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.
Int J Mol Sci ; 23(7)2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35409280

RESUMO

The transcription factor, early growth response-1 (EGR-1), is involved in the regulation of cell differentiation, proliferation, and apoptosis in response to different stimuli. EGR-1 is described to be involved in pancreatic endoderm differentiation, but the regulatory mechanisms controlling its action are not fully elucidated. Our previous investigation reported that exposure of mouse embryonic stem cells (mESCs) to the chemical nitric oxide (NO) donor diethylenetriamine nitric oxide adduct (DETA-NO) induces the expression of early differentiation genes such as pancreatic and duodenal homeobox 1 (Pdx1). We have also evidenced that Pdx1 expression is associated with the release of polycomb repressive complex 2 (PRC2) and P300 from the Pdx1 promoter; these events were accompanied by epigenetic changes to histones and site-specific changes in the DNA methylation. Here, we investigate the role of EGR-1 on Pdx1 regulation in mESCs. This study reveals that EGR-1 plays a negative role in Pdx1 expression and shows that the binding capacity of EGR-1 to the Pdx1 promoter depends on the methylation level of its DNA binding site and its acetylation state. These results suggest that targeting EGR-1 at early differentiation stages might be relevant for directing pluripotent cells into Pdx1-dependent cell lineages.


Assuntos
Endoderma , Células-Tronco Embrionárias Murinas , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias , Endoderma/metabolismo , Camundongos , Óxido Nítrico/metabolismo
2.
Lancet Reg Health Am ; 15: 100347, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36778067

RESUMO

Background: Malaria is one of the biggest impediments to global progress. In Peru, it is still a major public health problem. Measures of health and economic burden due to malaria are relevant considerations for the assessment of current policies. Methods: We used estimates from the Global Burden of Diseases Study 2019 for malaria in Peru, grouped by gender and age, from 1990 to 2019. Results are presented as absolute numbers and age-standardized rates with 95% uncertainty intervals (UI). We collected economic data from the World Bank and The National Institute of Statistics and Informatics of Peru and Loreto to calculate the economic burden of productivity loss (EBPL) using the human capital approach. Economic values were presented in constant dollars, soles, and percentages. Findings: Rates of deaths, years of life lost (YLLs), years lived with disability (YLDs), and disability-adjusted life years (DALYs), as well as the EBPL, were drastically reduced from 1990 to 2019. DALYs had a greater percentage of YLDs in 2019 than in 1990. DALYs rates showed no preference between sexes, but the "< 1 year" age group had the highest DALYs values over the study period. We found that the EBPL due to malaria for Loreto was considerably higher than Peru's in terms of GDP percentage. Interpretation: Our study shows that the fight against malaria in Peru reduced remarkably the impact of the disease since 1990; however, during the last decade the estimates were stable or even increased. Our results help to measure the malaria impact on the health status of the Peruvian population as well as the economic pressure that it exerts, constituting remarkable tools for policymaking aimed at reducing the burden of this disease. Strengthening the malaria elimination program is important to achieve the elimination of the disease in the coming years. Funding: This study was supported by the Universidad Nacional Toribio Rodríguez de Mendoza and FONDECYT: Contrato Nº 09-2019-FONDECYT-BMINC.INV and FONDECYT-BM, Perú (Program INCORPORACIÓN DE INVESTIGADORES E038-2019-01, Registry Number: 64007).

3.
Parasitol Int ; 85: 102428, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34329752

RESUMO

Fascioliasis is a zoonotic disease caused by parasites of the genus Fasciola spp. which cause an important loss to the livestock industry. The objectives of this study were: to estimate the prevalence of fascioliasis in three provinces of Amazonas, to evaluate possible risk factors of infection in cattle and to genetically characterize the Fasciola haplotypes circulating in this area. According to the results the prevalence of fascioliasis in cattle was 90.13% (712/790). Odds ratio results showed a significant association between fascioliasis and the Brown Swiss breed (OR = 2.62; 95% CI: 1.57-4.35; p < 0.001), and with female cattle older than 30 months (OR = 1.71; 95% CI: 1.05-2.79; p < 0.031). According to the molecular genetic studies using the gene marker NAD1, six haplotypes of Fasciola hepatica were found in the 35 infected livers collected. The results obtained in this study are concerning due to the high prevalence presented and it reveals the necessity of a continuing monitoring because of the high risk of transmission to humans.


Assuntos
Doenças dos Bovinos/epidemiologia , Fasciolíase/veterinária , Animais , Bovinos , Doenças dos Bovinos/parasitologia , Fasciolíase/epidemiologia , Fasciolíase/parasitologia , Feminino , Masculino , Peru/epidemiologia , Prevalência , Fatores de Risco
4.
ACS Omega ; 5(32): 20270-20282, 2020 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-32832780

RESUMO

Pluripotent stem cells maintain the property of self-renewal and differentiate into all cell types under clear environments. Though the gene regulatory mechanism for pluripotency has been investigated in recent years, it is still not completely understood. Here, we show several signaling pathways involved in the maintenance of pluripotency. To investigate whether AMPK is involved in maintaining the pluripotency in mouse embryonic stem cells (mESCs) and elucidating the possible molecular mechanisms, implicated D3 and R1/E mESC lines were used in this study. Cells were cultured in the absence or presence of LIF and treated with 1 mM and 0.5 mM 5-aminoimidazole-4-carboxamide-1-ß-d-ribofuranoside (AICAR), 2 mM metformin, compound C, and the PI3K inhibitor LY294002 for 24, 72, and 120 h. The levels of Nanog, Oct3/4, and REX1 and Brachyury, Notch2, and Gata4 mRNAs and Nanog or OCT3/4 protein levels were analyzed. Alkaline phosphatase and the cellular cycle were determined. The pGSK3ß, GSK3ß, p-ß-catenin, and ß-catenin protein levels were also investigated. We found that AMPK activators such as AICAR and metformin increase mRNA expression of pluripotency markers and decrease mRNA expression of differentiation markers in R1/E and D3 ES cells. AICAR increases phosphatase activity and arrests the cellular cycle in the G1 phase in these cells. We describe that AICAR effects were mediated by AMPK activation using a chemical inhibitor or by silencing this gene. AICAR effects were also mediated by PI3K, GSK3ß, and ß-catenin in R1/E ES cells. According to our findings, we provide a mechanism by which AICAR increases and maintains a pluripotency state through enhanced Nanog expression, involving AMPK/PI3K and p-GSK3ß Ser21/9 pathways backing up the AICAR function as a potential target for this drug controlling pluripotency. The highlights of this study are that AICAR (5-aminoimidazole-4-carboxamied-1-b-riboside), an AMP protein kinase (AMPK) activator, blocks the ESC differentiation and AMPK is a key enzyme for pluripotency and shows valuable data to clarify the molecular pluripotency mechanism.

5.
Front Immunol ; 11: 609961, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33633730

RESUMO

Snakebite envenoming is a global neglected disease with an incidence of up to 2.7 million new cases every year. Although antivenoms are so-far the most effective treatment to reverse the acute systemic effects induced by snakebite envenoming, they have a limited therapeutic potential, being unable to completely neutralize the local venom effects. Local damage, such as dermonecrosis and myonecrosis, can lead to permanent sequelae with physical, social, and psychological implications. The strong inflammatory process induced by snake venoms is associated with poor tissue regeneration, in particular the lack of or reduced skeletal muscle regeneration. Mesenchymal stromal cells (MSCs)-based therapies have shown both anti-inflammatory and pro-regenerative properties. We postulate that using allogeneic MSCs or their cell-free products can induce skeletal muscle regeneration in snakebite victims, improving all the three steps of the skeletal muscle regeneration process, mainly by anti-inflammatory activity, paracrine effects, neovascularization induction, and inhibition of tissue damage, instrumental for microenvironment remodeling and regeneration. Since snakebite envenoming occurs mainly in areas with poor healthcare, we enlist the principles and potential of MSCs-based therapies and discuss regulatory issues, good manufacturing practices, transportation, storage, and related-procedures that could allow the administration of these therapies, looking forward to a safe and cost-effective treatment for a so far unsolved and neglected health problem.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Desenvolvimento Muscular , Músculo Esquelético/fisiopatologia , Regeneração , Mordeduras de Serpentes/cirurgia , Animais , Humanos , Mediadores da Inflamação/metabolismo , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Necrose , Fenótipo , Transdução de Sinais , Mordeduras de Serpentes/diagnóstico , Mordeduras de Serpentes/metabolismo , Mordeduras de Serpentes/fisiopatologia , Resultado do Tratamento
6.
World J Stem Cells ; 9(2): 26-36, 2017 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-28289506

RESUMO

Mitochondrial dysfunction and endoplasmic reticulum stress (ERS) are global processes that are interrelated and regulated by several stress factors. Nitric oxide (NO) is a multifunctional biomolecule with many varieties of physiological and pathological functions, such as the regulation of cytochrome c inhibition and activation of the immune response, ERS and DNA damage; these actions are dose-dependent. It has been reported that in embryonic stem cells, NO has a dual role, controlling differentiation, survival and pluripotency, but the molecular mechanisms by which it modulates these functions are not yet known. Low levels of NO maintain pluripotency and induce mitochondrial biogenesis. It is well established that NO disrupts the mitochondrial respiratory chain and causes changes in mitochondrial Ca2+ flux that induce ERS. Thus, at high concentrations, NO becomes a potential differentiation agent due to the relationship between ERS and the unfolded protein response in many differentiated cell lines. Nevertheless, many studies have demonstrated the need for physiological levels of NO for a proper ERS response. In this review, we stress the importance of the relationships between NO levels, ERS and mitochondrial dysfunction that control stem cell fate as a new approach to possible cell therapy strategies.

7.
J Cell Biochem ; 117(9): 2078-88, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26853909

RESUMO

Nitric oxide (NO) delays mouse embryonic stem cell (mESC) differentiation by regulating genes linked to pluripotency and differentiation. Nevertheless, no profound study has been conducted on cell differentiation regulation by this molecule through signaling on essential biological functions. We sought to demonstrate that NO positively regulates the pluripotency transcriptional core, enforcing changes in the chromatin structure, in addition to regulating cell proliferation, and signaling pathways with key roles in stemness. Culturing mESCs with 2 µM of the NO donor diethylenetriamine/NO (DETA/NO) in the absence of leukemia inhibitory factor (LIF) induced significant changes in the expression of 16 genes of the pluripotency transcriptional core. Furthermore, treatment with DETA/NO resulted in a high occupancy of activating H3K4me3 at the Oct4 and Nanog promoters and repressive H3K9me3 and H3k27me3 at the Brachyury promoter. Additionally, the activation of signaling pathways involved in pluripotency, such as Gsk3-ß/ß-catenin, was observed, in addition to activation of PI3 K/Akt, which is consistent with the protection of mESCs from cell death. Finally, a decrease in cell proliferation coincides with cell cycle arrest in G2/M. Our results provide novel insights into NO-mediated gene regulation and cell proliferation and suggest that NO is necessary but not sufficient for the maintenance of pluripotency and the prevention of cell differentiation. J. Cell. Biochem. 117: 2078-2088, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular/fisiologia , Pontos de Checagem da Fase G2 do Ciclo Celular/fisiologia , Expressão Gênica/fisiologia , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Células-Tronco Embrionárias Murinas/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triazenos/farmacologia
8.
World J Stem Cells ; 7(3): 605-17, 2015 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-25914767

RESUMO

Stem cell pluripotency and differentiation are global processes regulated by several pathways that have been studied intensively over recent years. Nitric oxide (NO) is an important molecule that affects gene expression at the level of transcription and translation and regulates cell survival and proliferation in diverse cell types. In embryonic stem cells NO has a dual role, controlling differentiation and survival, but the molecular mechanisms by which it modulates these functions are not completely defined. NO is a physiological regulator of cell respiration through the inhibition of cytochrome c oxidase. Many researchers have been examining the role that NO plays in other aspects of metabolism such as the cellular bioenergetics state, the hypoxia response and the relationship of these areas to stem cell stemness.

9.
Redox Biol ; 5: 417-418, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-28162281

RESUMO

The expansion of pluripotent cells (ESCs and iPSCs) under conditions that maintain their pluripotency is necessary to implement a cell therapy program. Previously, we have described that low nitric oxide (NO) donor diethylenetriamine/nitric oxide adduct (DETA-NO) added to the culture medium, promote the expansion of these cell types. The molecular mechanisms are not yet known. We present evidences that ESC and iPSCs in normoxia in presence of low NO triggers a similar response to hypoxia, thus maintaining the pluripotency. We have studied the stability of HIF-1α (Hypoxia Inducible Factor) in presence of low NO. Because of the close relationship between hypoxia, metabolism, mitochondrial function and pluripotency we have analyzed by q RT-PCR the expression of genes involved in the glucose metabolism such as: HK2, LDHA and PDK1; besides other HIF-1α target gene. We further analyzed the expression of genes involved in mitochondrial biogenesis such as PGC1α, TFAM and NRF1 and we have observed that low NO maintains the same pattern of expression that in hypoxia. The study of the mitochondrial membrane potential using Mito-Tracker dye showed that NO decrease the mitochondrial function. We will analyze other metabolic parameters, to determinate if low NO regulates mitochondrial function and mimics Hypoxia Response. The knowledge of the role of NO in the Hypoxia Response and the mechanism that helps to maintain self-renewal in pluripotent cells in normoxia, can help to the design of culture media where NO could be optimal for stem cell expansion in the performance of future cell therapies.


Assuntos
Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Óxido Nítrico/metabolismo , Animais , Hipóxia Celular/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Mitocôndrias/metabolismo
10.
Stem Cells Int ; 2014: 379678, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25544848

RESUMO

The function of pluripotency genes in differentiation is a matter of investigation. We report here that Nanog and Oct4 are reexpressed in two mouse embryonic stem cell (mESC) lines following exposure to the differentiating agent DETA/NO. Both cell lines express a battery of both endoderm and mesoderm markers following induction of differentiation with DETA/NO-based protocols. Confocal analysis of cells undergoing directed differentiation shows that the majority of cells expressing Nanog express also endoderm genes such as Gata4 and FoxA2 (75.4% and 96.2%, resp.). Simultaneously, mRNA of mesodermal markers Flk1 and Mef2c are also regulated by the treatment. Acetylated histone H3 occupancy at the promoter of Nanog is involved in the process of reexpression. Furthermore, Nanog binding to the promoter of Brachyury leads to repression of this gene, thus disrupting mesendoderm transition.

11.
Islets ; 6(5-6): e995997, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25658244

RESUMO

Homeostatic levels of nitric oxide (NO) protect efficiently against apoptotic death in both human and rodent pancreatic ß cells, but the protein profile of this action remains to be determined. We have applied a 2 dimensional LC-MS-MALDI-TOF/TOF-based analysis to study the impact of protective NO in rat insulin-producing RINm5F cell line and in mouse and human pancreatic islets (HPI) exposed to serum deprivation condition. 24 proteins in RINm5F and 22 in HPI were identified to undergo changes in at least one experimental condition. These include stress response mitochondrial proteins (UQCRC2, VDAC1, ATP5C1, ATP5A1) in RINm5F cells and stress response endoplasmic reticulum proteins (HSPA5, PDIA6, VCP, GANAB) in HPI. In addition, metabolic and structural proteins, oxidoreductases and chaperones related with protein metabolism are also regulated by NO treatment. Network analysis of differentially expressed proteins shows their interaction in glucocorticoid receptor and NRF2-mediated oxidative stress response pathways and eNOS signaling. The results indicate that exposure to exogenous NO counteracts the impact of serum deprivation on pancreatic ß cell proteome. Species differences in the proteins involved are apparent.


Assuntos
Ilhotas Pancreáticas/efeitos dos fármacos , Óxido Nítrico/farmacologia , Adenosina Trifosfatases/análise , Animais , Proteínas de Ciclo Celular/análise , Linhagem Celular , Complexo III da Cadeia de Transporte de Elétrons/análise , Chaperona BiP do Retículo Endoplasmático , Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico/análise , Humanos , Células Secretoras de Insulina/química , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/citologia , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/análise , Proteômica , Ratos , Proteína com Valosina , Canal de Ânion 1 Dependente de Voltagem/análise
12.
Islets ; 4(2): 108-18, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22614339

RESUMO

The reduction of pancreatic ß-cell mass is an important factor in the development of type 1 and type 2 diabetes. Understanding the mechanisms that regulate the maintenance of pancreatic ß-cell mass as well as ß-cell death is necessary for the establishment of therapeutic strategies. In this context, nitric oxide (NO) is a diatomic, gaseous, highly reactive molecule with biological activity that participates in the regulation of pancreatic ß-cell mass. Two types of cellular responses can be distinguished depending on the level of NO production. First, pancreatic ß-cells exposed to inflammatory cytokines, lipid stress or hyperglycaemia produce high concentrations of NO, mainly due to the activation of inducible NO synthase (iNOS), thus promoting cell death. Meanwhile, under homeostatic conditions, low concentrations of NO, constitutively produced by endothelial NO synthase (eNOS), promote cell survival. Here, we will discuss the current knowledge of the NO-dependent mechanisms activated during cellular responses, emphasizing those related to the regulation of cell survival.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Óxido Nítrico/metabolismo , Animais , Sobrevivência Celular/fisiologia , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Humanos , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Óxido Nítrico Sintase Tipo II/metabolismo
13.
Cell Signal ; 20(2): 301-10, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18023142

RESUMO

Generation of low levels of nitric oxide (NO) contributes to beta cell survival in vitro. The purpose of this study was to explore the link between NO and the survival pathway triggered by insulin-like growth factor-1 (IGF-1) and insulin in insulin producing RINm5F cells and in pancreatic islets. Results show that exposure of cells to IGF-1/insulin protects against serum deprivation-induced apoptosis. This action is prevented with inhibitors of NO generation, PI3K and Akt. Moreover, transfection with the negative dominant form of the tyrosine kinase c-Src abrogates the effect of IGF-1 and insulin on DNA fragmentation. An increase in the expression level of NOS3 protein and in the enzyme activity is observed following exposure of serum-deprived RINm5F cells to IGF-1 and insulin. Phosphorylation of IRS-1, IRS-2 and to less extent IRS-3 takes place when serum-deprived RINm5F cells and rat pancreatic islets are exposed to either IGF-1, insulin, or diethylenetriamine nitric oxide adduct (DETA/NO). In human islets, IRS-1 and IRS-2 proteins are present and tyrosine phosphorylated upon exposure to IGF-1, insulin and DETA/NO. Both rat and human pancreatic islets undergo DNA fragmentation when cultured in serum-free medium and IGF-1, insulin and DETA/NO protect efficiently from this damage. We then conclude that generation of NO participates in the activation of survival pathways by IGF-1 and insulin in beta cells.


Assuntos
Fator de Crescimento Insulin-Like I/farmacologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/farmacologia , Óxido Nítrico/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , DEET/farmacologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Proteínas Substratos do Receptor de Insulina , Células Secretoras de Insulina/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Ratos , Ratos Wistar , Soro
14.
Endocrinology ; 145(5): 2319-27, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14764634

RESUMO

Mechanisms involved in the protective action of nitric oxide (NO) in insulin-producing cells are a matter of debate. We have previously shown that pharmacological inhibition of c-Src cancels the antiapoptotic action of low and sustained concentrations of exogenous NO. In this study, using insulin-producing RINm5F cells that overexpress Src either permanently active (v-Src) or dominant negative (dn-Src) forms, we determine that this tyrosine kinase is the principal mediator of the protective action of NO. We also show that Src-directed activation of insulin receptor substrate-1, phosphatidylinositol 3-kinase (PI3K), Akt, and Bad phosphorylation conform a substantial component of the survival route because pharmacological inhibition of PI3K and Akt canceled the antiapoptotic effects of NO. Studies performed with the protein kinase G (PKG) inhibitor KT-5823 revealed that NO-dependent activation of c-Src/ insulin receptor substrate-1 is not affected by PKG activation. By contrast, Akt and Bad activation are partially dependent on PKG activation. Endogenous production of NO after overexpression of endothelial nitric oxide synthase in RINm5F cells mimics the effects produced by generation of low amounts of NO from exogenous diethylenetriamine/NO. In addition, we found that NO produces c-Src/PI3K- and PKG-dependent activation of ERK 1/2. The MAPK kinase inhibitor PD 98059 suppresses NO-dependent protection from DNA fragmentation induced by serum deprivation. The protective action of low and sustained concentration of NO is also observed in staurosporine- and Taxol-induced apoptosis. Finally, NO also protects isolated rat islets from DNA fragmentation induced by serum deprivation. These data strengthen the notion that NO production at physiological levels plays a role in protection from apoptosis in pancreatic beta-cells.


Assuntos
Ilhotas Pancreáticas/metabolismo , Óxido Nítrico/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Proteína Tirosina Quinase CSK , Linhagem Celular , Sobrevivência Celular , Meios de Cultura Livres de Soro , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Proteínas Substratos do Receptor de Insulina , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo III , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Wistar , Quinases da Família src
15.
Exp Cell Res ; 293(1): 22-30, 2004 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-14729054

RESUMO

Generation of high levels of nitric oxide (NO) following induction of NOS2 by interleukin-1 beta (IL-1beta) triggers beta cell apoptosis in insulin-secreting RINm5F cells. Mitochondrial and nuclear events such as downregulation of the antiapoptotic protein Bcl-2, activation of the pore responsible for the permeability transition (PT) and DNA fragmentation are involved in the process. We report in the present paper that exposure of insulin-producing RINm5F cells to NO donors and to IL-1beta leads to oxidative carbonylation of both Bcl-2 and the adenine nucleotide translocator (ANT) component of the mitochondrial PT pore. When the effect of endogenous generation of high concentrations of NO following exposure of cells to IL-1beta was studied, carbonylation of Bcl-2 preceded downregulation of the protein. Overexpression of Mn-SOD decreases substantially the extent of Bcl-2 carbonylation in SIN-1-exposed cells. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) inhibition, carbonylation and translocation from cytoplasm to nucleus and DNA fragmentation were also induced by DETA/NO exposure. DETA/NO-induced carbonylation of Bcl-2 and ANT proteins takes place 6 h before apoptotic release of histone-associated DNA to cytoplasm. Time course studies also reveal a close parallel between GAPDH translocation to nucleus and carbonylation. Inhibitors of lipooxidation end products formation such as piridoxamine (PM) and aminoguanidine (AG) block NO-triggered carbonylation of Bcl-2, ANT and GAPDH, prevent NO-induced GAPDH enzyme inhibition and nuclear translocation and DNA fragmentation. Our results support the notion that the oxidative carbonylation of proteins plays a role in the control of NO-induced apoptosis.


Assuntos
Nucleotídeos de Adenina/metabolismo , Apoptose/efeitos dos fármacos , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Molsidomina/análogos & derivados , Óxido Nítrico/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , DEET/farmacologia , Sequestradores de Radicais Livres , Gliceraldeído-3-Fosfato Desidrogenases/antagonistas & inibidores , Gliceraldeído-3-Fosfato Desidrogenases/efeitos dos fármacos , Insulina/metabolismo , Secreção de Insulina , Interleucina-1/farmacologia , Cinética , Peroxidação de Lipídeos/efeitos dos fármacos , Molsidomina/farmacologia , Doadores de Óxido Nítrico/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Ratos , Superóxido Dismutase/metabolismo
16.
Biochem Pharmacol ; 66(10): 1963-71, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14599554

RESUMO

We have explored the impact of nitric oxide (NO) exposure on oxidation damage of lipids, and proteins, and the contribution of this type of damage to the activation of the apoptotic program in insulin secreting RINm5F cells. Exposure of cells to NO donors and to interleukin-1 beta (IL-1beta) led to generation of lipooxidation products such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE). Addition of superoxide dismutase (SOD) and catalase (Cat) to cells decreased by 50% MDA and 4-HNE production induced by IL-1beta. Over-expression of Mn-SOD in cells conferred a remarkable decrease (75%) in IL-1beta-induced lipid peroxidation. These data suggest that peroxynitrite (ONOO(-)) mediates peroxidative damage to lipids in this cell system. Inhibitors of advanced lipooxidation end products (ALEs) formation such as aminoguanidine (AG) and pyridoxamine (PM) prevented partially apoptotic events triggered by NO such as DNA fragmentation, caspase-3 activation and cytochrome c release from mitochondria. These findings indicate that ALEs are involved in NO-induced apoptosis. In fact, NO-induced carbonylation of PARP protein preceded its apoptotic degradation and inhibitors of ALEs formation prevented both events. We thus propose that carbonylation of proteins is instrumental in linking NO-dependent lipid oxidation and apoptosis in this cell system.


Assuntos
Apoptose , Peroxidação de Lipídeos/efeitos dos fármacos , Óxido Nítrico/farmacologia , Proteínas/metabolismo , Animais , Células Cultivadas , Fragmentação do DNA/efeitos dos fármacos , Insulina/metabolismo , Secreção de Insulina , Interleucina-1/farmacologia , L-Lactato Desidrogenase/metabolismo , Oxirredução/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases , Ratos , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...