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.
Ophthalmologe ; 116(3): 288-290, 2019 Mar.
Artigo em Alemão | MEDLINE | ID: mdl-30367232

RESUMO

The manual capsulorhexis created by Neuhann is still the standard procedure for opening the anterior capsule for cataract surgery. A limitation is the inaccuracy in the size and placement of the opening due to manual execution. In addition to the femtosecond laser a possible improvement in the standardization of capsulorhexis is provided by the Zepto procedure (precision pulse capsulotomy, PPC). In this case a 5.25 mm rhexis is created in a standardized fashion with a flexible suction adapter in which a nitinol ring is located. Whether the strength of PPC is comparable or better than that of the manual technique and how it behaves in terms of capsule shrinkage has not yet been finally clarified.


Assuntos
Extração de Catarata , Terapia a Laser , Capsulorrexe , Facoemulsificação
2.
Z Rheumatol ; 77(6): 469-476, 2018 Aug.
Artigo em Alemão | MEDLINE | ID: mdl-29881952

RESUMO

Intraocular inflammation with the imprecise and broad umbrella term "uveitis" is a diagnostic and therapeutic challenge in ophthalmology. Uveitis is one of the most common causes of blindness worldwide and due to the associated costs is comparable to diabetic retinopathy. Patients can be affected by uveitis at any age. Any part of the eye may be affected. The symptoms range from complete absence of symptoms, through all types of vision deterioration up to a red and even very painful eye. Uveitis can be strictly unilateral (also alternating from the left to the right eye) or bilateral with a relapsing or chronic course. The transitions are smooth and the differential diagnoses are very broad. In addition to infectious forms and ocular syndromes restricted to the eye, it also includes those with extraocular systemic diseases, such as ankylosing spondylitis or sarcoidosis. All commonly administered immunosuppressive treatment strategies in rheumatology can be used for non-infectious forms in addition to local and regional forms of treatment. The diagnostic and therapeutic impulses of this interdisciplinary interface between rheumatology and ophthalmology is discussed in more detail in this article.


Assuntos
Oftalmologistas , Reumatologia , Sarcoidose , Uveíte , Humanos , Reumatologistas
3.
J Mol Endocrinol ; 55(1): 21-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26108484

RESUMO

Oxidative folding of nascent proteins in the endoplasmic reticulum (ER), catalysed by one or more members of the protein disulfide isomerase family and the sulfhydryl oxidase ER oxidoreductin 1 (ERO1), is accompanied by generation of hydrogen peroxide (H2O2). Because of the high rate of insulin biosynthesis and the low expression of H2O2-inactivating enzymes in pancreatic ß cells, it has been proposed that the luminal H2O2 concentration might be very high. As the role of this H2O2 in ER stress and proinsulin processing is still unsolved, an ER-targeted and luminal-active catalase variant, ER-Catalase N244, was expressed in insulin-secreting INS-1E cells. In these cells, the influence of ER-specific H2O2 removal on cytokine-mediated cytotoxicity and ER stress, insulin gene expression, insulin content and secretion was analysed. The expression of ER-Catalase N244 reduced the toxicity of exogenously added H2O2 significantly with a threefold increase of the EC50 value for H2O2. However, the expression of cytokine-induced ER stress genes and viability after incubation with ß cell toxic cytokines (IL1ß alone or together with TNFα+IFNγ) was not affected by ER-Catalase N244. In control and ER-Catalase N244 expressing cells, insulin secretion and proinsulin content was identical, while removal of luminal H2O2 reduced insulin gene expression and insulin content in ER-Catalase N244 expressing cells. These data show that ER-Catalase N244 reduced H2O2 toxicity but did not provide protection against pro-inflammatory cytokine-mediated toxicity and ER stress. Insulin secretion was not affected by decreasing H2O2 in the ER in spite of a reduced insulin transcription and processing.


Assuntos
Retículo Endoplasmático/metabolismo , Peróxido de Hidrogênio/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Catalase/metabolismo , Células Cultivadas , Estresse do Retículo Endoplasmático/fisiologia , Insulina/metabolismo , Oxirredução , Estresse Oxidativo/fisiologia , Oxirredutases/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo Enxofre/metabolismo , Dobramento de Proteína , Ratos , Espécies Reativas de Oxigênio/metabolismo , Técnicas de Cultura de Tecidos
4.
Free Radic Biol Med ; 80: 77-83, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25499853

RESUMO

Disulfide bond formation during protein folding of nascent proteins is associated with the generation of H2O2 in the endoplasmic reticulum (ER). Approaches to quantifying H2O2 directly within the ER failed because of the oxidative environment in the ER lumen, and ER-specific catalase expression to detoxify high H2O2 concentrations resulted in an inactive protein owing to N-glycosylation. Therefore, the N-glycosylation motifs at asparagine-244 and -439 of the human catalase protein were deleted by site-directed mutagenesis. The ER-targeted expression of these variants revealed that the deletion of the N-glycosylation motif only at asparagine-244 (N244) was associated with the maintenance of full enzymatic activity in the ER. Expression of catalase N244 in the ER (ER-Catalase N244) was ER-specific and protected the cells significantly against exogenously added H2O2. With the expression of ER-Catalase N244, a highly effective H2O2 inactivation within the ER was achieved for the first time. Catalase has a high H2O2-inactivation capacity without the need of reducing cofactors, which might interfere with the ER redox homeostasis, and is not involved in protein folding. With these characteristics ER-Catalase N244 is an ideal tool to explore the impact of ER-generated H2O2 on the generation of disulfide bonds or to study the induction of ER-stress pathways through protein folding overload and accumulation of H2O2.


Assuntos
Catalase/metabolismo , Retículo Endoplasmático/enzimologia , Peróxido de Hidrogênio/farmacologia , Células Secretoras de Insulina/enzimologia , Engenharia de Proteínas , Animais , Asparagina/química , Asparagina/metabolismo , Sequência de Bases , Catalase/química , Catalase/genética , Linhagem Celular Tumoral , Dissulfetos/química , Dissulfetos/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Vetores Genéticos , Glicosilação , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Lentivirus/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Oxirredução , Estresse Oxidativo , Plasmídeos/química , Plasmídeos/metabolismo , Dobramento de Proteína , Ratos , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
5.
J Mol Endocrinol ; 52(3): 301-10, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24850829

RESUMO

Free intracellular ferrous iron (Fe(2+)) is essential for the generation of the extremely toxic hydroxyl radicals, which contribute to ß-cell destruction by cytokines. Therefore the expression of the different divalent metal transporter 1 (Dmt1) isoforms and ferritin (Ft) subunits, responsible for iron import and chelation, was analyzed under pro-inflammatory conditions (IL1ß alone or together with TNFα+IFNγ). The Dmt1 isoforms (1A/1B and +IRE/-IRE) and the total Dmt1 expression in insulin-producing cells (RINm5F and INS-1E), in primary rat islets and, for comparison, in the neuroendocrine PC12 cell line were quantified by qRT-PCR. In addition, the expression of the light (L-Ft) and heavy Ft (H-Ft) subunits and the mitochondrial Ft isoform (Mtft) in insulin-producing cells under control conditions and after cytokine treatment was estimated. The 1B isoform was the predominant Dmt1 mRNA in all insulin-producing cells, accounting for almost 100% of the 1A/1B isoform expression. For the IRE variants, +IRE expression was higher than -IRE expression. Pro-inflammatory cytokines accelerated the expression of Dmt1 isoforms significantly with an overall 2.5- to 3-fold increase in the total Dmt1 expression. In contrast, the expression of the iron-buffering ferritin subunits L- and H-Ft was unaffected by IL1ß and only slightly induced by the cytokine mixture. Mtft expression was also not increased. Dmt1 expression was significantly elevated through pro-inflammatory cytokines, whereas Ft expression was marginally increased. This imbalance between the increased iron transport capacity and the almost unaffected iron storage capacity can foster cytokine-mediated formation of hydroxyl radicals and thus pro-inflammatory cytokine toxicity through elevated free iron concentrations.


Assuntos
Proteínas de Transporte de Cátions/biossíntese , Ferritinas/biossíntese , Inflamação/imunologia , Células Secretoras de Insulina/metabolismo , Ferro/metabolismo , Animais , Transporte Biológico , Proteínas de Transporte de Cátions/genética , Hipóxia Celular , Linhagem Celular , Diabetes Mellitus , Ferritinas/genética , Interferon gama/metabolismo , Interleucina-1beta/metabolismo , Masculino , Células PC12 , Isoformas de Proteínas/biossíntese , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos Lew , Fator de Necrose Tumoral alfa/metabolismo
6.
Ophthalmologe ; 111(2): 121-7, 2014 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-24337343

RESUMO

A broad spectrum of anti-inflammatory drugs with different mechanisms is available for the treatment of intraocular inflammation. Corticosteroids are the mainstay of therapy. Mechanisms of action are quite well understood for most drugs in particular taken from animal research studies. However, pharmacokinetic evidence for treatment of ocular disease is generally limited for the human eye and especially for ocular inflammation. The bioavailability of a particular drug in an inflamed eye is expected to be faster due to a barrier breakdown. Therefore, intraocular level of effective substances should be lowered more rapidly than in a non-inflamed eye due to improved drainage. This article reviews current knowledge firstly about local, regional and systemic anti-inflammatory therapy of uveitis and finally on immuosuppressive systemic therapy.


Assuntos
Administração Oftálmica , Imunossupressores/administração & dosagem , Imunossupressores/farmacocinética , Modelos Biológicos , Absorção Ocular , Uveíte/tratamento farmacológico , Uveíte/metabolismo , Disponibilidade Biológica , Humanos , Injeções Intra-Arteriais , Injeções Intravenosas , Taxa de Depuração Metabólica
7.
Diabetologia ; 56(4): 774-82, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23306382

RESUMO

AIMS/HYPOTHESIS: Hydrogen peroxide (H2O2)-inactivating enzymes such as catalase are produced in extraordinarily low levels in beta cells. Whether this low expression might be related to a signalling function of H2O2 within the beta cell is unknown. A high level of H2O2-inactivating enzymes could potentially be incompatible with glucose-induced insulin secretion. Therefore the effect of catalase overexpression on mitochondrial function and physiological insulin secretion was studied in insulin-secreting INS-1E and primary islet cells. METHODS: INS-1E and rat islet cells were lentivirally transduced to overexpress catalase in the cytosol (CytoCat) or in mitochondria (MitoCat). Cell viability and caspase-3 activation were assessed after cytokine incubation and hypoxia. Insulin secretion was quantified and expression of the gene encoding the mitochondrial uncoupling protein 2 (Ucp2) was measured in parallel to mitochondrial membrane potential and reactive oxygen species (ROS) formation. RESULTS: The ability to secret insulin in a glucose-dependent manner was not suppressed by catalase overexpression, although the glucose-dependent increase in the mitochondrial membrane potential was attenuated in MitoCat cells along with an increased Ucp2 expression and reduced mitochondrial ROS formation. In addition, MitoCat overexpressing cells were significantly more resistant against pro-inflammatory cytokines and hypoxia than CytoCat and control cells. CONCLUSIONS/INTERPRETATION: The results demonstrate that an improved antioxidative defence status of insulin-secreting cells allowing efficient H2O2 inactivation is not incompatible with proper insulin secretory responsiveness to glucose stimulation and provide no support for a signalling role of H2O2 in insulin-secreting cells. Interestingly, the results also document for the first time that the decreased ROS formation with increasing glucose concentrations is of mitochondrial origin.


Assuntos
Catalase/metabolismo , Regulação Enzimológica da Expressão Gênica , Glucose/metabolismo , Ilhotas Pancreáticas/citologia , 2,4-Dinitrofenol/farmacologia , Animais , Glicemia/metabolismo , Citocinas/metabolismo , Citosol/metabolismo , Citometria de Fluxo , Peróxido de Hidrogênio/farmacologia , Insulina/metabolismo , Canais Iônicos/metabolismo , Masculino , Potenciais da Membrana , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Estresse Oxidativo , Ratos , Ratos Endogâmicos Lew , Espécies Reativas de Oxigênio , Transdução de Sinais , Proteína Desacopladora 2
8.
Diabetologia ; 53(6): 1120-30, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20349222

RESUMO

AIMS/HYPOTHESIS: Pro-inflammatory cytokines involved in the pathogenesis of type 1 diabetes deplete endoplasmic reticulum (ER) Ca2+ stores, leading to ER-stress and beta cell apoptosis. However, the cytokine-induced ER-stress response in beta cells is atypical and characterised by induction of the pro-apoptotic PKR-like ER kinase (PERK)-C/EBP homologous protein (CHOP) branch of the unfolded protein response, but defective X-box binding protein 1 (XBP1) splicing and activating transcription factor 6 activation. The purpose of this study was to overexpress spliced/active Xbp1 (XBP1s) to increase beta cell resistance to cytokine-induced ER-stress and apoptosis. METHODS: Xbp1s was overexpressed using adenoviruses and knocked down using small interference RNA in rat islet cells. In selected experiments, Xbp1 was also knocked down in FACS-purified rat beta cells and rat fibroblasts. Expression and production of XBP1s and key downstream genes and proteins was measured and beta cell function and viability were evaluated. RESULTS: Adenoviral-mediated overproduction of Xbp1s resulted in increased XBP1 activity and induction of several XBP1s target genes. Surprisingly, XBP1s overexpression impaired glucose-stimulated insulin secretion and increased beta cell apoptosis, whereas it protected fibroblasts against cell death induced by ER-stress. mRNA expression of Pdx1 and Mafa was inhibited in cells overproducing XBP1s, leading to decreased insulin expression. XBP1s knockdown partially restored cytokine/ER-stress-driven insulin and Pdx1 inhibition but had no effect on cytokine-induced ER-stress and apoptosis. CONCLUSIONS/INTERPRETATION: XBP1 has a distinct inhibitory role in beta cell as compared with other cell types. Prolonged XBP1s production hampers beta cell function via inhibition of insulin, Pdx1 and Mafa expression, eventually leading to beta cell apoptosis.


Assuntos
Apoptose/fisiologia , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição/metabolismo , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas de Ligação a DNA/genética , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Imunofluorescência , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Indóis/farmacologia , Insulina/genética , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-8/farmacologia , Fatores de Transcrição Maf/genética , Fatores de Transcrição Maf/metabolismo , Masculino , Interferência de RNA , RNA Interferente Pequeno , Ratos , Ratos Wistar , Fatores de Transcrição de Fator Regulador X , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Transfecção , Proteína 1 de Ligação a X-Box
9.
Diabetologia ; 48(8): 1541-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15986238

RESUMO

AIMS/HYPOTHESIS: Free radicals generated in mitochondria play a crucial role in the toxic effects of cytokines upon insulin-producing cells. This study therefore investigated the role of manganese superoxide dismutase (MnSOD) in cytokine-mediated toxicity in insulin-producing cells. METHODS: MnSOD was either stably overexpressed (MnSODsense) or stably suppressed (MnSODantisense) in insulin-producing RINm5F cells. Cell viability was quantified after incubation with different chemical reactive oxygen species (ROS) generators and with cytokines (IL-1beta alone or a mixture of IL-1beta, TNF-alpha and IFN-gamma). Additionally, cell proliferation and endogenous MnSOD protein expression were determined after exposure to cytokines. RESULTS: After incubation with hydrogen peroxide (H(2)O(2)) or hypoxanthine/xanthine oxidase no significant differences were observed in viability between control and MnSODsense or MnSODantisense clones. MnSOD overexpression reduced the viability of MnSODsense cells after exposure to the intracellular ROS generator menadione compared with control and MnSODantisense cells. MnSODsense cells also showed the highest susceptibility to cytokine toxicity with more than 75% loss of viability and a significant reduction of the proliferation rate after 72 h of incubation with a cytokine mixture. In comparison with control cells (67% viability loss), the reduction of viability in MnSODantisense cells was lower (50%), indicating a sensitising role of MnSOD in the progression of cytokine toxicity. The cell proliferation rate decreased in parallel to the reduction of cell viability. The MnSOD expression level after exposure to cytokines was also significantly lower in MnSODantisense cells than in control or MnSODsense cells. CONCLUSIONS/INTERPRETATION: The increase of the mitochondrial imbalance between the superoxide- and the H(2)O(2)-inactivating enzyme activities corresponds with a greater susceptibility to cytokines. Thus optimal antioxidative strategies to protect insulin-producing cells against cytokine toxicity may comprise a combined overexpression of H(2)O(2)-inactivating enzymes or suppression of MnSOD activity.


Assuntos
Citocinas/toxicidade , Insulina/biossíntese , Mitocôndrias/enzimologia , Espécies Reativas de Oxigênio/toxicidade , Superóxido Dismutase/fisiologia , Western Blotting , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA Complementar/biossíntese , DNA Complementar/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Interferon gama/toxicidade , Interleucina-1/toxicidade , Oxidantes/farmacologia , RNA Antissenso/biossíntese , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Sais de Tetrazólio , Tiazóis , Fator de Necrose Tumoral alfa/toxicidade , Vitamina K 3/farmacologia
10.
J Mol Endocrinol ; 31(3): 509-18, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14664711

RESUMO

Pancreatic beta-cells have low activities of the antioxidant enzyme catalase. Nitric oxide interacts with the haem group of catalase inhibiting its activity. We have studied the activity of catalase in beta-cells under conditions mimicking prediabetes and in which nitric oxide is generated from cytokine treatment in vitro. We also studied whether there is regulation of catalase enzyme activity by nitric oxide at the protein or gene expression level. RINm5F insulin-producing cells, treated for 24 h with cytokines, showed increased medium nitrite production (17+/-2.2 vs 0.3+/-0.2 pmol/ micro g protein) and significantly decreased cellular catalase activity (42.4+/-4.5%) compared with control cells. A similar reduction was seen in catalase-overexpressing RIN-CAT cells and in rat or human pancreatic islets of Langerhans. Catalase activity was also suppressed by the long-acting nitric oxide donor diethylenetriamine/nitric oxide adduct (Deta-NO) and this inhibition was reversible. The inhibition of catalase activity by cytokines in RINm5F cells was significantly reversed by the addition of the nitric oxide synthase 2 (NOS2) inhibitors nitro monomethylarginine or N-(3-(aminomethyl)benzyl)acetamidine (1400W). Protein expression was found to be unchanged in cytokine- or Deta-NO-treated RINm5F cells, while mRNA expression was marginally increased. We have shown that inhibition of catalase activity by cytokines is nitric oxide dependent and propose that this inhibition may confer increased susceptibility to cytokine- or nitric oxide-induced cell killing.


Assuntos
Catalase/metabolismo , Citocinas/farmacologia , Insulinoma/enzimologia , Óxido Nítrico/farmacologia , Neoplasias Pancreáticas/enzimologia , Amidinas/farmacologia , Animais , Benzilaminas/farmacologia , Morte Celular/fisiologia , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Insulina/metabolismo , Ilhotas Pancreáticas/enzimologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , RNA Mensageiro/genética , Ratos , Triazenos/farmacologia , Células Tumorais Cultivadas
11.
Diabetologia ; 44(9): 1189-96, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11596676

RESUMO

AIMS/HYPOTHESIS: We describe a new Type I (insulin-dependent) diabetes mellitus rat model (LEW.1AR1/Ztm-iddm) which arose through a spontaneous mutation in a congenic Lewis rat strain with a defined MHC haplotype (RT1.Aa B/Du Cu). METHODS: The development of diabetes was characterised using biochemical, immunological and morphological methods. RESULTS: Diabetes appeared in the rats with an incidence of 20 % without major sex preference at 58+/-2 days. The disease was characterised by hyperglycaemia, glycosuria, ketonuria and polyuria. In peripheral blood, the proportion of T lymphocytes was in the normal range expressing the RT6.1 differentiation antigen. Islets were heavily infiltrated with B and T lymphocytes, macrophages and NK cells with beta cells rapidly destroyed through apoptosis in areas of insulitis. CONCLUSION/INTERPRETATION: This Type I diabetic rat develops a spontaneous insulin-dependent autoimmune diabetes through beta cell apoptosis. It could prove to be a valuable new animal model for clarifying the mechanisms involved in the development of autoimmune diabetes.


Assuntos
Doenças Autoimunes , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Animais , Apoptose , Linfócitos B/patologia , Glicemia/análise , Diabetes Mellitus Tipo 1/metabolismo , Glicosúria , Hiperglicemia , Insulina/sangue , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Corpos Cetônicos/urina , Células Matadoras Naturais/patologia , Macrófagos/patologia , Masculino , Microscopia Eletrônica , Poliúria , Ratos , Ratos Endogâmicos Lew , Ratos Mutantes , Linfócitos T/patologia
12.
Diabetes ; 49(7): 1123-30, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10909968

RESUMO

Nitric oxide (NO) and reactive oxygen species (ROS) are crucial elements in cytokine-mediated beta-cell destruction. In insulin-producing RINm5F cells, overexpression of cytoprotective enzymes provides significant protection against the synergistic toxicity of NO and ROS. We therefore examined whether overexpression of catalase (Cat), glutathione peroxidase (Gpx), and Cu/Zn superoxide dismutase (SOD) can provide protection for bioengineered RINm5F cells against cytokine-mediated toxicity. A 72-h exposure of RINm5F control cells to interleukin-1beta (IL-1beta) alone or a combination of IL-1beta, tumor necrosis factor-alpha, and gamma-interferon resulted in a time- and concentration-dependent decrease of cell viability in the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) cytotoxicity assay. Although IL-1beta alone caused only a moderate reduction of viability in the range of 25%, the cytokine mixture induced a significant loss of viability of >75%. This increased toxicity of the cytokine mixture compared with that of IL-1beta alone could be explained by a higher rate of NO generation within the early 24-48 h incubation period that would favor the toxic synergism of NO and oxygen free radicals. Overexpression of Cat, Gpx, and Cu/Zn SOD protected against toxicity of the cytokine mixture but not against that of IL-1beta alone. The reduction of cytokine-mediated toxicity was evident also because of an increased proliferation rate and a drastic decrease in the cell death rate. The improved antioxidant defense status did not prevent the activation of iNOS after cytokine exposure. However, RINm5F cells overexpressing cytoprotective enzymes showed a significantly lower level of ROS-damaged protein residues. Thus, protection through Cat, Gpx, and Cu/Zn SOD overexpression was apparently because of an inactivation of ROS generated in the signal cascades of the cytokines. Overexpression of cytoprotective enzymes thus represents a feasible strategy to protect insulin-producing cells against cytokine-mediated cytotoxicity.


Assuntos
Catalase/genética , Citocinas/toxicidade , Regulação Enzimológica da Expressão Gênica , Glutationa Peroxidase/genética , Insulina/biossíntese , Ilhotas Pancreáticas/efeitos dos fármacos , Superóxido Dismutase/genética , Animais , Antioxidantes/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Interferon gama/toxicidade , Interleucina-1/toxicidade , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Cinética , Nitritos/metabolismo , Fator de Necrose Tumoral alfa/toxicidade
13.
Diabetologia ; 42(7): 849-55, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10440128

RESUMO

AIMS/HYPOTHESIS: The importance of different antioxidative enzymes for the defence of insulin-producing cells against the toxicity of nitric oxide (NO) was characterised in bioengineered RINm5F cells. METHODS: RINm5F insulin-producing cells stably overexpressing glutathione peroxidase (GPX), catalase (CAT) or Cu/Zn superoxide dismutase (SOD) were exposed to S-nitroso-N-acetyl-D,L-penicillamine (SNAP), sodium nitroprusside (SNP) and 3 morpholinosydnonimine (SIN-1), which generate both NO and reactive oxygen species, and to the polyamine/ NO, complex DETA/NO which generates NO alone. The viability of the cells was tested by the MTT assay. RESULTS: Overexpression of antioxidant enzymes provided significant protection against the toxicity of SNAP, SNP and SIN-1, with an individual specificity related to their chemical characteristics, but was without effect upon the toxicity of DETA/NO. Cells overexpressing GPX were well protected against SNP and SNAP, while CAT was most effective against SIN-1. SOD overexpression provided less protection against the toxicity of SNAP and SNP than overexpression of GPX but was more effective in protecting against SIN-1. Co-incubation of cells with NO donors and hydrogen peroxide or hypoxanthine and xanthine oxidase showed an overadditive synergism of toxicity. CONCLUSION/INTERPRETATION: The results emphasise the importance of a synergism between NO and reactive oxygen species for pancreatic beta-cell death. Such a synergism has also been observed after exposure of beta cells to cytokines. The component of the toxicity that is mediated by oxygen radicals can be suppressed effectively through overexpression of CAT, GPX or SOD or both.


Assuntos
Antioxidantes/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Doadores de Óxido Nítrico/toxicidade , Espécies Reativas de Oxigênio/fisiologia , Animais , Catalase/farmacologia , Citotoxicidade Imunológica , DEET/toxicidade , Sinergismo Farmacológico , Expressão Gênica , Glutationa Peroxidase/farmacologia , Peróxido de Hidrogênio/toxicidade , Hipoxantina/toxicidade , Molsidomina/análogos & derivados , Molsidomina/toxicidade , Nitroprussiato/toxicidade , Penicilamina/análogos & derivados , Penicilamina/toxicidade , S-Nitroso-N-Acetilpenicilamina , Superóxido Dismutase/farmacologia , Células Tumorais Cultivadas
14.
Diabetes ; 47(10): 1578-85, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9753295

RESUMO

To determine the importance of different antioxidative enzymes for the defense status of insulin-producing cells, the effects of stable overexpression of glutathione peroxidase (Gpx), catalase (Cat), or Cu/Zn superoxide dismutase (SOD) in insulin-producing RINm5F cells on the cytotoxicity of hydrogen peroxide (H2O2), hypoxanthine/xanthine oxidase (H/XO), and menadione have been investigated. Single overexpression of Cat or Gpx provided less protection than the combined expression of Cat plus SOD or Cat plus Gpx, while single overexpression of SOD either had no effect on the toxicity of the test compounds or increased it. RINm5F cells were also susceptible to butylalloxan, a lipophilic alloxan derivative that is selectively toxic to pancreatic beta-cells. Overexpression of enzymes, both alone and in combination, did not protect against butylalloxan-induced toxicity while SOD overexpression increased it, as evident from a half maximally effective concentration (EC50) value. The addition of Cat to the culture medium completely prevented the toxic effects of H2O2 and H/XO but had no significant effect on the toxicity of menadione or butylalloxan. Extracellular SOD had no effect on the toxicity of any of the test compounds. The results of this study show the importance of a combination of antioxidant enzymes in protecting against the toxicity of reactive oxygen species. Thus, overexpression of Cat and Gpx, alone or in combination with SOD, by use of molecular biology techniques can protect insulin-producing cells against oxidative damage. This may represent a strategy to protect pancreatic beta-cells against destruction during the development of autoimmune diabetes and emphasizes the importance of optimal antioxidative enzyme equipment for protection against free radical-mediated diseases.


Assuntos
Catalase/genética , Expressão Gênica , Glutationa Peroxidase/genética , Ilhotas Pancreáticas/enzimologia , Espécies Reativas de Oxigênio , Superóxido Dismutase/genética , Aloxano/toxicidade , Animais , Antioxidantes/metabolismo , Catalase/metabolismo , Catalase/farmacologia , Glutationa Peroxidase/metabolismo , Glutationa Peroxidase/farmacologia , Peróxido de Hidrogênio/toxicidade , Insulina/biossíntese , Ilhotas Pancreáticas/efeitos dos fármacos , Ratos , Superóxido Dismutase/metabolismo , Superóxido Dismutase/farmacologia , Transfecção , Células Tumorais Cultivadas , Vitamina K/toxicidade , Xantina/toxicidade , Xantina Oxidase/toxicidade
15.
Diabetes ; 46(11): 1733-42, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9356019

RESUMO

Antioxidant enzyme expression was determined in rat pancreatic islets and RINm5F insulin-producing cells on the level of mRNA, protein, and enzyme activity in comparison with 11 other rat tissues. Although superoxide dismutase expression was in the range of 30% of the liver values, the expression of the hydrogen peroxide-inactivating enzymes catalase and glutathione peroxidase was extremely low, in the range of 5% of the liver. Pancreatic islets but not RINm5F cells expressed an additional phospholipid hydroperoxide glutathione peroxidase that exerted protective effects against lipid peroxidation of the plasma membrane. Regression analysis for mRNA and protein expression and enzyme activities from 12 rat tissues revealed that the mRNA levels determine the enzyme activities of the tissues. The induction of cellular stress by high glucose, high oxygen, and heat shock treatment did not affect antioxidant enzyme expression in rat pancreatic islets or in RINm5F cells. Thus insulin-producing cells cannot adapt the low antioxidant enzyme activity levels to typical situations of cellular stress by an upregulation of gene expression. Through stable transfection, however, we were able to increase catalase and glutathione peroxidase gene expression in RINm5F cells, resulting in enzyme activities more than 100-fold higher than in nontransfected controls. Catalase-transfected RINm5F cells showed a 10-fold greater resistance toward hydrogen peroxide toxicity, whereas glutathione peroxidase overexpression was much less effective. Thus inactivation of hydrogen peroxide through catalase seems to be a step of critical importance for the removal of reactive oxygen species in insulin-producing cells. Overexpression of catalase may therefore be an effective means of preventing the toxic action of reactive oxygen species.


Assuntos
Antioxidantes/metabolismo , Catalase/biossíntese , Glutationa Peroxidase/biossíntese , Ilhotas Pancreáticas/enzimologia , Superóxido Dismutase/biossíntese , Transcrição Gênica , Animais , Catalase/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Isoenzimas/biossíntese , Isoenzimas/metabolismo , Cinética , Masculino , Especificidade de Órgãos , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Análise de Regressão , Superóxido Dismutase/metabolismo , Transfecção
16.
Biochem Pharmacol ; 51(7): 893-6, 1996 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-8651938

RESUMO

The effect of the antihyperglycaemic agent metformin was studied on gene expression of the energy-dependent sodium-hexose cotransporter (SGLT1) and the facilitative hexose transporters GLUT2 and GLUT5 in rat intestine. Metformin treatment (125 mg/kg body wt. twice daily for a period of 3 days) significantly increased SGLT1 gene expression in duodenum and jejunum. GLUT5 gene expression was increased by metformin treatment only in the jejunum. Gene expression of GLUT2 in the intestine was not significantly affected by metformin treatment. This increase in transporter gene expression offers the potential for increases in hexose uptake at the brush border membrane, and may compensate for other effects of the drug that have been suggested to decrease glucose uptake by SGLT1, as well as for metformin stimulation of glucose utilization by the intestinal mucosa.


Assuntos
Intestino Delgado/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Metformina/farmacologia , Proteínas de Transporte de Monossacarídeos/efeitos dos fármacos , Proteínas de Transporte de Monossacarídeos/metabolismo , Animais , Expressão Gênica/efeitos dos fármacos , Transportador de Glucose Tipo 2 , Transportador de Glucose Tipo 5 , Masculino , Ratos , Ratos Wistar , Transportador 1 de Glucose-Sódio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...