 3'UTR regions of mRNAs coding for Pdx1 (pancreatic and duodenal homeobox 1), FoxO1 (forkhead box protein O1), and of a novel regulator of insulin handling, Grp94 (glucose-regulated protein 94), MCPIP1-target structures were detected. Overexpression of the wild type MCPIP1wt, but not of the mutant MCPIP1D141N (lacking the RNase activity), decreased the expression of genes involved in insulin production and GSIS. Additionally INS1-E-MCPIP1wt cells exhibited a higher Ire1 (inositol-requiring enzyme 1) expression. MCPIP1wt overexpression blunted GSIS and glucose-mediated calcium influx with no deleterious effects on glucose uptake or glucokinase activity. We identify MCPIP1 as a new common link between diabetogenic conditions and beta-cell failure. MCPIP1 may serve as an interesting target for novel beta-cell protective approaches.


Assuntos
Diabetes Mellitus/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ribonucleases/metabolismo , Fatores de Transcrição/metabolismo , Regiões 3' não Traduzidas/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular , Citocinas/metabolismo , Diabetes Mellitus/patologia , Proteína Forkhead Box O1/metabolismo , Glucose/metabolismo , Humanos , Células Secretoras de Insulina/patologia , RNA Mensageiro/metabolismo , Ratos
2.
Cell Death Dis ; 10(1): 29, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30631045

RESUMO

The autoimmune-mediated beta-cell death in type 1 diabetes (T1DM) is associated with local inflammation (insulitis). We examined the role of MCPIP1 (monocyte chemotactic protein-induced protein 1), a novel cytokine-induced antiinflammatory protein, in this process. Basal MCPIP1 expression was lower in rat vs. human islets and beta-cells. Proinflammatory cytokines stimulated MCPIP1 expression in rat and human islets and in insulin-secreting cells. Moderate overexpression of MCPIP1 protected insulin-secreting INS1E cells against cytokine toxicity by a mechanism dependent on the presence of the PIN/DUB domain in MCPIP1. It also reduced cytokine-induced Chop and C/ebpß expression and maintained MCL-1 expression. The shRNA-mediated suppression of MCPIP1 led to the potentiation of cytokine-mediated NFκB activation and cytokine toxicity in human EndoC-ßH1 beta-cells. MCPIP1 expression was very high in infiltrated beta-cells before and after diabetes manifestation in the LEW.1AR1-iddm rat model of human T1DM. The extremely high expression of MCPIP1 in clonal beta-cells was associated with a failure of the regulatory feedback-loop mechanism, ER stress induction and high cytokine toxicity. In conclusion, our data indicate that the expression level of MCPIP1 affects the susceptibility of insulin-secreting cells to cytokines and regulates the mechanism of beta-cell death in T1DM.


Assuntos
Citocinas/toxicidade , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Ribonucleases/genética , Ribonucleases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Expressão Gênica , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Estresse Nitrosativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos Lew , Transfecção
3.
J Biol Chem ; 292(49): 20292-20304, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29070677

RESUMO

Increasing evidence suggests a crucial role of inflammation in cytokine-mediated ß-cell dysfunction and death in type 1 diabetes mellitus, although the mechanisms are incompletely understood. Sphingosine 1-phosphate (S1P) is a multifunctional bioactive sphingolipid involved in the development of many autoimmune and inflammatory diseases. Here, we investigated the role of intracellular S1P in insulin-secreting INS1E cells by genetically manipulating the S1P-metabolizing enzyme S1P lyase (SPL). The expression of spl was down-regulated by cytokines in INS1E cells and rat islets. Overexpression of SPL protected against cytokine toxicity. Interestingly, the SPL overexpression did not suppress the cytokine-induced NFκB-iNOS-NO pathway but attenuated calcium leakage from endoplasmic reticulum (ER) stores as manifested by lower cytosolic calcium levels, higher expression of the ER protein Sec61a, decreased dephosphorylation of Bcl-2-associated death promoter (Bad) protein, and weaker caspase-3 activation in cytokine-treated (IL-1ß, TNFα, and IFNγ) cells. This coincided with reduced cytokine-mediated ER stress, indicated by measurements of CCAAT/enhancer-binding protein homologous protein (chop) and immunoglobulin heavy chain binding protein (bip) levels. Moreover, cytokine-treated SPL-overexpressing cells exhibited increased expression of prohibitin 2 (Phb2), involved in the regulation of mitochondrial assembly and respiration. SPL-overexpressing cells were partially protected against cytokine-mediated ATP reduction and inhibition of glucose-induced insulin secretion. siRNA-mediated spl suppression resulted in effects opposite to those observed for SPL overexpression. Knockdown of phb2 partially reversed beneficial effects of SPL overexpression. In conclusion, the relatively low endogenous Spl expression level in insulin-secreting cells contributes to their extraordinary vulnerability to proinflammatory cytokine toxicity and may therefore represent a promising target for ß-cell protection in type 1 diabetes mellitus.


Assuntos
Aldeído Liases/genética , Aldeído Liases/fisiologia , Citocinas/toxicidade , Células Secretoras de Insulina/enzimologia , Trifosfato de Adenosina/metabolismo , Aldeído Liases/biossíntese , Animais , Linhagem Celular , Citocinas/farmacologia , Diabetes Mellitus Tipo 1/patologia , Estresse do Retículo Endoplasmático , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/enzimologia , Ratos
4.
Biochem Biophys Res Commun ; 464(1): 13-9, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26028562

RESUMO

In the new human EndoC-ßH1 ß-cell line, a detailed analysis of the physiological characteristics was performed. This new human ß-cell line expressed all target structures on the gene and protein level, which are crucial for physiological function and insulin secretion induced by glucose and other secretagogues. Glucose influx measurements revealed an excellent uptake capacity of EndoC-ßH1 ß-cells by the Glut1 and Glut2 glucose transporters. A high expression level of glucokinase enabled efficient glucose phosphorylation, increasing the ATP/ADP ratio along with stimulation of insulin secretion in the physiological glucose concentration range. The EC50 value of glucose for insulin secretion was 10.3 mM. Mannoheptulose, a specific glucokinase inhibitor, blocked glucose-induced insulin secretion (GSIS). The nutrient insulin secretagogues l-leucine and 2-ketoisocaproate also stimulated insulin secretion, with a potentiating effect of l-glutamine. The Kir 6.2 potassium channel blocker glibenclamide and Bay K 8644, an opener of the voltage-sensitive Ca(2+) channel significantly potentiated GSIS. Potentiation of GSIS by IBMX and forskolin went along with a strong stimulation of cAMP generation. In conclusion, the new human EndoC-ßH1 ß-cell line fully mirrors the analogous physiological characteristics of primary mouse, rat and human ß-cells. Thus, this new human EndoC-ßH1 ß-cell line is very well suited for physiological ß-cell studies.


Assuntos
Efeito Fundador , Glucose/metabolismo , Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/biossíntese , Transporte Biológico , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Linhagem Celular , Colforsina/farmacologia , Expressão Gênica , Glucoquinase/antagonistas & inibidores , Glucoquinase/genética , Glucoquinase/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 2/genética , Transportador de Glucose Tipo 2/metabolismo , Glutamina/metabolismo , Glutamina/farmacologia , Glibureto/farmacologia , Humanos , Células Secretoras de Insulina/citologia , Cetoácidos/metabolismo , Cetoácidos/farmacologia , Leucina/metabolismo , Leucina/farmacologia , Manoeptulose/metabolismo , Manoeptulose/farmacologia , Fosforilação , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
5.
Nitric Oxide ; 27(4): 235-41, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22921991

RESUMO

Nitric oxide (NO), produced by the action of the inducible NO synthase, plays a crucial role in cytokine toxicity to pancreatic beta cells during type 1 diabetes development. It was the aim of this study to analyze the role of the neuronal NOS (nNOS) in proinflammatory cytokine-mediated beta cell toxicity. Expression of different isoforms of nitric oxide synthase in insulin-secreting INS1E cells and rat islets was analyzed by quantitative real-time PCR and Western blotting. The expression of nNOS in insulin-secreting INS1E cells was similar to that found in rat brain, while two other isoforms, namely the endothelial eNOS and inducible iNOS were not expressed in untreated cells. IL-1ß alone or in combination with TNF-α and/or IFNγ induced iNOS but not eNOS expression. In contrast, nNOS expression was strongly decreased by the mixture of the three proinflammatory cytokines (IL-1ß, TNF-α and IFNγ) both on the gene and protein level in INS1E cells and rat islet cells. The effects of cytokines on glucose-induced insulin-secretion followed the pattern of nNOS expression reduction and, on the other hand, of the iNOS induction. The data indicate that a low level of nitric oxide originating from the constitutive expression of nNOS in pancreatic beta cells is not deleterious. In particular since proinflammatory cytokines reduce this expression. This nNOS suppression can compensate for NO generation by low concentrations of IL-1ß through iNOS induction. Thus, this basal nNOS expression level in pancreatic beta cells represents a protective element against cytokine toxicity.


Assuntos
Citocinas/toxicidade , Células Secretoras de Insulina/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Animais , Sequência de Bases , Citocinas/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Dados de Sequência Molecular , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
6.
Biochem J ; 445(3): 349-59, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22587331

RESUMO

Mimitin, a novel mitochondrial protein, has been shown to act as a molecular chaperone for the mitochondrial complex I and to regulate ATP synthesis. During Type 1 diabetes development, pro-inflammatory cytokines induce mitochondrial damage in pancreatic ß-cells, inhibit ATP synthesis and reduce glucose-induced insulin secretion. Mimitin was expressed in rat pancreatic islets including ß-cells and decreased by cytokines. In the ob/ob mouse, a model of insulin resistance and obesity, mimitin expression was down-regulated in liver and brain, up-regulated in heart and kidney, but not affected in islets. To further analyse the impact of mimitin on ß-cell function, two ß-cell lines, one with a low (INS1E) and another with a higher (MIN6) mimitin expression were studied. Mimitin overexpression protected INS1E cells against cytokine-induced caspase 3 activation, mitochondrial membrane potential reduction and ATP production inhibition, independently from the NF-κB (nuclear factor κB)-iNOS (inducible NO synthase) pathway. Mimitin overexpression increased basal and glucose-induced insulin secretion and prevented cytokine-mediated suppression of insulin secretion. Mimitin knockdown in MIN6 cells had opposite effects to those observed after overexpression. Thus mimitin has the capacity to modulate pancreatic islet function and to reduce cytokine toxicity.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Células Secretoras de Insulina/fisiologia , Proteínas Mitocondriais/metabolismo , Chaperonas Moleculares/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sequência de Bases , Caspases/metabolismo , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Citocinas/farmacologia , Primers do DNA/genética , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Complexo I de Transporte de Elétrons/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Técnicas In Vitro , Insulina/genética , Insulina/metabolismo , Resistência à Insulina/fisiologia , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Obesos , Proteínas Mitocondriais/antagonistas & inibidores , Proteínas Mitocondriais/genética , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/genética , Obesidade/fisiopatologia , Ratos , Ratos Endogâmicos Lew
7.
Endocrinology ; 153(6): 2612-22, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22495672

RESUMO

Arachidonic acid metabolites are crucial mediators of inflammation in diabetes. Although eicosanoids are established modulators of pancreatic ß-cell function, the role of prostacyclin (prostaglandin I2) is unknown. Therefore, this study aimed to analyze the role of prostacyclin in ß-cell function. Prostacyclin synthase (PGIS) was weakly expressed in rat islet cells but nevertheless significantly increased by incubation with 30 mM glucose, especially in non-ß-cells. PGIS was overexpressed in INS1E cells, and the regulation of insulin secretion was analyzed. PGIS overexpression strongly potentiated glucose-induced insulin secretion along with increased insulin content and ATP production. Importantly, overexpression of PGIS potentiated only nutrient-induced insulin secretion. The effect of PGIS overexpression was mediated by prostacyclin released from insulin-secreting cells and dependent on prostacyclin receptor (IP receptor) activation, with concomitant cAMP production. The cAMP-mediated potentiation of glucose-induced insulin secretion by prostacyclin was independent of the protein kinase A pathway but strongly attenuated by the knockdown of the exchange protein directly activated by cAMP 2 (Epac2), pointing to a crucial role for Epac2 in this process. Thus, prostacyclin is a powerful potentiator of glucose-induced insulin secretion. It improves the secretory capacity by inducing insulin biosynthesis and probably by stimulating exocytosis. Our findings open a new therapeutical perspective for an improved treatment of type 2 diabetes.


Assuntos
Epoprostenol/farmacologia , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Western Blotting , Células Cultivadas , AMP Cíclico/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Sinergismo Farmacológico , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Ilhotas Pancreáticas/metabolismo , Masculino , Interferência de RNA , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
J Endocrinol ; 197(1): 139-50, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18372240

RESUMO

Pro-inflammatory cytokines cause beta-cell dysfunction and death. The aim of this study was to investigate the interactions between different pro- and anti-inflammatory cytokines and their effects on apoptotic beta-cell death pathways. Insulin-producing RINm5F cells were exposed to different combinations of cytokines. Gene expression analyses of manganese superoxide dismutase (MnSOD) and inducible nitric oxide synthase (iNOS) were performed by real-time RT-PCR. Cell viability was measured by the MTT assay, NFkappaB activation using a SEAP reporter gene assay, protein expression by western blotting and caspase-3 activity using the DEVD cleavage method. IL-1beta, tumour necrosis factor alpha (TNFalpha) and a combination of all three pro-inflammatory cytokines increased while IFNgamma alone did not affect NFkappaB activity and iNOS gene and protein expression. Interestingly, the anti-inflammatory cytokines IL-4, IL-13 and IL-10 decreased IL-1beta-stimulated NFkappaB activation and iNOS expression. IL-1beta, TNFalpha and the pro-inflammatory cytokine combination also increased MnSOD gene and protein expression. But IL-4, IL-13 and IL-10 did not affect MnSOD expression and did not modulate IL-1beta-stimulated MnSOD expression. Caspase-3 activity was increased by IL-1beta and the pro-inflammatory cytokine combination, and to a lesser extent by TNFalpha. In contrast, IFNgamma had no effect on caspase-3 activity. IL-4, IL-13 and IL-10 decreased caspase-3 activity and increased viability of insulin-producing cells treated with pro-inflammatory cytokines. The anti-inflammatory cytokines counteracted the cytotoxic effects of pro-inflammatory cytokines in insulin-producing cells. This was achieved through the reduction of nitrosative stress. Thus, a balance between the anti-inflammatory and the pro-inflammatory cytokines is of crucial importance for the prevention of pancreatic beta-cell destruction.


Assuntos
Citocinas/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Secretoras de Insulina/metabolismo , Interferon gama/farmacologia , Interleucina-1beta/farmacologia , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Nitroarginina/farmacologia , Ratos , Superóxido Dismutase/genética
9.
Antioxid Redox Signal ; 10(4): 691-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18177230

RESUMO

The diabetogenic agent alloxan is a triketone that selectively destroys pancreatic beta cells. To investigate the importance of the triketone structure of alloxan for its cytotoxic potency, alloxan was compared with ninhydrin, also a triketone, and the amino derivative of alloxan uramil, which is not a triketone, because the 5-keto group of the alloxan is replaced by an amino group. Both compounds are cytotoxic but not diabetogenic. Ninhydrin was capable of generating cytotoxic reactive oxygen species (ROS) through redox cycling with dithiols, and uramil could also generate cytotoxic ROS. Both ninhydrin and uramil could not redox cycle with glutathione (GSH) and were not selectively toxic to beta cells; their structure does not allow selective cellular uptake via the GLUT2 glucose transporter. Thus, the results show that the 5-keto group in the pyrimidine ring structure of the triketone alloxan is crucially important for its ability to be selectively taken up into the beta cells via the specific glucose transporter GLUT2. The 5-keto group of the molecule enables redox cycling of alloxan through reaction with glutathione (GSH), thereby generating the cytotoxic ROS. Thus, the unique combination of these two properties confers on alloxan the beta cell-selective toxicity and diabetogenicity. Replacement of the 5-keto group by an amino group, as in uramil, abolishes selective beta cell toxicity because of the loss of the glucose analogue structure and the capability to generate ROS via redox cycling with GSH and cysteine.


Assuntos
Aloxano/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Ninidrina/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Aloxano/química , Animais , Células Cultivadas , Transportador de Glucose Tipo 2/metabolismo , Glutationa/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Estrutura Molecular , Ninidrina/química , Oxigênio/metabolismo , Fosforilação , Ratos , Relação Estrutura-Atividade , Tolueno/análogos & derivados , Tolueno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA