Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Endocrinol ; 72(2)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38054640

RESUMO

The early phase of type 2 diabetes mellitus (T2DM) is characterised by insulin resistance, which can initially be compensated by elevated insulin secretion. However, as postulated by the workload hypothesis, over time harming insulin requirements contribute to ß-cell dysfunction and death. The mechanisms behind this transition are complex and not fully understood but involve factors such as endoplasmic reticulum (ER) stress raised by gluco/lipotoxicity. To investigate the effect of excessive insulin folding on ER luminal H2O2 generation, ER stress and viability, insulin was expressed glucose-independently by a doxycycline-regulated Tet-On system in insulin-producing RINm5F cells. Additionally, the effect of palmitic acid (PA) as a subsidiary T2DM-associated factor was examined in this model system. Elevated insulin expression increased ER luminal H2O2 concentration quantified by the fluorescent sensor protein TriPer and reduced viability, but did not activate apoptosis. However, when combined with PA, insulin expression resulted in a significant increase in ER stress and apoptosis. Expression of ER-localised catalase verified the specificity of the applied H2O2 detection method without attenuating ER stress, caspase activation or viability loss. These findings suggest that hyperinsulinism alone can cause increased ER luminal H2O2 generation, mild ER stress and reduced viability, while hyperinsulinism in combination with PA accelerates these processes and triggers apoptosis. The inability of ER catalase to counteract these effects suggests that further damaging factors besides H2O2 are involved in cell dysfunction. Finally, reducing the high insulin demand in the initial phase of T2DM may be crucial in preventing further ß-cell damage caused by gluco/lipotoxicity.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Células Secretoras de Insulina , Humanos , Ácido Palmítico/farmacologia , Catalase/metabolismo , Catalase/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Peróxido de Hidrogênio/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Apoptose , Estresse Oxidativo , Estresse do Retículo Endoplasmático
2.
FASEB J ; 37(1): e22685, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36468845

RESUMO

The endoplasmic reticulum (ER) lumen is not only the major site for the assembly and folding of newly synthesized proteins but also the main intracellular Ca2+ store. Ca2+ ions are involved in versatile biochemical processes, including posttranslational processing and folding of nascent proteins. Disruption of ER Ca2+ homeostasis is usually accompanied by an ER stress response that can ultimately lead to apoptosis if unresolved. Abnormal ER Ca2+ depletion has been linked to pancreatic ß-cell dysfunction and death under lipotoxic conditions. However, the underlying mechanisms how the ß-cell toxic saturated free fatty acid palmitate perturbs ER Ca2+ homeostasis and its interplay with other organelles are not fully understood. In the present study, we demonstrate that treatment of insulin-secreting INS-1E cells with palmitate diminished ER Ca2+ levels, elevated cytosolic/mitochondrial Ca2+ content, lowered the mitochondrial membrane potential, and ATP content. In addition, palmitate-pretreated ß-cells contained significantly less luminal Ca2+ , revealed a severely impaired ER Ca2+ reuptake rate, and substantially lower insulin content. Importantly, detoxification of luminal H2 O2 by expression of the ER-resident glutathione peroxidase 8 (GPx8) abrogated the lipotoxic effects of palmitate. Moreover, GPx8 supported oxidative protein folding and preserved insulin content under lipotoxic conditions. A direct involvement of luminal H2 O2 in palmitate-mediated ER Ca2+ depletion could be corroborated by the ectopic expression of an ER-luminal active catalase. Our data point to the critical role of luminal H2 O2 in palmitate-mediated depletion of ER Ca2+ through redox-dependent impairment of Ca2+ ATPase pump activity upstream of mitochondrial dysfunction in insulin-secreting INS-1E cells.


Assuntos
Células Secretoras de Insulina , Palmitatos , Palmitatos/metabolismo , Células Secretoras de Insulina/metabolismo , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Insulina/metabolismo
3.
Cells ; 10(9)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34571827

RESUMO

During type 1 diabetes mellitus (T1DM) development, beta-cells undergo intense endoplasmic reticulum (ER) stress that could result in apoptosis through the failure of adaptation to the unfolded protein response (UPR). Islet transplantation is considered an attractive alternative among beta-cell replacement therapies for T1DM. To avoid the loss of beta-cells that will jeopardize the transplant's outcome, several strategies are being studied. We have previously shown that prolactin induces protection against proinflammatory cytokines and redox imbalance-induced beta-cell death by increasing heat-shock protein B1 (HSPB1) levels. Since the role of HSPB1 in beta cells has not been deeply studied, we investigated the mechanisms involved in unbalanced protein homeostasis caused by intense ER stress and overload of the proteasomal protein degradation pathway. We tested whether HSPB1-mediated cytoprotective effects involved UPR modulation and improvement of protein degradation via the ubiquitin-proteasome system. We demonstrated that increased levels of HSPB1 attenuated levels of pro-apoptotic proteins such as CHOP and BIM, as well as increased protein ubiquitination and the speed of proteasomal protein degradation. Our data showed that HSPB1 induced resistance to proteotoxic stress and, thus, enhanced cell survival via an increase in beta-cell proteolytic capacity. These results could contribute to generate strategies aimed at the optimization of beta-cell replacement therapies.


Assuntos
Proteínas de Choque Térmico/metabolismo , Células Secretoras de Insulina/metabolismo , Chaperonas Moleculares/metabolismo , Animais , Apoptose/fisiologia , Morte Celular/fisiologia , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Oxirredução , Complexo de Endopeptidases do Proteassoma/metabolismo , Transporte Proteico/fisiologia , Proteólise , Resposta a Proteínas não Dobradas/fisiologia
4.
Biochim Biophys Acta Biomembr ; 1862(2): 183096, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31672544

RESUMO

Hydrogen peroxide (H2O2) plays a central role in redox signalling and in oxidative stress-mediated cell death. It is generated through multiple mechanisms at various intracellular sites. Due to its chemical stability it can reach distant sites of action. However, its hydrophilicity can hamper lipid membrane passage. We therefore studied the kinetics of H2O2 diffusion through subcellular membranes employing the H2O2 biosensor HyPer in insulin-producing RINm5F cells. Plasma- and ER-membrane-bound HyPer sensors facing the cytosolic compartment reacted twice as fast to H2O2 compared to sensors expressed in peroxisomes and mitochondria. Overexpression of the H2O2-inactivating enzyme catalase in the ER-lumen and in the peroxisomes retarded the reaction time of HyPer, both localised within the peroxisomes as well as at the cytosolic surface of the ER. The unsaturated fatty acid oleic acid did not affect the reaction of the peroxisomal HyPer sensor to H2O2, while the saturated fatty acid palmitic acid accelerated its reaction time to H2O2 in this organelle. The results show that the plasma-, peroxisomal, and mitochondrial membrane of insulin-producing RINm5F cells are permeable for H2O2. Nonetheless, the organelle membranes retard H2O2 diffusion due to a barrier function of the lipid membrane, as documented by retarded reaction times of the intraorganellar sensors. Accelerated decomposition of H2O2 by catalase, expressed in the peroxisomes or the ER, further retarded the HyPer sensor reaction time. The results show that redox signalling and oxidative stress-mediated toxicity are crucially dependent on physicochemical membrane properties and antioxidative defence mechanisms in health and disease.


Assuntos
Membrana Celular/metabolismo , Peróxido de Hidrogênio/metabolismo , Células Secretoras de Insulina/ultraestrutura , Técnicas Biossensoriais , Difusão , Humanos , Células Secretoras de Insulina/metabolismo , Cinética , Oxirredução , Estresse Oxidativo , Permeabilidade
5.
Free Radic Biol Med ; 134: 394-405, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30699366

RESUMO

Maintaining islet cell viability in vitro, although challenging, appears to be a strategy for improving the outcome of pancreatic islet transplantation. We have shown that prolactin (PRL) leads to beta-cell cytoprotection against apoptosis, an effect mediated by heat shock protein B1 (HSPB1). Since the role of HSPB1 in beta-cells is still unclear and the hormone concentration used is not compatible with clinical applications because of all the side effects displayed by the hormone in other tissues, we explored the molecular mechanisms by which HSPB1 mediates beta-cell cytoprotection. Lysates from PRL- and/or cytokine-treated MIN6 beta-cells were subjected to HSPB1 immunoprecipitation followed by identification through mass spectrometry. PRL-treated cells presented an enrichment of several proteins co-precipitating with HSPB1. Of note were oxidative stress resistance-, protein degradation- and carbohydrate metabolism-related proteins. Wild type, HSPB1 silenced or overexpressing MIN6 cells were exposed to menadione and hydrogen peroxide and analysed for several oxidative stress parameters. HSPB1 knockdown rendered cells more sensitive to oxidative stress and led to a reduced antioxidant capacity, while prolactin induced an HSPB1-mediated cytoprotection against oxidative stress. HSPB1 overexpression, however, led to opposite effects. PRL treatment, HSPB1 silencing or overexpression did not change the expression nor activities of antioxidant enzymes, it also did not lead to a modulation of total glutathione levels nor G6PD expression. However, HSPB1 levels are related to a modulation of GSH/GSSG ratio, G6PD activity and NADPH/NADP + ratio. We have shown that HSPB1 is important for pro-survival effects against oxidative stress-induced beta-cell death. These results are in accordance with PRL-induced enrichment of HSPB1-interacting proteins related to protection against oxidative stress. Finally, our results outline the need of further studies investigating the importance of HSPB1 for beta-cell viability, since this could lead to the mitigation of beta-cell death through the up-regulation of an endogenous protective pathway.


Assuntos
Citoproteção , Proteínas de Choque Térmico/metabolismo , Células Secretoras de Insulina/citologia , Insulinoma/patologia , Chaperonas Moleculares/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Prolactina/farmacologia , Animais , Apoptose , Glutationa/metabolismo , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Insulinoma/tratamento farmacológico , Insulinoma/metabolismo , Camundongos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/genética , Oxirredução , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Transporte Proteico , Proteólise , Células Tumorais Cultivadas
6.
Free Radic Biol Med ; 112: 121-130, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28751022

RESUMO

Increased circulating levels of saturated fatty acids (FFAs) and glucose are considered to be major mediators of ß-cell dysfunction and death in T2DM. Although it has been proposed that endoplasmic reticulum (ER) and oxidative stress play a crucial role in gluco/lipotoxicity, their interplay and relative contribution to ß-cell dysfunction and apoptosis has not been fully elucidated. In addition it is still unclear how palmitate - the physiologically most abundant long-chain saturated FFA - elicits ER stress and which immediate signals commit ß-cells to apoptosis. To study the underlying mechanisms of palmitate-mediated ER stress and ß-cell toxicity, we exploited the observation that the recently described ER-resident GPx7 and GPx8 are not expressed in rat ß-cells. Expression of GPx7 or GPx8 attenuated FFAs-mediated H2O2 generation, ER stress, and apoptosis induction. These results could be confirmed by a H2O2-specific inactivating ER catalase, indicating that accumulation of H2O2 in the ER lumen is critical in FFA-induced ER stress. Furthermore, neither the expression of GPx7 nor of GPx8 increased insulin content or facilitated disulfide bond formation in insulin-secreting INS-1E cells. Hence, reduction of H2O2 by ER-GPx isoforms is not rate-limiting in oxidative protein folding in rat ß-cells. These data suggest that FFA-mediated ER stress is partially dependent on oxidative stress and selective expression of GPx7 or GPx8 improves the ER antioxidative capacity of rat ß-cells without compromising insulin production and the oxidative protein folding machinery.


Assuntos
Retículo Endoplasmático/efeitos dos fármacos , Glucose/toxicidade , Ácido Palmítico/toxicidade , Peroxidases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Expressão Gênica , Glutationa Peroxidase , Peróxido de Hidrogênio/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Estresse Oxidativo , Peroxidases/genética , Plasmídeos/química , Plasmídeos/metabolismo , Ratos , Transfecção , Transgenes
7.
BMC Biol ; 15(1): 24, 2017 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-28347335

RESUMO

BACKGROUND: The fate of hydrogen peroxide (H2O2) in the endoplasmic reticulum (ER) has been inferred indirectly from the activity of ER-localized thiol oxidases and peroxiredoxins, in vitro, and the consequences of their genetic manipulation, in vivo. Over the years hints have suggested that glutathione, puzzlingly abundant in the ER lumen, might have a role in reducing the heavy burden of H2O2 produced by the luminal enzymatic machinery for disulfide bond formation. However, limitations in existing organelle-targeted H2O2 probes have rendered them inert in the thiol-oxidizing ER, precluding experimental follow-up of glutathione's role in ER H2O2 metabolism. RESULTS: Here we report on the development of TriPer, a vital optical probe sensitive to changes in the concentration of H2O2 in the thiol-oxidizing environment of the ER. Consistent with the hypothesized contribution of oxidative protein folding to H2O2 production, ER-localized TriPer detected an increase in the luminal H2O2 signal upon induction of pro-insulin (a disulfide-bonded protein of pancreatic ß-cells), which was attenuated by the ectopic expression of catalase in the ER lumen. Interfering with glutathione production in the cytosol by buthionine sulfoximine (BSO) or enhancing its localized destruction by expression of the glutathione-degrading enzyme ChaC1 in the lumen of the ER further enhanced the luminal H2O2 signal and eroded ß-cell viability. CONCLUSIONS: A tri-cysteine system with a single peroxidatic thiol enables H2O2 detection in oxidizing milieux such as that of the ER. Tracking ER H2O2 in live pancreatic ß-cells points to a role for glutathione in H2O2 turnover.


Assuntos
Retículo Endoplasmático/metabolismo , Peróxido de Hidrogênio/metabolismo , Sondas Moleculares/metabolismo , Fenômenos Ópticos , Animais , Catálise , Linhagem Celular , Retículo Endoplasmático/efeitos dos fármacos , Fluorescência , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Cinética , Camundongos , Oxirredução , Compostos de Sulfidrila/metabolismo
8.
J Biol Chem ; 289(39): 26904-26913, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25122762

RESUMO

Oxidative folding of (pro)insulin is crucial for its assembly and biological function. This process takes place in the endoplasmic reticulum (ER) and is accomplished by protein disulfide isomerase and ER oxidoreductin 1ß, generating stoichiometric amounts of hydrogen peroxide (H2O2) as byproduct. During insulin resistance in the prediabetic state, increased insulin biosynthesis can overwhelm the ER antioxidative and folding capacity, causing an imbalance in the ER redox homeostasis and oxidative stress. Peroxiredoxin 4 (Prdx4), an ER-specific antioxidative peroxidase can utilize luminal H2O2 as driving force for reoxidizing protein disulfide isomerase family members, thus efficiently contributing to disulfide bond formation. Here, we examined the functional significance of Prdx4 on ß-cell function with emphasis on insulin content and secretion during stimulation with nutrient secretagogues. Overexpression of Prdx4 in glucose-responsive insulin-secreting INS-1E cells significantly metabolized luminal H2O2 and improved the glucose-induced insulin secretion, which was accompanied by the enhanced proinsulin mRNA transcription and insulin content. This ß-cell beneficial effect was also observed upon stimulation with the nutrient insulin secretagogue combination of leucine plus glutamine, indicating that the effect is not restricted to glucose. However, knockdown of Prdx4 had no impact on H2O2 metabolism or ß-cell function due to the fact that Prdx4 expression is negligibly low in pancreatic ß-cells. Moreover, we provide evidence that the constitutively low expression of Prdx4 is highly susceptible to hyperoxidation in the presence of high glucose. Overall, these data suggest an important role of Prdx4 in maintaining insulin levels and improving the ER folding capacity also under conditions of a high insulin requirement.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Peroxirredoxinas/biossíntese , Edulcorantes/farmacologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Células Hep G2 , Humanos , Peróxido de Hidrogênio/metabolismo , Insulina/genética , Secreção de Insulina , Células Secretoras de Insulina/citologia , Oxirredução/efeitos dos fármacos , Peroxirredoxinas/genética , Dobramento de Proteína/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Edulcorantes/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
9.
Free Radic Biol Med ; 53(7): 1451-8, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22921589

RESUMO

Oxidative protein folding in the endoplasmic reticulum (ER) is associated with the formation of native disulfide bonds, which inevitably results in the formation of hydrogen peroxide (H(2)O(2)). Particularly in pancreatic ß-cells with their high secretory activity and extremely low antioxidant capacity, the H(2)O(2) molecules generated during oxidative protein folding could represent a significant oxidative burden. Therefore this study was conducted to elucidate the H(2)O(2) generation during disulfide bond formation in insulin-producing RINm5F cells by targeting and specifically expressing the H(2)O(2)-sensitive biosensor HyPer in the ER (ER-HyPer). In addition the influence of overexpression of the H(2)O(2)-metabolizing ER-resident peroxiredoxin IV (PRDXIV) on H(2)O(2) levels was examined. The ER-HyPer fluorescent protein was completely oxidized, whereas HyPer expressed in cytosol, peroxisomes, and mitochondria was prevalently in the reduced state, indicating a high basal H(2)O(2) concentration in the ER lumen. These results could also be confirmed in non-insulin-producing COS-7 cells. Overexpression of PRDXIV in RINm5F cells effectively protected against H(2)O(2)-mediated toxicity; however, it did not affect the fluorescence signal of ER-HyPer. Moreover, the inhibition of de novo protein synthesis and the associated H(2)O(2) generation by cycloheximide had no influence on the ER-HyPer redox state. Taken together, these findings strongly suggest that the H(2)O(2)-sensitive biosensor reflects exclusively the oxidative milieu in the ER and not the H(2)O(2) concentration in the ER lumen.


Assuntos
Dissulfetos/metabolismo , Retículo Endoplasmático/metabolismo , Peróxido de Hidrogênio/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Luminescentes/metabolismo , Compostos de Sulfidrila/metabolismo , Animais , Técnicas Biossensoriais , Linhagem Celular , Cicloeximida/farmacologia , Citosol/efeitos dos fármacos , Citosol/metabolismo , Fluorescência , Expressão Gênica , Genes Reporter , Peróxido de Hidrogênio/farmacologia , Células Secretoras de Insulina/citologia , Proteínas Luminescentes/genética , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Sondas Moleculares , Oxirredução , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Peroxissomos/efeitos dos fármacos , Peroxissomos/metabolismo , Dobramento de Proteína , Ratos
10.
Biochim Biophys Acta ; 1813(10): 1827-35, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21784110

RESUMO

Pro-inflammatory cytokine-mediated beta cell apoptosis is activated through multiple signaling pathways involving mitochondria and endoplasmic reticulum. Activation of organelle-specific caspases has been implicated in the progression and execution of cell death. This study was therefore performed to elucidate the effects of pro-inflammatory cytokines on a possible cross-talk between the compartment-specific caspases 9 and 12 and their differential contribution to beta cell apoptosis. Moreover, the occurrence of ROS-mediated mitochondrial damage in response to beta cell toxic cytokines has been quantified. ER-specific caspase-12 was strongly activated in response to pro-inflammatory cytokines; however, its inhibition did not abolish cytokine-induced mitochondrial caspase-9 activation and loss of cell viability. In addition, there was a significant induction of oxidative mitochondrial DNA damage and elevated cardiolipin peroxidation in insulin-producing RINm5F cells and rat islet cells. Overexpression of the H(2)O(2) detoxifying enzyme catalase effectively reduced the observed cytokine-induced oxidative damage of mitochondrial structures. Taken together, the results strongly indicate that mitochondrial caspase-9 is not a downstream substrate of ER-specific caspase-12 and that pro-inflammatory cytokines cause apoptotic beta cell death through activation of caspase-9 primarily by hydroxyl radical-mediated mitochondrial damage.


Assuntos
Apoptose/fisiologia , Caspase 12/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Mitocôndrias/metabolismo , Estresse Oxidativo/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspase 12/genética , Caspase 9/metabolismo , Caspase 9/fisiologia , Catalase/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citocinas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ativação Enzimática/fisiologia , Mediadores da Inflamação/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Ratos , Ratos Endogâmicos Lew
11.
J Mol Med (Berl) ; 89(8): 785-98, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21487676

RESUMO

Although nitric oxide (NO) and oxidative stress both contribute to proinflammatory cytokine toxicity in pancreatic ß-cells during type 1 diabetes mellitus (T1DM) development, the interactions between NO and reactive oxygen species (ROS) in cytokine-mediated ß-cell death have not been clarified. Exposure of insulin-producing RINm5F cells to IL-1ß generated NO, while exposure to a combination of IL-1ß, TNF-α, and IFN-γ, which simulates T1DM conditions, generated both NO and ROS. In theory, two reactions between NO and ROS are possible, one with the superoxide radical yielding peroxynitrite, and the other with hydrogen peroxide (H(2)O(2)) yielding hydroxyl radicals. Results of the present work exclude peroxynitrite involvement in cytokine toxicity to ß-cells because its generation did not correlate with the toxic action of cytokines. On the other hand, we show that H(2)O(2), produced upon exposure of insulin-producing cell clones and primary rat islet cells to cytokines almost exclusively in the mitochondria, reacted in the presence of trace metal (Fe(++)) with NO forming highly toxic hydroxyl radicals, thus explaining the severe toxicity that causes apoptotic ß-cell death. Expression of the H(2)O(2)-inactivating enzyme catalase in mitochondria protected against cytokine toxicity by preventing hydroxyl radical formation. We therefore conclude that proinflammatory cytokine-mediated ß-cell death is due to nitro-oxidative stress-mediated hydroxyl radical formation in the mitochondria.


Assuntos
Citocinas/toxicidade , Radical Hidroxila/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Mitocôndrias/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Caspase 3/metabolismo , Linhagem Celular , Células Clonais , Inibidores Enzimáticos/farmacologia , Fluoresceínas/metabolismo , Peróxido de Hidrogênio/metabolismo , Insulina/biossíntese , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Mitocôndrias/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Nitritos/metabolismo , Oxirredução/efeitos dos fármacos , Ácido Peroxinitroso/biossíntese , Ratos , Ratos Endogâmicos Lew , Tirosina/análogos & derivados , Tirosina/metabolismo
12.
Mol Cell Endocrinol ; 332(1-2): 88-96, 2011 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-20933054

RESUMO

Pro-inflammatory cytokines are key mediators in the selective and progressive destruction of insulin-producing beta cells during type 1 diabetes development. However, the mechanisms of cytokine-induced beta cell apoptosis are not fully understood. This study demonstrates that pro-inflammatory cytokines strongly modified the expression of the anti-apoptotic protein Bcl-2 and the pro-apoptotic BH3-only proteins Bad, Bim, and Bid in primary rat islets and insulin-producing RINm5F cells. Overexpression of mitochondrially located catalase (MitoCatalase) specifically increased basal Bcl-2 and decreased basal Bax expression, suppressed cytokine-mediated reduction of Bcl-2, and thereby prevented the release of cytochrome c, Smac/DIABLO and the activation of caspase-9 and -3. Thus, cytokine-mediated decrease of Bcl-2 expression and the sequentially changed Bax/Bcl-2 ratio are responsible for the release of pro-apoptotic mitochondrial factors, activation of caspase-9, and ultimately caspase-3. These results indicate that activation of the intrinsic/mitochondrial apoptosis pathway is essential for cytokine-induced beta cell death and the mitochondrial generation of reactive oxygen species, in particular mitochondrial hydrogen peroxide, differentially regulates the Bax/Bcl-2 ratio.


Assuntos
Antioxidantes/metabolismo , Citocinas/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Proteína 11 Semelhante a Bcl-2 , Caspase 3/metabolismo , Caspase 9/metabolismo , Catalase/genética , Catalase/metabolismo , Linhagem Celular , Células Secretoras de Insulina/citologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Ratos , Ratos Endogâmicos Lew , Espécies Reativas de Oxigênio/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
13.
Life Sci ; 80(22): 2045-50, 2007 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-17408701

RESUMO

Thyroid hormones reduce glucose tolerance in humans and animals. This effect is related to a decrease of glucose-induced insulin secretion following a reduction of pancreatic beta cell mass due to beta cell loss. The aim of this study was to analyze in vitro the mechanisms underlying the effects of triiodothyronine (T(3)) on the cell viability and cell cycle caused by changes of cell death or proliferation rate of insulin-producing INS-1 cells. 72-h Exposure of INS-1 cells to increasing T(3) concentrations up to 500 microM resulted in a significant viability reduction. This T(3) toxicity was caused by an increased apoptotic cell death rate, which was accompanied by a decreased proliferation rate. Inhibitory effects of T(3) on glucose-induced insulin secretion were already seen after 24 h of incubation, indicating that the deleterious effects of T(3) were time-dependent, changing from specific cellular dysfunctions to a severe and extended disturbance of the cellular survival program. Only T(3) concentrations higher than 250 microM were able to decrease cell viability and proliferation rate, to increase the rate of apoptosis and to reduce glucose-induced insulin secretion. These micromolar T(3) concentrations were significantly higher than the concentration range of T(3) receptor binding, indicating that other non-receptor-mediated mechanisms beyond the receptor level must be responsible for the observed toxic effects of T(3) in vitro.


Assuntos
Apoptose , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Transdução de Sinais , Tri-Iodotironina/metabolismo , Tri-Iodotironina/toxicidade , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Células Secretoras de Insulina/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos
14.
Diabetes ; 53(9): 2271-80, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15331536

RESUMO

Insulin-producing cells are known for their extremely low antioxidant equipment with hydrogen peroxide (H(2)O(2))-inactivating enzymes. Therefore, catalase was stably overexpressed in mitochondria and for comparison in the cytoplasmic compartment of insulin-producing RINm5F cells and analyzed for its protective effect against toxicity of reactive oxygen species (ROS) and proinflammatory cytokines. Only mitochondrial overexpression of catalase provided protection against menadione toxicity, a chemical agent that preferentially generates superoxide radicals intramitochondrially. On the other hand, the cytoplasmic catalase overexpression provided better protection against H(2)O(2) toxicity. Mitochondrial catalase overexpression also preferentially protected against the toxicity of interleukin-1beta (IL-1beta) and a proinflammatory cytokine mixture (IL-1beta, tumor necrosis factor-alpha [TNF-alpha], and gamma-interferon [IFN-gamma]) that is more toxic than IL-1beta alone. Thus, it can be concluded that targeted overexpression of catalase in the mitochondria provides particularly effective protection against cell death in all situations in which ROS are generated intramitochondrially. The observed higher rate of cell death after exposure to a cytokine mixture in comparison with the weaker effect of IL-1beta alone may be due to an additive toxicity of TNF-alpha through ROS formation in mitochondria. The results emphasize the central role of mitochondrially generated ROS in the cytokine-mediated cell destruction of insulin-producing cells.


Assuntos
Catalase/genética , Citocinas/toxicidade , Ilhotas Pancreáticas/enzimologia , Mitocôndrias/enzimologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Antifibrinolíticos/toxicidade , Antineoplásicos/toxicidade , Catalase/metabolismo , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica , Glucose/metabolismo , Peróxido de Hidrogênio/toxicidade , Insulina/metabolismo , Insulinoma , Interferon gama/toxicidade , Interleucina-1/toxicidade , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/imunologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Oxidantes/toxicidade , Neoplasias Pancreáticas , Regiões Promotoras Genéticas/fisiologia , Ratos , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/toxicidade , Vitamina K 3/toxicidade
15.
Free Radic Biol Med ; 34(6): 683-8, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12633745

RESUMO

Insulin-producing cells show very low activity levels of the cytoprotective enzymes catalase, glutathione peroxidase, and superoxide dismutase. This weak antioxidative defense status has been considered a major feature of the poor resistance against oxidative stress. Therefore, we analyzed the protective effect of a combined overexpression of Cu,ZnSOD or MnSOD together with different levels of catalase. Catalase alone was able to increase the resistance of transfected RINm5F insulin-producing tissue culture cells against H(2)O(2) and HX/XO, but no protection was seen in the case of menadione. In combination with an increase of the MnSOD or Cu,ZnSOD expression, the protective action of catalase overexpression could be further increased and extended to the toxicity of menadione. Thus, optimal protection of insulin-producing cells against oxidative stress-mediated toxicity requires a combined overexpression of both superoxide- and hydrogen peroxide-inactivating enzymes. This treatment can compensate for the constitutively low level of antioxidant enzyme expression in insulin-producing cells and may provide an improved protection in situations of free radical-mediated destruction of pancreatic beta cells in the process of autoimmune diabetes development.


Assuntos
Catalase/metabolismo , Expressão Gênica , Glutationa Peroxidase/metabolismo , Insulina/metabolismo , Espécies Reativas de Oxigênio , Superóxido Dismutase/metabolismo , Animais , Antifibrinolíticos/toxicidade , Catalase/genética , Sequestradores de Radicais Livres/metabolismo , Glutationa Peroxidase/genética , Peróxido de Hidrogênio/toxicidade , Hipoglicemiantes/metabolismo , Oxidantes/toxicidade , Ratos , Superóxido Dismutase/genética , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/enzimologia , Células Tumorais Cultivadas/metabolismo , Regulação para Cima , Vitamina K 3/toxicidade , Xantina/toxicidade , Xantina Oxidase/toxicidade
16.
Diabetes ; 52(1): 93-101, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12502498

RESUMO

Proinflammatory cytokines (interleukin-1beta [IL-1beta], tumor necrosis factor-alpha [TNF-alpha], and gamma-interferon [IFN-gamma]) initiate a variety of signal cascades in pancreatic beta-cells that affect the expression level of genes involved in both the destruction and the protection of the beta-cell. The generation of nitric oxide (NO) via the inducible NO synthase (iNOS) and oxygen free radicals play a key role in cytokine-mediated beta-cell destruction. Within these signal cascades, the activation of the transcription factor nuclear factor-kappaB (NF-kappaB) is crucial, and many cytokine-sensitive genes contain binding sites for this transcription factor in their promoter regions. The aim of this study was to characterize the cytokine-mediated activation of NF-kappaB and the subsequent expression of iNOS protein in insulin-producing RINm5F cells with an improved antioxidant defense status by overexpression of the cytoprotective enzymes catalase (Cat), glutathione peroxidase (Gpx), and the cytoplasmic Cu/Zn superoxide dismutase (Cu/ZnSOD). RINm5F cells with diverse mitochondrial antioxidative defense status were generated by stable overexpression of MnSOD constructs in sense (MnSOD sense) and antisense orientation (MnSOD antisense). Cytokine-induced (IL-1beta or cytokine mix consisting of IL-1beta + TNF-alpha + IFN-gamma) activation of NF-kappaB in RINm5F cells was reduced by >80% through overexpression of MnSOD. The activity of the iNOS promoter remained at basal levels in cytokine-stimulated MnSOD sense cells. In contrast, the suppression of MnSOD gene expression in cytokine-stimulated MnSOD antisense cells resulted in a threefold higher activation of NF-kappaB and a twofold higher activation of the iNOS promoter as compared with control cells. The iNOS protein expression was significantly reduced after a 6- and 8-h cytokine incubation of MnSOD sense cells. The low activity level of MnSOD in RINm5F MnSOD antisense cells increased the iNOS protein expression in particular during the early phase of cytokine-mediated toxicity. Cat, Gpx, and the cytoplasmic Cu/ZnSOD did not affect the activation of NF-kappaB and the iNOS promoter. In conclusion, the overexpression of MnSOD, which inactivates specifically mitochondrially derived oxygen free radicals, significantly reduced the activation of NF-kappaB in insulin-producing cells. As a consequence of this protective effect in the early cytokine signaling pathways, the induction of iNOS, an important event in the beta-cell destruction process, was also significantly reduced. The results provide evidence that mitochondrially derived reactive oxygen species (ROS) play a critical role in the activation of the cytokine-sensitive transcription factor NF-kappaB. Overexpression of MnSOD may thus be beneficial for beta-cell survival through suppression of oxygen free radical formation, prevention of NF-kappaB activation, and iNOS expression.


Assuntos
Citocinas/fisiologia , Insulina/biossíntese , Insulinoma/metabolismo , Mitocôndrias/enzimologia , NF-kappa B/fisiologia , Oxirredutases/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Catalase/metabolismo , Citocinas/farmacologia , Combinação de Medicamentos , Glutationa Peroxidase/metabolismo , Interleucina-1/farmacologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Regiões Promotoras Genéticas/fisiologia , Superóxido Dismutase/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...