Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 36(28): 4087, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28288137

RESUMO

This corrects the article DOI: 10.1038/onc.2014.355.

2.
Redox Biol ; 12: 129-138, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28231483

RESUMO

The signaling of reactive oxygen species (ROS) is essential for the maintenance of normal cellular function. However, whether and how ROS regulate stem cells are unclear. Here, we demonstrate that, in transgenic mice expressing the human manganese superoxide dismutase (MnSOD) gene, a scavenger of ROS in mitochondria, the number and function of mouse hematopoietic stem/progenitor cells (HSPC) under physiological conditions are enhanced. Importantly, giving MnTnBuOE-2-PyP5+(MnP), a redox- active MnSOD mimetic, to mouse primary bone marrow cells or to C57B/L6 mice significantly enhances the number of HSPCs. Mechanistically, MnP reduces superoxide to hydrogen peroxide, which activates intracellular Nrf2 signaling leading to the induction of antioxidant enzymes, including MnSOD and catalase, and mitochondrial uncoupling protein 3. The results reveal a novel role of ROS signaling in regulating stem cell function, and suggest a possible beneficial effect of MnP in treating pathological bone marrow cell loss and in increasing stem cell population for bone marrow transplantation.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Metaloporfirinas/farmacologia , Superóxido Dismutase/metabolismo , Animais , Células Cultivadas , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Superóxido Dismutase/genética
3.
Oncogene ; 34(41): 5229-39, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25659582

RESUMO

Epithelial-mesenchymal transition (EMT) promotes cancer cell invasion, metastasis and treatment failure. EMT may be activated in cancer cells by reactive oxygen species (ROS). EMT may promote conversion of a subset of cancer cells from a CD44(low)-CD24(high) (CD44L) epithelial phenotype to a CD44(high)-CD24(-/low) (CD44H) mesenchymal phenotype, the latter associated with increased malignant properties of cancer cells. ROS are required for cells undergoing EMT, although excessive ROS may induce cell death or senescence; however, little is known as to how cellular antioxidant capabilities may be regulated during EMT. Mitochondrial superoxide dismutase 2 (SOD2) is frequently overexpressed in oral and esophageal cancers. Here, we investigate mechanisms of SOD2 transcriptional regulation in EMT, as well as the functional role of this antioxidant in EMT. Using well-characterized genetically engineered oral and esophageal human epithelial cell lines coupled with RNA interference and flow cytometric approaches, we find that transforming growth factor (TGF)-ß stimulates EMT, resulting in conversion of CD44L to CD44H cells, the latter of which display SOD2 upregulation. SOD2 induction in transformed keratinocytes was concurrent with suppression of TGF-ß-mediated induction of both ROS and senescence. SOD2 gene expression appeared to be transcriptionally regulated by NF-κB and ZEB2, but not ZEB1. Moreover, SOD2-mediated antioxidant activity may restrict conversion of CD44L cells to CD44H cells at the early stages of EMT. These data provide novel mechanistic insights into the dynamic expression of SOD2 during EMT. In addition, we delineate a functional role for SOD2 in EMT via the influence of this antioxidant upon distinct CD44L and CD44H subsets of cancer cells that have been implicated in oral and esophageal tumor biology.


Assuntos
Transição Epitelial-Mesenquimal , Superóxido Dismutase/fisiologia , Linhagem Celular , Regulação Enzimológica da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Receptores de Hialuronatos , Mitocôndrias/enzimologia , NF-kappa B/metabolismo , Proteínas Repressoras/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco
4.
Oncogene ; 34(32): 4229-37, 2015 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-25362851

RESUMO

Manganese superoxide dismutase (MnSOD) is a mitochondrially localized primary antioxidant enzyme, known to be essential for the survival of aerobic life and to have important roles in tumorigenesis. Here, we show that MnSOD deficiency in skin tissues of MnSOD-heterozygous knockout (Sod2(+/-)) mice leads to increased expresson of uncoupling proteins (UCPs). When MnSOD is deficient, superoxide radical and its resulting reactive oxygen species (ROS) activate ligand binding to peroxisome proliferator-activated receptor alpha (PPARα), suggesting that the activation of PPARα signaling is a major mechanism underlying MnSOD-dependent UCPs expression that consequently triggers the PI3K/Akt/mTOR pathway, leading to increased aerobic glycolysis. Knockdown of UCPs and mTOR suppresses lactate production and increases ATP levels, suggesting that UCPs contribute to increased glycolysis. These results highlight the existence of a free radical-mediated mechanism that activates mitochondria uncoupling to reduce ROS production, which precedes the glycolytic adaptation described as the Warburg Effect.


Assuntos
Glicólise , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Superóxido Dismutase/deficiência , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Células Cultivadas , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Lactatos/metabolismo , Camundongos Knockout , Mitocôndrias/genética , Proteínas Mitocondriais/genética , PPAR alfa/genética , PPAR alfa/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Superóxido Dismutase/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteína Desacopladora 1 , Proteína Desacopladora 2
5.
Free Radic Biol Med ; 72: 55-65, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24632380

RESUMO

Doxorubicin (DOX), one of the most effective anticancer drugs, is known to generate progressive cardiac damage, which is due, in part, to DOX-induced reactive oxygen species (ROS). The elevated ROS often induce oxidative protein modifications that result in alteration of protein functions. This study demonstrates that the level of proteins adducted by 4-hydroxy-2-nonenal (HNE), a lipid peroxidation product, is significantly increased in mouse heart mitochondria after DOX treatment. A redox proteomics method involving two-dimensional electrophoresis followed by mass spectrometry and investigation of protein databases identified several HNE-modified mitochondrial proteins, which were verified by HNE-specific immunoprecipitation in cardiac mitochondria from the DOX-treated mice. The majority of the identified proteins are related to mitochondrial energy metabolism. These include proteins in the citric acid cycle and electron transport chain. The enzymatic activities of the HNE-adducted proteins were significantly reduced in DOX-treated mice. Consistent with the decline in the function of the HNE-adducted proteins, the respiratory function of cardiac mitochondria as determined by oxygen consumption rate was also significantly reduced after DOX treatment. Treatment with Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, an SOD mimic, averted the doxorubicin-induced mitochondrial dysfunctions as well as the HNE-protein adductions. Together, the results demonstrate that free radical-mediated alteration of energy metabolism is an important mechanism mediating DOX-induced cardiac injury, suggesting that metabolic intervention may represent a novel approach to preventing cardiac injury after chemotherapy.


Assuntos
Aldeídos/metabolismo , Antibióticos Antineoplásicos/toxicidade , Doxorrubicina/toxicidade , Metabolismo Energético/efeitos dos fármacos , Mitocôndrias Cardíacas/efeitos dos fármacos , Animais , Eletroforese em Gel Bidimensional , Immunoblotting , Imunoprecipitação , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/metabolismo , Oxirredução , Proteômica
6.
Oncogene ; 31(17): 2129-39, 2012 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21909133

RESUMO

Manganese superoxide dismutase is a nuclear encoded primary antioxidant enzyme localized exclusively in the mitochondrial matrix. Genotoxic agents, such as ultraviolet (UV) radiation, generates oxidative stress and cause mitochondrial DNA (mtDNA) damage. The mtDNA polymerase (Polγ), a major constituent of nucleoids, is responsible for the replication and repair of the mitochondrial genome. Recent studies suggest that the mitochondria contain fidelity proteins and MnSOD constitutes an integral part of the nucleoid complex. However, it is not known whether or how MnSOD participates in the mitochondrial repair processes. Using skin tissue from C57BL/6 mice exposed to UVB radiation, we demonstrate that MnSOD has a critical role in preventing mtDNA damage by protecting the function of Polγ. Quantitative-PCR analysis shows an increase in mtDNA damage after UVB exposure. Immunofluorescence and immunoblotting studies demonstrate p53 translocation to the mitochondria and interaction with Polγ after UVB exposure. The mtDNA immunoprecipitation assay with Polγ and p53 antibodies in p53(+/+) and p53(-/-) mice demonstrates an interaction between MnSOD, p53 and Polγ. The results suggest that these proteins form a complex for the repair of UVB-associated mtDNA damage. The data also demonstrate that UVB exposure injures the mtDNA D-loop in a p53-dependent manner. Using MnSOD-deficient mice we demonstrate that UVB-induced mtDNA damage is MnSOD dependent. Exposure to UVB results in nitration and inactivation of Polγ, which is prevented by addition of the MnSOD mimetic Mn(III)TE-2-PyP(5+). These results demonstrate for the first time that MnSOD is a fidelity protein that maintains the activity of Polγ by preventing UVB-induced nitration and inactivation of Polγ. The data also demonstrate that MnSOD has a role along with p53 to prevent mtDNA damage.


Assuntos
DNA Polimerase Dirigida por DNA/efeitos da radiação , Superóxido Dismutase/fisiologia , Raios Ultravioleta , Animais , DNA Polimerase gama , Reparo do DNA/efeitos dos fármacos , DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/efeitos da radiação , Metaloporfirinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , Proteína Supressora de Tumor p53/metabolismo
7.
Neuroscience ; 175: 292-9, 2011 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-21074598

RESUMO

Doxorubicin (Dox) is a potent, broad-spectrum chemotherapeutic drug used around the world. Despite its effectiveness, it has a wide range of toxic side effects, many of which most likely result from its inherent pro-oxidant activity. It has been reported that Dox has toxic effects on normal tissues, including brain tissue. The present study tested the protective effect of a xanthone derivative of Garcinia Mangostana against Dox-induced neuronal toxicity. Xanthone can prevent Dox from causing mononuclear cells to increase the level of tumor necrosis factor-alpha (TNFα). We show that xanthone given to mice before Dox administration suppresses protein carbonyl, nitrotyrosine and 4-hydroxy-2'-nonenal (4HNE)-adducted proteins in brain tissue. The levels of the pro-apoptotic proteins p53 and Bax and the anti-apoptotic protein Bcl-xL were significantly increased in Dox-treated mice compared with the control group. Consistent with the increase of apoptotic markers, the levels of caspase-3 activity and TUNEL-positive cells were also increased in Dox-treated mice. Pretreatment with xanthone suppressed Dox-induced increases in all indicators of injury tested. Together, the results suggest that xanthone prevents Dox-induced central nervous system toxicity, at least in part, by suppression of Dox-mediated increases in circulating TNFα. Thus, xanthone is a good candidate for prevention of systemic effects resulting from reactive oxygen generating anticancer therapeutics.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Garcinia mangostana , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/antagonistas & inibidores , Neurotoxinas/toxicidade , Xantonas/farmacologia , Animais , Química Encefálica/efeitos dos fármacos , Química Encefálica/fisiologia , Linhagem Celular , Modelos Animais de Doenças , Garcinia mangostana/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/induzido quimicamente , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Xantonas/uso terapêutico
8.
Neuroscience ; 166(3): 796-807, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20096337

RESUMO

Adriamycin (ADR) is a chemotherapeutic for the treatment of solid tumors. This quinone-containing anthracycline is well known to produce large amounts of reactive oxygen species (ROS) in vivo. A common complaint of patients undergoing long-term treatment with ADR is somnolence, often referred to as "chemobrain." While ADR itself does not cross the blood brain barrier (BBB), we recently showed that ADR administration causes a peripheral increase in tumor necrosis factor alpha (TNF-alpha), which migrates across the BBB and leads to inflammation and oxidative stress in brain, most likely contributing to the observed decline in cognition. In the current study, we measured levels of the antioxidant glutathione (GSH) in brains of mice injected intraparitoneally (i.p.) with ADR, as well as the levels and activities of several enzymes involved in brain GSH metabolism. We observed significantly decreased GSH levels, as well as altered GSH/GSSG ratio in brains of ADR treated mice relative to saline-treated controls. Also observed in brains of ADR treated mice were increased levels of glutathione peroxidase (GPx), glutathione-S-transferase (GST), and glutathione reductase (GR). We also observed increased activity of GPx, but a significant reduction in GST and GR activity in mice brain, 72 h post i.p. injection of ADR (20 mg/kg body weight). Furthermore, we used redox proteomics to identify specific proteins that are oxidized and/or have differential levels in mice brains as a result of a single i.p. injection of ADR. Visinin like protein 1 (VLP1), peptidyl prolyl isomerase 1 (Pin1), and syntaxin 1 (SYNT1) showed differential levels in ADR treated mice relative to saline-treated controls. Triose phosphate isomerase (TPI), enolase, and peroxiredoxin 1 (PRX-1) showed significantly increased specific carbonylation in ADR treated mice brain. These results further support the notion ADR induces oxidative stress in brain despite not crossing the BBB, and that antioxidant intervention may prevent ADR-induced cognitive dysfunction.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Antioxidantes/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Doxorrubicina/efeitos adversos , Estresse Oxidativo , Animais , Encéfalo/enzimologia , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Injeções Intraperitoneais , Masculino , Camundongos , Oxirredução , Carbonilação Proteica , Proteômica
9.
Neuroscience ; 153(1): 120-30, 2008 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-18353561

RESUMO

Alzheimer's disease (AD) is associated with beta-amyloid accumulation, oxidative stress and mitochondrial dysfunction. However, the effects of genetic mutation of AD on oxidative status and mitochondrial manganese superoxide dismutase (MnSOD) production during neuronal development are unclear. To investigate the consequences of genetic mutation of AD on oxidative damages and production of MnSOD during neuronal development, we used primary neurons from new born wild-type (WT/WT) and amyloid precursor protein (APP) (NLh/NLh) and presenilin 1 (PS1) (P264L) knock-in mice (APP/PS1) which incorporated humanized mutations in the genome. Increasing levels of oxidative damages, including protein carbonyl, 4-hydroxynonenal (4-HNE) and 3-nitrotyrosine (3-NT), were accompanied by a reduction in mitochondrial membrane potential in both developing and mature APP/PS1 neurons compared with WT/WT neurons suggesting mitochondrial dysfunction under oxidative stress. Interestingly, developing APP/PS1 neurons were significantly more resistant to beta-amyloid 1-42 treatment, whereas mature APP/PS1 neurons were more vulnerable than WT/WT neurons of the same age. Consistent with the protective function of MnSOD, developing APP/PS1 neurons have increased MnSOD protein and activity, indicating an adaptive response to oxidative stress in developing neurons. In contrast, mature APP/PS1 neurons exhibited lower MnSOD levels compared with mature WT/WT neurons indicating that mature APP/PS1 neurons lost the adaptive response. Moreover, mature APP/PS1 neurons had more co-localization of MnSOD with nitrotyrosine indicating a greater inhibition of MnSOD by nitrotyrosine. Overexpression of MnSOD or addition of MnTE-2-PyP(5+) (SOD mimetic) protected against beta-amyloid-induced neuronal death and improved mitochondrial respiratory function. Together, the results demonstrate that compensatory induction of MnSOD in response to an early increase in oxidative stress protects developing neurons against beta-amyloid toxicity. However, continuing development of neurons under oxidative damage conditions may suppress the expression of MnSOD and enhance cell death in mature neurons.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/genética , Aldeídos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Encéfalo/fisiopatologia , Respiração Celular/efeitos dos fármacos , Respiração Celular/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Humanos , Potencial da Membrana Mitocondrial/genética , Metaloporfirinas/farmacologia , Camundongos , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Doenças Mitocondriais/fisiopatologia , Mutação/genética , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Presenilina-1/genética , Carbonilação Proteica/fisiologia , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Tirosina/análogos & derivados , Tirosina/metabolismo
10.
Neuroscience ; 151(2): 622-9, 2008 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-18160224

RESUMO

Tumor necrosis factor-alpha (TNF-alpha), a ubiquitous pro-inflammatory cytokine, is an important mediator in the immune-neuroendocrine system that affects the CNS. The present study demonstrates that treatment with TNF-alpha activates microglia to increase TNF-alpha production in primary cultures of glial cells isolated from wild-type (WT) mice and mice deficient in the inducible form of nitric oxide synthase (iNOSKO). However, mitochondrial dysfunction in WT neurons occurs at lower concentrations of TNF-alpha when neurons are directly treated with TNF-alpha or co-cultured with TNF-alpha-treated microglia than iNOSKO neurons similarly treated. Immunofluorescent staining of primary neurons co-cultured with TNF-alpha-treated microglia reveals that the antioxidant enzyme in mitochondria, manganese superoxide dismutase (MnSOD), is co-localized with nitrotyrosine in WT but not in iNOSKO primary neuronal cells. Importantly, the percentage of surviving neurons is significantly reduced in WT neurons compared with iNOSKO neurons under identical treatment conditions. Together, the results suggest that TNF-alpha activates microglia to produce high levels of TNF-alpha and that production of nitric oxide (NO) in neurons is an important factor affecting MnSOD nitration and subsequent mitochondrial dysfunction.


Assuntos
Mitocôndrias/fisiologia , Neuroglia/fisiologia , Neurônios/metabolismo , Nitratos/metabolismo , Óxido Nítrico/biossíntese , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Morte Celular/genética , Morte Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Sais de Tetrazólio , Tiazóis , Tirosina/análogos & derivados , Tirosina/metabolismo
11.
Oncogene ; 25(10): 1554-9, 2006 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-16261162

RESUMO

The relationship between NF-kappaB and resistance to radiation treatment in many tumor cell types has been generally well recognized. However, which members of the NF-kappaB family contribute to radiation resistance is unclear. In the present study, we demonstrate that RelB plays an important radioprotective role in aggressive prostate cancer cells, in part by the induction of antioxidant and antiapoptotic manganese superoxide dismutase (MnSOD) gene. RelB is both constitutively present and is inducible by radiation in aggressive prostate cancer cells. Using ectopically expressed dominant negative inhibitor, p100 mutant, and the siRNA approach, we demonstrate that selective inhibition of RelB significantly decreases the levels of MnSOD resulting in a significant increase in the sensitivity of prostate cancer cells to radiation treatment. These results demonstrate that RelB plays an important role in redox regulation of the cell and protects aggressive prostate cancer cells against radiation-induced cell death. Thus, inhibition of RelB could be a novel mechanism to radiosensitize prostate cancer.


Assuntos
Raios gama , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/radioterapia , Superóxido Dismutase/genética , Fator de Transcrição RelB/fisiologia , Indução Enzimática/efeitos da radiação , Humanos , Masculino , Estresse Oxidativo/efeitos da radiação , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , RNA Mensageiro/metabolismo , Tolerância a Radiação , Superóxido Dismutase/biossíntese , Superóxido Dismutase/efeitos da radiação , Fator de Transcrição RelB/antagonistas & inibidores , Fator de Transcrição RelB/efeitos da radiação
12.
DNA Cell Biol ; 20(8): 473-81, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11560779

RESUMO

Induction of manganese superoxide dismutase (MnSOD) in response to oxidative stress has been well established in animals, tissues, and cell culture. However, the role of the human MnSOD (hMnSOD) promoter in stimulus-dependent activation of transcription is unknown. The hMnSOD promoter lacks both a TATA and a CAAT box but possesses several GC motifs. In a previous study, we showed that the basal promoter contains multiple Sp1 and AP-2 binding sites and that Sp1 is essential for the constitutive expression of the hMnSOD gene. In this study, we identified an Egr-1 binding site in the basal promoter of hMnSOD. We also found that the basal promoter is responsive to 12-O-tetradecanoylphorbol-13-acetate (TPA)-activated hMnSOD transcription in the human hepatocarcinoma cell line HepG2. The contributions of these binding sites and the roles of the transcription factors Egr-1, AP-2, and Sp1 in the activation of hMnSOD transcription by TPA were investigated by site-directed mutation analysis, Western blotting, and overexpression of transcription factors. The results showed that Sp1 plays a positive role for both basal and TPA-activated hMnSOD transcription, whereas overexpression of Egr-1 has a negative role in the basal promoter activity without any effect on TPA-mediated activation of hMnSOD transcription.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Enzimológica da Expressão Gênica , Proteínas Imediatamente Precoces , Fator de Transcrição Sp1/metabolismo , Superóxido Dismutase/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Sítios de Ligação , Proteína 1 de Resposta de Crescimento Precoce , Humanos , Manganês , Regiões Promotoras Genéticas , Acetato de Tetradecanoilforbol/farmacologia , Fator de Transcrição AP-2 , Células Tumorais Cultivadas
13.
Antioxid Redox Signal ; 3(3): 375-86, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11499385

RESUMO

Stable transfection of neomycin and human manganese superoxide dismutase (MnSOD2) expression plasmids into a murine fibrosarcoma cell line (FSa-II) was previously done in our laboratory. Treatment with 10 microM 5-azacytidine induced apoptosis in the control cell line (NEO), whereas the MnSOD-overexpressing cell line (SOD-H) demonstrated differentiated-appearing morphology. The levels of the myogenic transcription factor, MyoD, and the muscle-specific marker, alpha-actin, were increased over time with 5-azacytidine treatment in the SOD-H cell line. Nuclear transcription factor NFkappaB was activated in the SOD-H cell line, whereas inhibition of NFkappaB activation reduced the levels of MyoD and alpha-actin. Members of mitogen-activated protein kinase pathway and the Raf1/MEK/ERK cascade were shown to play a positive role in this event. Overexpression of MnSOD not only can protect cells from the toxic effects of 5-azacytidine, but can also promote the fibrosarcoma cells to enter a differentiation program.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Apoptose , Azacitidina/farmacologia , Fibrossarcoma/metabolismo , Fibrossarcoma/terapia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Actinas/biossíntese , Animais , Caspase 3 , Caspases/metabolismo , Diferenciação Celular , Linhagem Celular , Relação Dose-Resposta a Droga , Fibrossarcoma/patologia , Cinética , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C3H , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína MyoD/biossíntese , NF-kappa B/fisiologia , Superóxido Dismutase/fisiologia , Transfecção , Células Tumorais Cultivadas
14.
Anticancer Res ; 20(1A): 7-10, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10769628

RESUMO

The relationship between spontaneous apoptosis and overexpression of manganese superoxide dismutase (MnSOD) gene was examined in vivo. The mouse fibrosarcoma cells expressing high MnSOD activities due to transfection with the human MnSOD cDNA (SOD-H), or the fibrosarcoma cells transfected with the selectable marker alone (NEO), were transplanted into immune-deficient Fox Chase SCID C.B-17/Icr-scid Jcl mice. Apoptosis in tumors was visually quantified by the in situ end-labeling method. The number of apoptotic cells in the SOD-H tumors was significantly less than that in the NEO tumors. The tumor growth time of the SOD-H tumors to grow from 34 to 500 mm3 in one-half of the mice was slightly longer than that of the NEO tumors, but the difference was not statistically significant. These results suggest that overexpression of MnSOD gene is involved in the suppression of spontaneous apoptosis, without a resultant alteration in the tumor growth.


Assuntos
Apoptose/genética , Fibrossarcoma/patologia , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/fisiologia , Superóxido Dismutase/fisiologia , Animais , DNA Complementar/genética , Indução Enzimática , Humanos , Manganês/fisiologia , Camundongos , Camundongos SCID , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Proteínas Recombinantes de Fusão/fisiologia , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Transfecção , Células Tumorais Cultivadas
15.
Free Radic Biol Med ; 28(3): 397-402, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10699751

RESUMO

Manganese superoxide dismutase (Mn-SOD) plays an important role in attenuating free radical-induced oxidative damage. The purpose of this research was to determine if increased expression of Mn-SOD gene alters intracellular redox status. Twelve week old male B6C3 mice, engineered to express human Mn-SOD in multiple organs, and their nontransgenic littermates were assessed for oxidative stress and antioxidant status in heart, brain, lung, skeletal muscle, liver, and kidney. Relative to their nontransgenic littermates, transgenic mice had significantly (p <.01) higher activity of Mn-SOD in heart, skeletal muscle, lung, and brain. Copper, zinc (Cu,Zn)-SOD activity was significantly higher in kidney, whereas catalase activity was lower in brain and liver. The activities of selenium (Se)-GSH peroxidase and non-Se-GSH peroxidase, and levels of vitamin E, ascorbic acid and GSH were not significantly different in any tissues measured between Mn-SOD transgenic mice and their nontransgenic controls. The levels of malondialdehyde were significantly lower in the muscle and heart of Mn-SOD mice, and conjugated dienes and protein carbonyls were not altered in any tissues measured. The results obtained showed that expression of human SOD gene did not systematical alter antioxidant systems or adversely affect the redox state of the transgenic mice. The results also suggest that expression of human SOD gene confers protection against peroxidative damage to membrane lipids.


Assuntos
Antioxidantes/metabolismo , Catalase/metabolismo , Glutationa Peroxidase/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Animais , Ácido Ascórbico/metabolismo , Glutationa/metabolismo , Humanos , Masculino , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Especificidade de Órgãos , Oxidantes/metabolismo , Oxirredução , Vitamina E/metabolismo
16.
DNA Cell Biol ; 18(9): 709-22, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10492402

RESUMO

Tumor necrosis factor-alpha (TNF) and interleukin-1beta (IL-1) are cytokines that induce expression of various genes through activation of the redox-sensitive transcription factor nuclear factor-kappaB (NF-kappaB). We have previously cloned the entire human MnSOD (SOD2) gene and found several NF-kappaB-binding sites in the 5' and 3' flanking and intronic regions. To test whether these putative NF-kappaB-binding sites are able to respond to TNF and IL-1, we performed induction analysis using various deletion constructs ligated to a luciferase reporter gene. We found that the 5' and 3' flanking regions containing several NF-kappaB-binding sites do not mediate MnSOD induction by TNF or IL-1. When a 342-bp intron 2 fragment containing NF-kappaB, C/EBP, and NF-1 binding sites was linked to the basal promoter of the SOD2 gene, transcriptional activities were significantly increased in response to TNF and IL-1 in an orientation- and position-independent manner. To accurately identify the element that is most critical for the enhancer activity, deletions and specific mutations of each individual site were studied. The results indicated that the NF-kappaB binding site is essential but not sufficient for TNF- or IL-1-mediated induction. Furthermore, NF-kappaB elements in the 5' and 3' flanking regions could be made to function in TNF or IL-1 induction when they were transposed to the intronic fragment. Taken together, these results suggest that an NF-kappaB element and its location in the SOD2 gene is critical for TNF/IL-1-mediated induction. However, a complex interaction between NF-kappaB and other transcription elements is needed for a high-level induction.


Assuntos
Regulação Enzimológica da Expressão Gênica/genética , Interleucina-1/farmacologia , Íntrons , NF-kappa B/metabolismo , Superóxido Dismutase/genética , Fator de Necrose Tumoral alfa/farmacologia , Sequência de Bases , Sítios de Ligação , Primers do DNA , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Mutagênese Sítio-Dirigida
17.
FASEB J ; 13(12): 1601-10, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10463952

RESUMO

Mitochondria have recently been shown to serve a central role in programmed cell death. In addition, reactive oxygen species (ROS) have been implicated in cell death pathways upon treatment with a variety of agents; however, the specific cellular source of the ROS generation is unknown. We hypothesize that mitochondria-derived free radicals play a critical role in apoptotic cell death. To directly test this hypothesis, we treated murine fibrosarcoma cell lines, which expressed a range of mitochondrial manganese superoxide dismutase (MnSOD) activities, with respiratory chain inhibitors. Apoptosis was confirmed by DNA fragmentation analysis and electron microscopy. MnSOD overexpression specifically protected against cell death upon treatment with rotenone or antimycin. We examined bcl-x(L), p53 and poly(ADP-ribose) polymerase (PARP) to identify specific cellular pathways that might contribute to the mitochondrial-initiated ROS-mediated cell death. Cells overexpressing MnSOD contained less bcl-x(L) within the mitochondria compared to control (NEO) cells, therefore excluding the role of bcl-x(L). p53 was undetectable by Western analysis and examination of the proapoptotic protein bax, a p53 target gene, did not increase with treatment. Activation of caspase-3 (CPP-32) occurred in the NEO cells independent of cytochrome c release from the mitochondria. PARP, a target protein of CPP-32 activity, was cleaved to a 64 kDa fragment in the NEO cells prior to generation of nucleosomal fragments. Taken together, these findings suggest that mitochondrial-mediated ROS generation is a key event by which inhibition of respiration causes cell death, and identifies CPP-32 and the PARP-linked pathway as targets of mitochondrial-derived ROS-induced cell death.


Assuntos
Apoptose/fisiologia , Mitocôndrias/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/fisiologia , Superóxido Dismutase/metabolismo , Animais , Antimicina A/análogos & derivados , Antimicina A/farmacologia , Caspase 3 , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Núcleo Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Fragmentação do DNA , Fibrossarcoma , Citometria de Fluxo , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Camundongos , Mitocôndrias/enzimologia , Mitocôndrias/ultraestrutura , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Superóxido Dismutase/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2 , Proteína bcl-X
18.
J Neurosci ; 19(15): 6248-56, 1999 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10414954

RESUMO

Tumor necrosis factor alpha (TNFalpha) is widely expressed in both neurons and glia and has been shown to be upregulated after traumatic brain injury (TBI). TNFalpha receptor activation results in activation of the transcription factor nuclear factor kappaB (NF-kappaB), which may serve an antiapoptotic role via the induction of target genes manganese superoxide dismutase (MnSOD) and/or calbindin. In the present study, we used a controlled cortical impact model of TBI with pertinent lines of transgenic mice to combine both morphological characterization and molecular analysis to elucidate the role of TNFalpha after TBI. Measurements of both the lesion volume and the blood-brain barrier breach indicated exacerbations in mice rendered genetically deficient in both the p55 and p75 TNFalpha receptors (TNFR-KO) compared with wild-type animals. Additionally, animals genetically altered to overexpress MnSOD showed a significant decrease in lesion volume compared with that of control littermates, whereas no alterations were observed in mice lacking the calcium-binding protein calbindin D28k. Analysis of NF-kappaB activation and relative levels of MnSOD revealed delayed responses in the injured cortex of TNFR-KO animals compared with wild-type animals, implying that endogenous TNFalpha may be neuroprotective after TBI.


Assuntos
Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , NF-kappa B/fisiologia , Receptores do Fator de Necrose Tumoral/deficiência , Animais , Barreira Hematoencefálica/fisiologia , Calbindina 1 , Calbindinas , Córtex Cerebral/lesões , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Feminino , Camundongos , Camundongos Knockout/genética , Receptores do Fator de Necrose Tumoral/genética , Proteína G de Ligação ao Cálcio S100/genética , Proteína G de Ligação ao Cálcio S100/metabolismo , Superóxido Dismutase/metabolismo , Ferimentos não Penetrantes/metabolismo , Ferimentos não Penetrantes/patologia
19.
Oncogene ; 18(1): 93-102, 1999 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-9926924

RESUMO

Manganese superoxide dismutase (MnSOD) has been shown to play an important role in preventing the development of cancer. MnSOD activity is reduced in many transformed cells and tumor tissues. We previously showed that the reduced level of MnSOD activity in cancer cells was not due to a defect in the primary structure of MnSOD protein, but rather was due to defects in gene expression. To elucidate the cause for the reduced expression of human MnSOD in cancer, we investigated the nucleotide sequence in the regulatory region of the MnSOD gene in a normal human cell line and various human tumor cell lines. A DNA fragment spanning 3.4 kb 5' flanking region of the MnSOD gene isolated from a normal human genomic DNA library was used to determine the DNA sequence of MnSOD promoter. PCR primers were used for amplification of the 3.4 kb 5' flanking region of the human MnSOD gene in cancer cells. Sequence analysis identified three heterozygous mutations in the proximal region of the promoter in five human tumor cell lines. These mutations, clustered around the GC-rich region of the human MnSOD promoter, change the binding pattern of AP-2 and lead to a reduction in transcription activity using a luciferase reporter assay system. These results suggest that the reduced level of MnSOD expression in some tumor cells is, at least in part, due to a defect in the DNA sequence of the promoter region.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Mutação , Regiões Promotoras Genéticas , Superóxido Dismutase/genética , Sequência de Bases , Clonagem Molecular , Proteínas de Ligação a DNA/metabolismo , Células HL-60 , Células HT29 , Humanos , Manganês , Dados de Sequência Molecular , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição AP-2 , Fatores de Transcrição/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
20.
Arch Biochem Biophys ; 362(1): 59-66, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9917329

RESUMO

Adriamycin (ADR) is a potent anticancer drug that causes severe cardiomyopathy. We have previously demonstrated that ADR-induced ultrastructural mitochondrial injury in the heart was attenuated in manganese superoxide dismutase (MnSOD) transgenic mice. To further investigate the biochemical mechanisms by which MnSOD protected mitochondria against ADR-induced damage, cardiac mitochondrial function and activities were evaluated. The results showed that ADR caused significant decrease in state 3 respiration and respiratory control ratio using both complex I and II substrates in nontransgenic mice. In transgenic mice, state 3 respiration for complex I substrates remained unaffected by ADR, but was reduced for complex II substrate. Complex I activity was significantly decreased in nontransgenic, but not in transgenic mice after ADR treatment, suggesting that mitochondrial complex I is sensitive to inactivation by superoxide radicals. The activities of complex II and mitochondrial creatine kinase were decreased by ADR in both nontransgenic and transgenic mice. These results support our previous observations on the protective role of MnSOD on the ultrastructural damage of the heart after ADR treatment and extend the understanding of its mechanisms in mitochondria.


Assuntos
Cardiomiopatias/enzimologia , Doxorrubicina/toxicidade , Mitocôndrias Cardíacas/enzimologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , Superóxido Dismutase/fisiologia , Animais , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/patologia , Creatina Quinase/metabolismo , Transporte de Elétrons/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/ultraestrutura , Miocárdio/enzimologia , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Consumo de Oxigênio/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...