Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Arch Toxicol ; 96(7): 1915-1920, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35416515

RESUMO

The toxic potential of H2O2 is limited, even if intracellular concentrations of H2O2 under conditions of oxidative stress increase to the micromolar concentration range. Its toxicity is mostly restricted to the oxidation of highly reactive thiol groups, some of which are functionally very important. Subsequently, the HO· radical is generated spontaneously from H2O2 in the Fenton reaction. The HO· radical is extremely toxic and destroys any biological structure. Due to the high reactivity, its action is limited to a locally restricted site of its generation. On the other hand, H2O2 with its stability and long half-life can reach virtually any site and distribute its toxic effect all over the cell. Thereby HO·, in spite of its ultra-short half-life (10-9 s), can execute its extraordinary toxic action at any target of the cell. In this oxidative stress scenario, H2O2 is the pro-radical, that spreads the toxic action of the HO· radical. It is the longevity of the H2O2 molecule allowing it to distribute its toxic action from the site of origin all over the cell and may even mediate intercellular communication. Thus, H2O2 acts as a spreader by transporting it to sites where the extremely short-lived toxic HO· radical can arise in the presence of "free iron". H2O2 and HO· act in concert due to their different complementary chemical properties. They are dependent upon each other while executing the toxic effects in oxidative stress under diabetic metabolic conditions in particular in the highly vulnerable pancreatic beta cell, which in contrast to many other cell types is so badly protected against oxidative stress due to its extremely low H2O2 inactivating enzyme capacity.


Assuntos
Radical Hidroxila , Células Secretoras de Insulina , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/toxicidade , Radical Hidroxila/química , Radical Hidroxila/metabolismo , Células Secretoras de Insulina/metabolismo , Ferro/metabolismo , Oxirredução
2.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35163462

RESUMO

The LEW.1AR1-iddm rat is an animal model of human type 1 diabetes (T1D). Previously, we have shown that combination with anti-TCR/anti-TNF-α antibody-based therapy re-established normoglycemia and increased proteinic arginine-dimethylation in the spleen, yet not in the pancreas. High blood glucose is often associated with elevated formation of advanced glycation end-products (AGEs) which act via their receptor (RAGE). Both anti-TCR and anti-TNF-α are inhibitors of RAGE. The aim of the present work was to investigate potential biochemical changes of anti-TCR/anti-TNF-α therapy in the LEW.1AR1-iddm rat. We determined by stable-isotope dilution gas chromatography-mass spectrometry (GC-MS) the content of free and proteinic AGEs and the Nε-monomethylation of lysine (Lys) residues in proteins of pancreas, kidney, liver, spleen and lymph nodes of normoglycemic control (ngCo, n = 6), acute diabetic (acT1D, n = 6), chronic diabetic (chT1D, n = 4), and cured (cuT1D, n = 4) rats after anti-TCR/anti-TNF-α therapy. Analyzed biomarkers included Lys and its metabolites Nε-carboxymethyl lysine (CML), furosine and Nε-monomethyl lysine (MML). Other amino acids were also determined. Statistical methods including ANOVA, principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were used to evaluate the effects. Most statistical differences between the study groups were observed for spleen, pancreas and kidney, with liver and lymph nodes showing no such differences. In the pancreas, the groups differed with respect to proteinic furosine (p = 0.0289) and free CML (p = 0.0023). In the kidneys, the groups differed with respect to proteinic furosine (p = 0.0076) and CML (p = 0.0270). In the spleen, group differences were found for proteinic furosine (p = 0.0114) and free furosine (p = 0.0368), as well as for proteinic CML (p = 0.0502) and proteinic MML (p = 0.0191). The acT1D rats had lower furosine, CML and MML levels in the spleen than the rats in all other groups. This observation corresponds to the lower citrullination levels previously measured in these rats. PCA revealed diametric associations between PC1 and PC2 for spleen (r = -0.8271, p < 0.0001) compared to pancreas (r = 0.5805, p = 0.0073) and kidney (r = 0.8692, p < 0.0001). These findings underscore the importance of the spleen in this animal model of human T1D. OPLS-DA showed that in total sixteen amino acids differed in the experimental groups.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Diabetes Mellitus Tipo 1/tratamento farmacológico , Lisina/análogos & derivados , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Estudos de Casos e Controles , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animais de Doenças , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Rim/química , Fígado/química , Linfonodos/química , Lisina/análise , Masculino , Pâncreas/química , Ratos , Ratos Endogâmicos Lew , Baço/química
3.
BMC Med ; 18(1): 33, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32106855

RESUMO

BACKGROUND: The cytokine IL-17 is a key player in autoimmune processes, while the cytokine IL-6 is responsible for the chronification of inflammation. However, their roles in type 1 diabetes development are still unknown. METHODS: Therefore, therapies for 5 days with anti-IL-17A or anti-IL-6 in combination with a T cell-specific antibody, anti-TCR, or in a triple combination were initiated immediately after disease manifestation to reverse the diabetic metabolic state in the LEW.1AR1-iddm (IDDM) rat, a model of human type 1 diabetes. RESULTS: Monotherapies with anti-IL-6 or anti-IL-17 showed no sustained anti-diabetic effects. Only the combination therapy of anti-TCR with anti-IL-6 or anti-IL-17 at starting blood glucose concentrations up to 12 mmol/l restored normoglycaemia. The triple antibody combination therapy was effective even up to very high initial blood glucose concentrations (17 mmol/l). The ß cell mass was raised to values of around 6 mg corresponding to those of normoglycaemic controls. In parallel, the apoptosis rate of ß cells was reduced and the proliferation rate increased as well as the islet immune cell infiltrate was strongly reduced in double and abolished in triple combination therapies. CONCLUSIONS: The anti-TCR combination therapy with anti-IL-17 preferentially raised the ß cell mass as a result of ß cell proliferation while anti-IL-6 strongly reduced ß cell apoptosis and the islet immune cell infiltrate with a modest increase of the ß cell mass only. The triple combination therapy achieved both goals in a complimentary anti-autoimmune and anti-inflammatory action resulting in sustained normoglycaemia with normalized serum C-peptide concentrations.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Interleucina-17/antagonistas & inibidores , Interleucina-6/antagonistas & inibidores , Indução de Remissão/métodos , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Ratos , Ratos Endogâmicos Lew
4.
Amino Acids ; 52(1): 103-110, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31832896

RESUMO

The LEW.1AR1-iddm rat is an animal model of human type 1 diabetes (T1D). We determined by GC-MS the extent of asymmetric dimethylation (prADMA) and citrullination (prCit) of L-arginine residues in organ proteins (pr) of normoglycaemic control (ngCo, n = 6), acutely diabetic (acT1D, n = 6), chronically diabetic (chT1D, n = 4), and cured (cuT1D, n = 4) rats after anti-TCR/anti-TNF-α therapy. Pancreatic prCit and prADMA did not differ between the groups but were correlated (r = 0.728, P = 0.0003, n = 20). acT1D rats had lower prCit levels in spleen and kidney than ngCo rats. cuT1D rats had higher prADMA levels than chT1D rats only in the spleen. Combination therapy re-established normoglycaemia and increased prADMA in the spleen without altering pancreatic prADMA and prCit. Western blotting demonstrated the presence of different prADMA pattern, especially an ≈ 50-kDa prADMA in spleen and pancreas, and an ≈ 25-kDa prADMA in the pancreas only, with the kidney showing only a very faint and small prADMA. Besides the changes in the pancreas during different metabolic states, the spleen may play a stronger role for the recognition of metabolic changes in T1D than thought thus far.


Assuntos
Anticorpos/farmacologia , Arginina/genética , Diabetes Mellitus Tipo 1/tratamento farmacológico , Fator de Necrose Tumoral alfa/genética , Animais , Anticorpos/imunologia , Glicemia/genética , Citrulinação/efeitos dos fármacos , Citrulinação/genética , Metilação de DNA/genética , Metilação de DNA/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Humanos , Masculino , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores de Antígenos de Linfócitos T alfa-beta/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Baço/efeitos dos fármacos , Baço/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
5.
Diabetologia ; 61(3): 641-657, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29185012

RESUMO

AIMS/HYPOTHESIS: Pancreatic islet beta cell failure causes type 2 diabetes in humans. To identify transcriptomic changes in type 2 diabetic islets, the Innovative Medicines Initiative for Diabetes: Improving beta-cell function and identification of diagnostic biomarkers for treatment monitoring in Diabetes (IMIDIA) consortium ( www.imidia.org ) established a comprehensive, unique multicentre biobank of human islets and pancreas tissues from organ donors and metabolically phenotyped pancreatectomised patients (PPP). METHODS: Affymetrix microarrays were used to assess the islet transcriptome of islets isolated either by enzymatic digestion from 103 organ donors (OD), including 84 non-diabetic and 19 type 2 diabetic individuals, or by laser capture microdissection (LCM) from surgical specimens of 103 PPP, including 32 non-diabetic, 36 with type 2 diabetes, 15 with impaired glucose tolerance (IGT) and 20 with recent-onset diabetes (<1 year), conceivably secondary to the pancreatic disorder leading to surgery (type 3c diabetes). Bioinformatics tools were used to (1) compare the islet transcriptome of type 2 diabetic vs non-diabetic OD and PPP as well as vs IGT and type 3c diabetes within the PPP group; and (2) identify transcription factors driving gene co-expression modules correlated with insulin secretion ex vivo and glucose tolerance in vivo. Selected genes of interest were validated for their expression and function in beta cells. RESULTS: Comparative transcriptomic analysis identified 19 genes differentially expressed (false discovery rate ≤0.05, fold change ≥1.5) in type 2 diabetic vs non-diabetic islets from OD and PPP. Nine out of these 19 dysregulated genes were not previously reported to be dysregulated in type 2 diabetic islets. Signature genes included TMEM37, which inhibited Ca2+-influx and insulin secretion in beta cells, and ARG2 and PPP1R1A, which promoted insulin secretion. Systems biology approaches identified HNF1A, PDX1 and REST as drivers of gene co-expression modules correlated with impaired insulin secretion or glucose tolerance, and 14 out of 19 differentially expressed type 2 diabetic islet signature genes were enriched in these modules. None of these signature genes was significantly dysregulated in islets of PPP with impaired glucose tolerance or type 3c diabetes. CONCLUSIONS/INTERPRETATION: These studies enabled the stringent definition of a novel transcriptomic signature of type 2 diabetic islets, regardless of islet source and isolation procedure. Lack of this signature in islets from PPP with IGT or type 3c diabetes indicates differences possibly due to peculiarities of these hyperglycaemic conditions and/or a role for duration and severity of hyperglycaemia. Alternatively, these transcriptomic changes capture, but may not precede, beta cell failure.


Assuntos
Bancos de Espécimes Biológicos , Diabetes Mellitus Tipo 2/metabolismo , Biologia de Sistemas/métodos , Doadores de Tecidos , Transcriptoma/genética , Idoso , Idoso de 80 Anos ou mais , Biologia Computacional , Feminino , Humanos , Masculino , Pancreatectomia
6.
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
7.
Amino Acids ; 50(7): 799-821, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29728915

RESUMO

We recently found that renal carbonic anhydrase (CA) is involved in the reabsorption of inorganic nitrite (NO2-), an abundant reservoir of nitric oxide (NO) in tissues and cells. Impaired NO synthesis in the endothelium and decreased NO bioavailability in the circulation are considered major contributors to the development and progression of renal and cardiovascular diseases in different conditions including diabetes. Isolated human and bovine erythrocytic CAII and CAIV can convert nitrite to nitrous acid (HONO) and its anhydride N2O3 which, in the presence of thiols (RSH), are further converted to S-nitrosothiols (RSNO) and NO. Thus, CA may be responsible both for the homeostasis of nitrite and for its bioactivation to RSNO/NO. We hypothesized that enhanced excretion of nitrite in the urine may contribute to NO-related dysfunctions in the renal and cardiovascular systems, and proposed the urinary nitrate-to-nitrite molar ratio, i.e., UNOxR, as a measure of renal CA-dependent excretion of nitrite. Based on results from clinical and experimental animal studies, here, we report on a first evaluation of UNOxR. We determined UNOxR values in preterm neonates, healthy children, and adults, in children suffering from type 1 diabetes mellitus (T1DM) or Duchenne muscular dystrophy (DMD), in elderly subjects suffering from chronic rheumatic diseases, type 2 diabetes mellitus (T2DM), coronary artery disease (CAD), or peripheral arterial occlusive disease (PAOD). We also determined UNOxR values in healthy young men who ingested isosorbide dinitrate (ISDN), pentaerythrityl tetranitrate (PETN), or inorganic nitrate. In addition, we tested the utility of UNOxR in two animal models, i.e., the LEW.1AR1-iddm rat, an animal model of human T1DM, and the APOE*3-Leiden.CETP mice, a model of human dyslipidemia. Mean UNOxR values were lower in adult patients with rheumatic diseases (187) and in T2DM patients of the DALI study (74) as compared to healthy elderly adults (660) and healthy young men (1500). The intra- and inter-variabilities of UNOxR were of the order of 50% in young and elderly healthy subjects. UNOxR values were lower in black compared to white boys (314 vs. 483, P = 0.007), which is in line with reported lower NO bioavailability in black ethnicity. Mean UNOxR values were lower in DMD (424) compared to healthy (730) children, but they were higher in T1DM children (1192). ISDN (3 × 30 mg) decreased stronger UNOxR compared to PETN (3 × 80 mg) after 1 day (P = 0.046) and after 5 days (P = 0.0016) of oral administration of therapeutically equivalent doses. In healthy young men who ingested NaNO3 (0.1 mmol/kg/d), UNOxR was higher than in those who ingested the same dose of NaCl (1709 vs. 369). In LEW.1AR1-iddm rats, mean UNOxR values were lower than in healthy rats (198 vs. 308) and comparable to those in APOE*3-Leiden.CETP mice (151).


Assuntos
Diabetes Mellitus Tipo 1/urina , Diabetes Mellitus Tipo 2/urina , Rim/metabolismo , Nitratos/urina , Nitritos/urina , Doenças Reumáticas/urina , Animais , Arteriopatias Oclusivas/sangue , Arteriopatias Oclusivas/urina , Anidrases Carbônicas/metabolismo , Bovinos , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/urina , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 2/sangue , Camundongos , Distrofia Muscular de Duchenne/sangue , Distrofia Muscular de Duchenne/urina , Óxido Nítrico/sangue , Ratos , Doenças Reumáticas/sangue
8.
Pediatr Diabetes ; 19(5): 963-971, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29527790

RESUMO

OBJECTIVE: Type 1 diabetes (T1D) develops in distinct stages, before and after disease onset. Whether the natural course translates into different immunologic patterns is still uncertain. This study aimed at identifying peripheral immune patterns at key time-points, in T1D children undergoing remission phase. METHODS: Children with new-onset T1D and healthy age and gender-matched controls were recruited at a pediatric hospital. Peripheral blood samples were evaluated by flow cytometry at 3 longitudinal time-points: onset (T1), remission phase (T2) and established disease (T3). Cytokine levels were quantified by multiplex assay. Fasting C-peptide, HbA1c, and 25OHD were also measured. RESULTS: T1D children (n = 28; 10.0 ± 2.6 years) showed significant differences from controls in circulating neutrophils, T helper (Th)17 and natural killer (NK) cells, with relevant variations during disease progression. At onset, neutrophils, NK, Th17 and T cytotoxic (Tc)17 cells were decreased. As disease progressed, neutrophil counts recovered whereas NK counts remained low. Th17 and Tc17 cells behavior followed the neutrophil variation pattern. B-cells were lowest in the remission phase and regulatory T-cells significantly declined after remission. Two cytokine response profiles were identified. Low cytokine-responders showed higher circulating fasting C-peptide levels at onset and longer remission periods. C-peptide inversely correlated with pro-inflammatory and cytotoxic cells. CONCLUSIONS: Our data suggest an association between immune cells, cytokine patterns and metabolic counterparts. The dynamic changes of circulating immune cells during disease progression involve key innate and acquired immune cell types. This longitudinal picture of T1D progression may enable disease staging and patient stratification, essential for individualized treatment.


Assuntos
Citocinas/sangue , Diabetes Mellitus Tipo 1/imunologia , Adolescente , Peptídeo C/sangue , Estudos de Casos e Controles , Criança , Diabetes Mellitus Tipo 1/sangue , Progressão da Doença , Feminino , Humanos , Contagem de Leucócitos , Estudos Longitudinais , Masculino
9.
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
10.
Diabetes Metab Res Rev ; 33(7)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28692149

RESUMO

Animal models of human type 1 diabetes will be of a great importance for the evaluation of new combination therapies with curative potential. However, reliable predictive power for successful translation to patients with type 1 diabetes is crucial. This will be particularly important in the future when evaluating success of new combination therapies that show great promise for preservation and restoration of beta cell mass and thereby reverse the type 1 diabetic hyperglycaemia. But not all spontaneous animal models are equally well suited for this purpose. The advantages and disadvantages of the three spontaneous rat models (BioBreeding diabetes-prone [BB] rat, Komeda [KDP] rat, and LEW.1AR1-iddm [IDDM] rat) as well as the NOD mouse, compared with the characteristics of human type 1 diabetes, are considered in this review.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Modelos Animais de Doenças , Hipoglicemiantes/uso terapêutico , Animais , Quimioterapia Combinada , Humanos , Camundongos , Ratos , Pesquisa Translacional Biomédica
11.
Biochim Biophys Acta Gen Subj ; 1861(8): 1929-1942, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28527893

RESUMO

BACKGROUND: Diabetes mellitus is a serious metabolic disease. Dysfunction and subsequent loss of the ß-cells in the islets of Langerhans through apoptosis ultimately cause a life-threatening insulin deficiency. The underlying reason for the particular vulnerability of the ß-cells is an extraordinary sensitivity to the toxicity of reactive oxygen and nitrogen species (ROS and RNS) due to its low antioxidative defense status. SCOPE REVIEW: This review considers the different aspects of the chemistry and biology of the biologically most important reactive species and their chemico-biological interactions in the ß-cell toxicity of proinflammatory cytokines in type 1 diabetes and of lipotoxicity in type 2 diabetes development. MAJOR CONCLUSION: The weak antioxidative defense equipment in the different subcellular organelles makes the ß-cells particularly vulnerable and prone to mitochondrial, peroxisomal and ER stress. Looking upon the enzyme deficiencies which are responsible for the low antioxidative defense status of the pancreatic ß-cells it is the lack of enzymatic capacity for H2O2 inactivation at all major subcellular sites. GENERAL SIGNIFICANCE: Diabetes is the most prevalent metabolic disorder with a steadily increasing incidence of both type 1 and type 2 diabetes worldwide. The weak protection of the pancreatic ß-cells against oxidative stress is a major reason for their particular vulnerability. Thus, careful protection of the ß-cells is required for prevention of the disease.


Assuntos
Antioxidantes/metabolismo , Células Secretoras de Insulina/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Diabetes Mellitus/metabolismo , Estresse do Retículo Endoplasmático , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Estresse Oxidativo
12.
Diabetologia ; 59(10): 2125-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27460666

RESUMO

AIMS/HYPOTHESIS: The aim of this study was to perform a detailed analysis of cytokine toxicity in the new human EndoC-ßH1 beta cell line. METHODS: The expression profile of the antioxidative enzymes in the new human EndoC-ßH1 beta cells was characterised and compared with that of primary beta cells in the human pancreas. The effects of proinflammatory cytokines on reactive oxygen species formation, insulin secretory responsiveness and apoptosis of EndoC-ßH1 beta cells were determined. RESULTS: EndoC-ßH1 beta cells were sensitive to the toxic action of proinflammatory cytokines. Glucose-dependent stimulation of insulin secretion and an increase in the ATP/ADP ratio was abolished by proinflammatory cytokines without induction of IL-1ß expression. Cytokine-mediated caspase-3 activation was accompanied by reactive oxygen species formation and developed more slowly than in rodent beta cells. Cytokines transiently increased the expression of unfolded protein response genes, without inducing endoplasmic reticulum stress-marker genes. Cytokine-mediated NFκB activation was too weak to induce inducible nitric oxide synthase expression. The resultant lack of nitric oxide generation in EndoC-ßH1 cells, in contrast to rodent beta cells, makes these cells dependent on exogenously generated nitric oxide, which is released from infiltrating immune cells in human type 1 diabetes, for full expression of proinflammatory cytokine toxicity. CONCLUSIONS/INTERPRETATION: EndoC-ßH1 beta cells are characterised by an imbalance between H2O2-generating and -inactivating enzymes, and react to cytokine exposure in a similar manner to primary human beta cells. They are a suitable beta cell surrogate for cytokine-toxicity studies.


Assuntos
Citocinas/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Western Blotting , Caspase 3/metabolismo , Linhagem Celular , Citometria de Fluxo , Imunofluorescência , Glucose/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Insulina/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pancrelipase/metabolismo , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase-1/metabolismo
13.
Diabetologia ; 58(12): 2800-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26363782

RESUMO

AIMS/HYPOTHESIS: The LEW.1AR1-iddm rat, an animal model of human type 1 diabetes, arose through a spontaneous mutation within the inbred strain LEW.1AR1. A susceptibility locus (Iddm8) on rat chromosome 1 (RNO1) has been identified previously, which is accompanied by autoimmune diabetes and the additional phenotype of a variable CD3(+) T cell frequency. METHODS: In the present study we characterised the Iddm8 region on RNO1 in backcross strains using the genetically divergent Brown Norway (BN) and Paris (PAR) rats. Candidate genes of the Iddm8 region were sequenced for mutation analysis. RESULTS: The Iddm8 region could be subdivided by single nucleotide polymorphism (SNP) analyses. In the first region, a mutation in exon 44 of the Dock8 gene was identified resulting in an amino acid exchange in the protein from glutamine to glutamate. This exchange is unique for the LEW.1AR1-iddm rat. In the second region, a SNP was detected in exon 11 of the Vwa2 gene with an exchange from arginine to tryptophan. This SNP is also present in other rat strains. CONCLUSIONS/INTERPRETATION: The Dock8 mutation gave rise to a new type 1 diabetes rat model with very close similarity to type 1 diabetes in humans, providing a deepened insight into the impact of genes involved in diabetes development.


Assuntos
Diabetes Mellitus Tipo 1/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Mutação , Alelos , Substituição de Aminoácidos , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Éxons/genética , Humanos , Células Matadoras Naturais , Modelos Moleculares , Polimorfismo de Nucleotídeo Único , Ratos , Ratos Endogâmicos Lew , Complexo Receptor-CD3 de Antígeno de Linfócitos T/genética , Fator de von Willebrand/genética
14.
J Biol Chem ; 289(18): 12189-94, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24637025

RESUMO

This minireview looks back at a century of glycolysis research with a focus on the mechanisms of flux regulation. Traditionally, glycolysis is regarded as a feeder pathway that prepares glucose for further catabolism and energy production. However, glycolysis is much more than that, in particular in those tissues that express the low affinity glucose-phosphorylating enzyme glucokinase. This enzyme equips the glycolytic pathway with a special steering function for the regulation of intermediary metabolism. In beta cells, glycolysis acts as a transducer for triggering and amplifying physiological glucose-induced insulin secretion. On the basis of these considerations, I have defined a glycolytic flux regulatory unit composed of the two fructose ester steps of this pathway with various enzymes and metabolites that regulate glycolysis.


Assuntos
Glucoquinase/metabolismo , Glicólise , Animais , Glucose/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Modelos Biológicos , Fosforilação
15.
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
16.
Biochim Biophys Acta ; 1843(3): 554-64, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24333860

RESUMO

The glucose phosphorylating enzyme glucokinase regulates glucose metabolism in the liver. Glucokinase activity is modulated by a liver-specific competitive inhibitor, the glucokinase regulatory protein (GRP), which mediates sequestration of glucokinase to the nucleus at low glucose concentrations. However, the mechanism of glucokinase nuclear export is not fully understood. In this study we investigated the dynamics of glucose-dependent interaction and translocation of glucokinase and GRP in primary hepatocytes using fluorescence resonance energy transfer, selective photoconversion and fluorescence recovery after photobleaching. The formation of the glucokinase:GRP complex in the nucleus of primary hepatocytes at 5 mmol/l glucose was significantly reduced after a 2 h incubation at 20 mmol/l glucose. The GRP was predominantly localized in the nucleus, but a mobile fraction moved between the nucleus and the cytoplasm. The glucose concentration only marginally affected GRP shuttling. In contrast, the nuclear export rate of glucokinase was significantly higher at 20 than at 5 mmol/l glucose. Thus, glucose was proven to be the driving-force for nuclear export of glucokinase in hepatocytes. Using the FLII2Pglu-700mu-delta6 glucose nanosensor it could be shown that in hepatocytes the kinetics of nuclear glucose influx, metabolism or efflux were significantly faster compared to insulin-secreting cells. The rapid equilibration kinetics of glucose flux into the nucleus facilitates dissociation of the glucokinase:GRP complex and also nuclear glucose metabolism by free glucokinase enzyme. In conclusion, we could show that a rise of glucose in the nucleus of hepatocytes releases active glucokinase from the glucokinase:GRP complex and promotes the subsequent nuclear export of glucokinase.


Assuntos
Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Glucoquinase/metabolismo , Glucose/metabolismo , Hepatócitos/metabolismo , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Citoplasma/metabolismo , Células Secretoras de Insulina/metabolismo , Cinética , Camundongos , Transporte Proteico , Ratos
17.
Cell Physiol Biochem ; 36(3): 852-65, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26044490

RESUMO

BACKGROUND/AIMS: Elevated levels of non-esterified fatty acids (NEFAs) are under suspicion to mediate ß-cell dysfunction and ß-cell loss in type 2 diabetes, a phenomenon known as lipotoxicity. Whereas saturated fatty acids show a strong cytotoxic effect upon insulin-producing cells, unsaturated fatty acids are not toxic and can even prevent toxicity. Experimental evidence suggests that oxidative stress mediates lipotoxicity and there is evidence that the subcellular site of ROS formation is the peroxisome. However, the interaction between unsaturated and saturated NEFAs in this process is unclear. METHODS: Toxicity of rat insulin-producing cells after NEFA incubation was measured by MTT and caspase assays. NEFA induced H2O2 formation was quantified by organelle specific expression of the H2O2 specific fluorescence sensor protein HyPer. RESULTS: The saturated NEFA palmitic acid had a significant toxic effect on the viability of rat insulin-producing cells. Unsaturated NEFAs with carbon chain lengths >14 showed, irrespective of the number of double bonds, a pronounced protection against palmitic acid induced toxicity. Palmitic acid induced H2O2 formation in the peroxisomes of insulin-producing cells. Oleic acid incubation led to lipid droplet formation, but in contrast to palmitic acid induced neither an ER stress response nor peroxisomal H2O2 generation. Furthermore, oleic acid prevented palmitic acid induced H2O2 production in the peroxisomes. CONCLUSION: Thus unsaturated NEFAs prevent deleterious hydrogen peroxide generation during peroxisomal ß-oxidation of long-chain saturated NEFAs in rat insulin-producing cells.


Assuntos
Peróxido de Hidrogênio/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Ácido Oleico/farmacologia , Ácido Palmítico/toxicidade , Peroxissomos/efeitos dos fármacos , Animais , Bioensaio , Sobrevivência Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Peróxido de Hidrogênio/antagonistas & inibidores , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Gotículas Lipídicas/efeitos dos fármacos , Gotículas Lipídicas/metabolismo , Masculino , Ácido Palmítico/antagonistas & inibidores , Peroxissomos/metabolismo , Cultura Primária de Células , Ratos , Ratos Endogâmicos Lew
18.
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
19.
Diabetologia ; 57(3): 512-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24310561

RESUMO

AIMS/HYPOTHESIS: Research on the pathogenesis of type 1 diabetes relies heavily on good animal models. The aim of this work was to study the translational value of animal models of type 1 diabetes to the human situation. METHODS: We compared the four major animal models of spontaneous type 1 diabetes, namely the NOD mouse, BioBreeding (BB) rat, Komeda rat and LEW.1AR1-iddm rat, by examining the immunohistochemistry and in situ RT-PCR of immune cell infiltrate and cytokine pattern in pancreatic islets, and by comparing findings with human data. RESULTS: After type 1 diabetes manifestation CD8(+) T cells, CD68(+) macrophages and CD4(+) T cells were observed as the main immune cell types with declining frequency, in infiltrated islets of all diabetic pancreases. IL-1ß and TNF-α were the main proinflammatory cytokines in the immune cell infiltrate in NOD mice, BB rats and LEW.1AR1-iddm rats, as well as in humans. The Komeda rat was the exception, with IFN-γ and TNF-α being the main cytokines. In addition, IL-17 and IL-6 and the anti-inflammatory cytokines IL-4, IL-10 and IL-13 were found in some infiltrating immune cells. Apoptotic as well as proliferating beta cells were observed in infiltrated islets. In healthy pancreases no proinflammatory cytokine expression was observed. CONCLUSIONS/INTERPRETATION: With the exception of the Komeda rat, the animal models mirror very well the situation in humans with type 1 diabetes. Thus animal models of type 1 diabetes can provide meaningful information on the disease processes in the pancreas of patients with type 1 diabetes.


Assuntos
Apoptose , Linfócitos B/patologia , Citocinas/metabolismo , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/patologia , Animais , Apoptose/imunologia , Linfócitos B/imunologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/imunologia , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Ratos , Ratos Endogâmicos BB , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/metabolismo
20.
J Biol Chem ; 288(50): 35824-39, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24187134

RESUMO

Glucokinase acts as a glucose sensor in pancreatic beta cells. Its posttranslational regulation is important but not yet fully understood. Therefore, a pancreatic islet yeast two-hybrid library was produced and searched for glucokinase-binding proteins. A protein sequence containing a full-length ubiquitin-like domain was identified to interact with glucokinase. Mammalian two-hybrid and fluorescence resonance energy transfer analyses confirmed the interaction between glucokinase and the ubiquitin-like domain in insulin-secreting MIN6 cells and revealed the highest binding affinity at low glucose. Overexpression of parkin, an ubiquitin E3 ligase exhibiting an ubiquitin-like domain with high homology to the identified, diminished insulin secretion in MIN6 cells but had only some effect on glucokinase activity. Overexpression of the elucidated ubiquitin-like domain or midnolin, containing exactly this ubiquitin-like domain, significantly reduced both intrinsic glucokinase activity and glucose-induced insulin secretion. Midnolin has been to date classified as a nucleolar protein regulating mouse development. However, we could not confirm localization of midnolin in nucleoli. Fluorescence microscopy analyses revealed localization of midnolin in nucleus and cytoplasm and co-localization with glucokinase in pancreatic beta cells. In addition we could show that midnolin gene expression in pancreatic islets is up-regulated at low glucose and that the midnolin protein is highly expressed in pancreatic beta cells and also in liver, muscle, and brain of the adult mouse and cell lines of human and rat origin. Thus, the results of our study suggest that midnolin plays a role in cellular signaling of adult tissues and regulates glucokinase enzyme activity in pancreatic beta cells.


Assuntos
Glucoquinase/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Ubiquitina/química , Sequência de Aminoácidos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Especificidade de Órgãos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Ratos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA