Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Mol Cell Biochem ; 367(1-2): 51-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22661379

ABSTRACT

Regulation and downstream effects of mitochondrial protein S-glutathionylation in response to oxidative stress are poorly understood. The study aim was to determine whether anti-oxidants such as catalase and estradiol alter mitochondrial protein S-glutathionylation and in turn affect apoptosis following ultraviolet B (UV-B) light irradiation. HeLa cells were transduced with increasing amounts of adenovirus encoding catalase (Ad-Cat) and ß-galactosidase (Ad-Lac Z) or pre-incubated with estradiol before induction of apoptosis by UV-B light exposure. Inhibition of mitochondrial protein S-glutathionylation was assessed using autoantibodies specific for the non-S-glutathionylated form of PDC-E2. The percentage of apoptotic cells following UV-B irradiation were not significantly different between mock cells (cells with no virus infection) and Ad-Cat and Ad-Lac Z infected cells at all viral doses (all p > 0.050). Autoantibody staining of non-S-glutathionylated PDC-E2 in apoptotic cells was three times greater in only Ad-Cat infected cells compared to only Ad-Lac Z infected cells (81.3 ± 16.7 vs 26 ± 7.2 %, respectively, p = 0.030). Similarly estradiol treatment (33 and 100 nM) also significantly increased PDC-E2 staining in apoptotic cells compared to non-treated cells (both p < 0.010). The percentage of apoptotic cells was not significantly different with any of the estradiol concentrations (all p > 0.100). The observed procaspase 12 cleavage following UV-B irradiation suggests that a mitochondrial-independent apoptotic pathway was activated. In conclusion, following an apoptotic stimulus, estradiol may inhibit mitochondrial protein S-glutathionylation without inhibiting apoptosis. This effect may play a role in ninefold greater prevalence of autoantibodies against PDC-E2 in women with primary biliary cirrhosis.


Subject(s)
Antioxidants/physiology , Catalase/physiology , Estradiol/physiology , Glutathione/metabolism , Mitochondrial Proteins/metabolism , Antioxidants/pharmacology , Apoptosis , Catalase/metabolism , Dihydrolipoyllysine-Residue Acetyltransferase/metabolism , Estradiol/pharmacology , Humans , Oxidative Stress
2.
BMC Genomics ; 10: 524, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19917080

ABSTRACT

BACKGROUND: The understanding of the biological function, regulation, and cellular interactions of the yeast genome and proteome, along with the high conservation in gene function found between yeast genes and their human homologues, has allowed for Saccharomyces cerevisiae to be used as a model organism to deduce biological processes in human cells. Here, we have completed a systematic screen of the entire set of 4,733 haploid S. cerevisiae gene deletion strains (the entire set of nonessential genes for this organism) to identify gene products that modulate cellular toxicity to nickel sulfate (NiSO(4)). RESULTS: We have identified 149 genes whose gene deletion causes sensitivity to NiSO(4) and 119 genes whose gene deletion confers resistance. Pathways analysis with proteins whose absence renders cells sensitive and resistant to nickel identified a wide range of cellular processes engaged in the toxicity of S. cerevisiae to NiSO(4). Functional categories overrepresented with proteins whose absence renders cells sensitive to NiSO(4) include homeostasis of protons, cation transport, transport ATPases, endocytosis, siderophore-iron transport, homeostasis of metal ions, and the diphthamide biosynthesis pathway. Functional categories overrepresented with proteins whose absence renders cells resistant to nickel include functioning and transport of the vacuole and lysosome, protein targeting, sorting, and translocation, intra-Golgi transport, regulation of C-compound and carbohydrate metabolism, transcriptional repression, and chromosome segregation/division. Interactome analysis mapped seven nickel toxicity modulating and ten nickel-resistance networks. Additionally, we studied the degree of sensitivity or resistance of the 111 nickel-sensitive and 72 -resistant strains whose gene deletion product has a similar protein in human cells. CONCLUSION: We have undertaken a whole genome approach in order to further understand the mechanism(s) regulating the cell's toxicity to nickel compounds. We have used computational methods to integrate the data and generate global models of the yeast's cellular response to NiSO(4). The results of our study shed light on molecular pathways associated with the cellular response of eukaryotic cells to nickel compounds and provide potential implications for further understanding the toxic effects of nickel compounds to human cells.


Subject(s)
Genome, Fungal/genetics , Nickel/toxicity , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Sequence Deletion/genetics , Signal Transduction/drug effects , Adenosine Triphosphatases/metabolism , Amino Acids/metabolism , Carbohydrate Metabolism/drug effects , Chromosome Segregation/drug effects , Endocytosis/drug effects , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genomics , Homeostasis/drug effects , Humans , Iron/metabolism , Models, Biological , Phenotype , Protein Transport/drug effects , Protons , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Siderophores/metabolism , Transcription, Genetic/drug effects
3.
Cell Cycle ; 8(22): 3688-94, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19855177

ABSTRACT

Higher order chromosome structures are the hallmark of mitotic and meiotic cells. Chromatin condensation and compaction are essential for rapid chromosome congression and accurate chromosome segregation during cell division. The core histones possess tails at their amino-termini. These tails, which extend from the surface of the nucleosomes, are highly dynamic and subject to an extensive array of covalent modifications. Modified histone tails play an important role, not only in the folding of nucleosomal arrays into higher order chromatin structures but also in gene regulation. The combination of these distinct covalent modifications of histones constitutes "the histone code" that regulates various cellular processes, including mitotic and meiotic progression.


Subject(s)
Gene Expression Regulation/physiology , Histones/metabolism , Meiosis/physiology , Mitosis/physiology , Nucleosomes/metabolism , Methylation , Phosphorylation , Ubiquitination
4.
Genomics ; 94(5): 294-307, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19631266

ABSTRACT

We have used Saccharomyces cerevisiae to identify toxicologically important proteins and pathways involved in arsenic-induced toxicity and carcinogenicity in humans. We performed a systemic screen of the complete set of 4733 haploid S. cerevisiae single-gene-deletion mutants to identify those that have decreased or increased growth, relative to wild type, after exposure to sodium arsenite (NaAsO(2)). IC(50) values for all mutants were determined to further validate our results. Ultimately we identified 248 mutants sensitive to arsenite and 5 mutants resistant to arsenite exposure. We analyzed the proteins corresponding to arsenite-sensitive mutants and determined that they belonged to functional categories that include protein binding, phosphate metabolism, vacuolar/lysosomal transport, protein targeting, sorting, and translocation, cell growth/morphogenesis, cell polarity and filament formation. Furthermore, these data were mapped onto a protein interactome to identify arsenite-toxicity-modulating networks. These networks are associated with the cytoskeleton, ubiquitination, histone acetylation and the MAPK signaling pathway. Our studies have potential implications for understanding toxicity and carcinogenesis in arsenic-induced human conditions, such as cancer and aging.


Subject(s)
Arsenites/toxicity , Gene Expression Regulation, Fungal/drug effects , Gene Regulatory Networks/drug effects , Genome, Fungal , Saccharomyces cerevisiae Proteins/drug effects , Saccharomyces cerevisiae/drug effects , Sodium Compounds/toxicity , Arsenites/pharmacology , Drug Resistance, Fungal , Gene Deletion , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests/methods , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sodium Compounds/pharmacology
5.
Cancer Res ; 68(11): 4077-85, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519666

ABSTRACT

Polo-like kinase 3 (Plk3) is an important mediator of the cellular responses to genotoxic stresses. In this study, we examined the physiologic function of Plk3 by generating Plk3-deficient mice. Plk3(-/-) mice displayed an increase in weight and developed tumors in various organs at advanced age. Many tumors in Plk3(-/-) mice were large in size, exhibiting enhanced angiogenesis. Plk3(-/-) mouse embryonic fibroblasts were hypersensitive to the induction of hypoxia-inducible factor-1 alpha (HIF-1 alpha) under hypoxic conditions or by nickel and cobalt ion treatments. Ectopic expression of the Plk3-kinase domain (Plk3-KD), but not its Polo-box domain or a Plk3-KD mutant, suppressed the nuclear accumulation of HIF-1 alpha induced by nickel or cobalt ions. Moreover, hypoxia-induced HIF-1 alpha expression was tightly associated with a significant down-regulation of Plk3 expression in HeLa cells. Given the importance of HIF-1 alpha in mediating the activation of the "survival machinery" in cancer cells, these studies strongly suggest that enhanced tumorigenesis in Plk3-null mice is at least partially mediated by a deregulated HIF-1 pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Base Sequence , Cell Line , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction
6.
Hepatology ; 47(5): 1483-94, 2008 May.
Article in English | MEDLINE | ID: mdl-18393316

ABSTRACT

Cytochrome P450 2E1 (CYP2E1) is suggested to play a role in alcoholic liver disease, which includes alcoholic fatty liver, alcoholic hepatitis, and alcoholic cirrhosis. In this study, we investigated whether CYP2E1 plays a role in experimental alcoholic fatty liver in an oral ethanol-feeding model. After 4 weeks of ethanol feeding, macrovesicular fat accumulation and accumulation of triglyceride in liver were observed in wild-type mice but not in CYP2E1-knockout mice. In contrast, free fatty acids (FFAs) were increased in CYP2E1-knockout mice but not in wild-type mice. CYP2E1 was induced by ethanol in wild-type mice, and oxidative stress induced by ethanol was higher in wild-type mice than in CYP2E1-knockout mice. Peroxisome proliferator-activated receptor alpha (PPARalpha), a regulator of fatty acid oxidation, was up-regulated in CYP2E1-knockout mice fed ethanol but not in wild-type mice. A PPARalpha target gene, acyl CoA oxidase, was decreased by ethanol in wild-type but not in CYP2E1-knockout mice. Chlormethiazole, an inhibitor of CYP2E1, lowered macrovesicular fat accumulation, inhibited oxidative stress, and up-regulated PPARalpha protein level in wild-type mice fed ethanol. The introduction of CYP2E1 to CYP2E1-knockout mice via an adenovirus restored macrovesicular fat accumulation. These results indicate that CYP2E1 contributes to experimental alcoholic fatty liver in this model and suggest that CYP2E1-derived oxidative stress may inhibit oxidation of fatty acids by preventing up-regulation of PPARalpha by ethanol, resulting in fatty liver.


Subject(s)
Cytochrome P-450 CYP2E1/metabolism , Ethanol/toxicity , Fatty Liver, Alcoholic/enzymology , Alanine Transaminase/blood , Animals , Cytochrome P-450 CYP2E1/deficiency , Cytochrome P-450 CYP2E1/genetics , Disease Models, Animal , Fatty Acids, Nonesterified/metabolism , Fatty Liver, Alcoholic/pathology , Female , Immunohistochemistry , Liver/pathology , Mice , Mice, Knockout , PPAR alpha/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
J Pharmacol Exp Ther ; 319(3): 1435-43, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16971506

ABSTRACT

Cycloheximide (CHX), an inhibitor of protein synthesis, has been reported to prevent cell death in a wide variety of cell types and produced by different apoptotic stimuli. However, the mechanisms by which CHX protects cells from apoptosis are still unclear. In this study, we investigated whether p53 plays a role in the protection by CHX against serum withdrawal-induced apoptosis. Deprivation of serum from the culture medium causes apoptosis in HepG2 cells, and CHX dramatically protects cells from death. p53, p21, and Bax protein levels were elevated, and cell cycle arrest was produced after serum withdrawal. CHX abolished this elevation of p53, p21, and Bax as well as the cell cycle arrest induced by serum deprivation. The p53 inhibitor pifithrin-alpha protects HepG2 cells against apoptosis induced by serum withdrawal. HepG2 cells expressing a dominant negative form of mutant p53 and Hep3B cells lacking p53 were resistant to serum withdrawal-induced apoptosis. Lowering of p53 by small interfering RNA protects HepG2 cells from serum withdrawal-induced apoptosis. p53 phosphorylation was induced by serum withdrawal and other chemotherapeutic reagents such as actinomycin D, doxorubicin, and etoposide. CHX decreases the levels of phosphorylated p53 (pp53) even in the presence of a proteasome inhibitor, which maintains the total p53 levels, whereas it does not affect the dephosphorylation of pp53. These results suggest the possibility that kinases that phosphorylate p53 might be affected by CHX administration. In summary, CHX protects HepG2 cells from serum withdrawal-induced apoptosis through inhibiting the synthesis of p53 and the phosphorylation of p53.


Subject(s)
Apoptosis/drug effects , Cycloheximide/pharmacology , Protein Synthesis Inhibitors/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Actins/metabolism , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Fragmentation/drug effects , Dactinomycin/pharmacology , Doxorubicin/pharmacology , Etoposide/pharmacology , Flow Cytometry , Humans , Phosphorylation , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Transfection , Trypan Blue , Tumor Suppressor Protein p53/metabolism
8.
Toxicol Sci ; 91(2): 365-71, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16549397

ABSTRACT

Induction of cytochrome P450 2E1 by ethanol is believed to be one of the central pathways by which ethanol generates a state of oxidative stress and causes hepatotoxicity. In order to evaluate the biochemical and toxicological actions of CYP2E1 and its sensitization of hepatotoxin-induced injury, an adenovirus which can mediate overexpression of CYP2E1 was constructed. Injecting this virus into mice through the tail vein elevated CYP2E1 protein and activity twofold in the liver of the mice compared with the mice injected with Ad-LacZ or saline. Transaminase levels were dramatically increased in mice injected with the CYP2E1 adenovirus. Histological evaluation of liver specimens of mice injected with Ad-2E1 showed liver cell injury. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) assay demonstrated that more cells were stained positively in the liver of the mice infected with Ad-2E1 than in the liver of the mice infected with Ad-LacZ. 3-Nitrotyrosine protein adducts and protein carbonyl adducts were increased in the liver of the mice infected with Ad-2E1 compared with Ad-LacZ. This potentiated toxicity most likely reflects interactions between CYP2E1- and adenovirus-mediated toxicity pathways. These results show that adenovirus-mediated overexpression of CYP2E1 could induce liver toxicity in mice and suggests a mechanism involving oxidative/nitrosative stress.


Subject(s)
Adenoviridae/genetics , Cytochrome P-450 CYP2E1/pharmacology , Liver/drug effects , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Caspases/metabolism , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Genetic Vectors , In Situ Nick-End Labeling , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Protein Binding , Tyrosine/analogs & derivatives , Tyrosine/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism , beta-Galactosidase/pharmacology
9.
J Biol Chem ; 281(8): 5128-36, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16380384

ABSTRACT

Induction of CYP2E1 by ethanol is one mechanism by which ethanol causes oxidative stress and alcohol liver disease. Although CYP2E1 is predominantly found in the endoplasmic reticulum, it is also located in rat hepatic mitochondria. In the current study, chronic alcohol consumption induced rat hepatic mitochondrial CYP2E1. To study the role of mitochondrial targeted CYP2E1 in generating oxidative stress and causing damage to mitochondria, HepG2 lines overexpressing CYP2E1 in mitochondria (mE10 and mE27 cells) were established by transfecting a plasmid containing human CYP2E1 cDNA lacking the hydrophobic endoplasmic reticulum targeting signal sequence into HepG2 cells followed by G418 selection. A 40-kDa catalytically active NH2-terminally truncated form of CYP2E1 (mtCYP2E1) was detected in the mitochondrial compartment in these cells by Western blot analysis. Cell death caused by depletion of GSH by buthionine sulfoximine (BSO) was increased in mE10 and mE27 cells as compared with cells transfected with empty vector (pCI-neo). Antioxidants were able to abolish the loss of cell viability. Increased levels of reactive oxygen species and mitochondrial 3-nitrotyrosine and 4-hydroxynonenal protein adducts and decreased mitochondrial aconitase activity and mitochondrial membrane potential were observed in mE10 and mE27 cells treated with BSO. The mitochondrial membrane stabilizer, cyclosporine A, was also able to protect these cells from BSO toxicity. These results revealed that CYP2E1 in the mitochondrial compartment could induce oxidative stress in the mitochondria, damage mitochondria membrane potential, and cause a loss of cell viability. The accumulation of CYP2E1 in hepatic mitochondria induced by ethanol consumption might play an important role in alcohol liver disease.


Subject(s)
Cytochrome P-450 CYP2E1/physiology , Glutathione/metabolism , Mitochondria/metabolism , Aldehydes/chemistry , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Blotting, Western , Buthionine Sulfoximine/chemistry , Catalysis , Cell Line , Cell Survival , Cyclosporine/chemistry , Cyclosporine/pharmacology , Cytochrome P-450 CYP2E1/metabolism , DNA, Complementary/metabolism , Endoplasmic Reticulum/metabolism , Ethanol/pharmacology , Ethanol/toxicity , Flow Cytometry , Humans , Liver/metabolism , Liver Diseases, Alcoholic/pathology , Male , Membrane Potentials , Microscopy, Confocal , Mitochondria, Liver/metabolism , Oxidative Stress , Plasmids/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions , Time Factors , Transfection , Tyrosine/analogs & derivatives , Tyrosine/chemistry
10.
Mol Cell Biochem ; 262(1-2): 165-76, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15532721

ABSTRACT

To study the biochemical and toxicological properties of cytochrome P450 2E1 (CYP2E1), an adenovirus containing human CYP2E1 cDNA (Ad-CYP2E1) was constructed and was shown to successfully mediate the overexpression of CYP2E1 in HepG2 cells. Acetaminophen (APAP) toxicity to HepG2 cells infected with Ad-CYP2E1 was characterized as a preliminary proof of principle experiment to validate the functionality of the CYP2E1 adenovirus. Compared with cells infected with Ad-LacZ, HepG2 cells infected with Ad-CYP2E1 were more sensitive to APAP induced necrosis and apoptosis when the cells were depleted of intracellular reduced glutathione (GSH). The APAP cytotoxicity was dependent on both the concentration of APAP and the multiplicity of infection of the Ad-CYP2E1 virus. Apoptosis induced by APAP in HepG2 cells overexpressing CYP2E1 was caspase dependent and could be inhibited by the pan-caspase inhibitor Z-VAD-fmk. After treatment with APAP, mitochondrial membrane potential was dramatically decreased in the CYP2E1-expressing cells. APAP protein adducts were elevated in HepG2 cells infected with Ad-CYP2E1 compared with that in cells infected with Ad-LacZ; two bands around 90 KD were found only in the CYP2E1-expressing cells. These results demonstrate that adenovirus-mediated overexpression of human CYP2E1 activates APAP to reactive metabolites which damage mitochondria, form protein adducts, and result in toxicity to HepG2 cells. The Ad-CYP2E1 may be useful for studies designed to investigate the role of CYP2E1 in APAP and alcoholic liver injury and to further characterize the actions and effects of CYP2E1.


Subject(s)
Acetaminophen/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cytochrome P-450 CYP2E1/pharmacology , Adenoviridae/genetics , Cell Line, Tumor , Cytochrome P-450 CYP2E1/genetics , Dose-Response Relationship, Drug , Drug Synergism , Genetic Vectors , Humans , Transfection
11.
Autoimmun Rev ; 2(1): 36-42, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12848974

ABSTRACT

Apoptosis is a normal physiologic form of cell death that follows activation of either an intrinsic or extrinsic pathway. In the intrinsic, various stimuli, such as oxidative stress, lead to mitochondrial dysfunction and the release of pro-apoptotic factors. Ligand binding to cell surface death receptors, such as Fas, activates the extrinsic pathway. Due to the rapid clearance of apoptotic cells, detection and quantification of apoptotic cells is prone to underestimation. In the liver, the importance of apoptosis is evident both during development and homeostasis of the biliary tree. Apoptosis also plays a prominent role in liver pathogenesis. Induction of the extrinsic apoptotic pathway by cytotoxic lymphocytes predominates in autoimmune liver diseases, viral hepatitis, and liver allograft rejection. Biliary cell apoptosis is highly regulated by bcl-2 family members. Both the extrinsic and intrinsic pathways are active in alcohol-related liver disease. Overexpression of anti-apoptotic proteins and FasL allow liver tumor cells to evade tumor specific cytotoxic lymphocytes. Agents that modulate apoptosis may be of future therapeutic benefit in a number of liver diseases.


Subject(s)
Apoptosis , Autoimmunity , Liver Diseases/physiopathology , Liver/physiology , Animals , Fas Ligand Protein , Homeostasis/physiology , Humans , Liver/immunology , Liver Diseases/immunology , Membrane Glycoproteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Signal Transduction , T-Lymphocytes, Cytotoxic/physiology , Tumor Necrosis Factor-alpha/physiology
12.
J Biol Chem ; 278(7): 4660-7, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12468545

ABSTRACT

Oxidants such as H(2)O(2) play a role in the toxicity of certain DNA-damaging agents, a process that often involves the tumor suppressor p53. H(2)O(2) is rapidly degraded by catalase, which protects cells against oxidant injury. To study the effect of catalase on apoptosis induced by DNA-damaging agents, HepG2 cells were infected with adenovirus containing the cDNA of catalase (Ad-Cat). Forty-eight hours after infection, catalase protein and activity was increased 7-10-fold compared with control cells infected with Ad-LacZ. After treatment with Vp16 or mitomycin C, control cells underwent apoptosis in a p53-dependent manner; however, overexpression of catalase inhibited this apoptosis. Basal levels as well as Vp16- or mitomycin C-stimulated levels of p53 and p21 protein were decreased in the catalase-overexpressing cells as compared with control cells; however, p53 mRNA levels were not decreased by catalase. There was no difference in p53 protein synthesis between catalase-overexpressing cells and control cells. However, pulse-chase experiments indicated that p53 protein degradation was enhanced in the catalase-overexpressing cells. Proteasome inhibitors but not calpeptin prevented the catalase-mediated decrease of p53 content. Whereas Vp16 increased, catalase overexpression decreased the phosphorylation of p53. The protein phosphatase inhibitor okadaic acid did not prevent the catalase-mediated down-regulation of p53 or phosphorylated p53. These results demonstrate that catalase protects HepG2 cells from apoptosis induced by DNA-damaging agents in association with decreasing p53 phosphorylation; the latter may lead to an acceleration in the degradation of p53 protein by the proteasome complex. This suggests that the level of catalase may play a critical role in cell-induced resistance to the effects of anti-cancer drugs which up-regulate p53.


Subject(s)
Apoptosis/genetics , Catalase/genetics , Genes, p53 , Oxidative Stress/genetics , Adenoviridae , Apoptosis/drug effects , Cell Line , DNA Damage/drug effects , DNA Damage/genetics , DNA, Complementary/genetics , Etoposide/pharmacology , Gene Expression Regulation, Enzymologic , Humans , Mitomycin/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Transfection
13.
J Pharmacol Exp Ther ; 301(1): 111-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11907164

ABSTRACT

Induction of cytochrome P450 CYP2E1 by ethanol appears to be one of the mechanisms by which ethanol creates a state of oxidative stress. Glutathione (GSH) is a key cellular antioxidant that detoxifies reactive oxygen species. Depletion of GSH, especially mitochondrial GSH, is believed to play a role in the ethanol-induced liver injury. Previous results reported that depletion of GSH by buthionine-(S,R)-sulfoximine (BSO) treatment caused apoptosis and necrosis in HepG2 cells, which overexpress CYP2E1. In the current work, adenoviral infection with vectors that resulted in expression of catalase either in the cytosol or mitochondrial compartments was able to abolish the loss of mitochondrial membrane potential or damage to mitochondria observed in HepG2 cells overexpressing CYP2E1 that were treated with BSO. Loss of cell viability, either necrotic or apoptotic, was also prevented by the catalase overexpression after infection with the adenoviral vectors. The protective effects of catalase were associated with the suppression of the increase in the production of reactive oxygen species and of mitochondrial lipid peroxidation observed after GSH depletion. These results reveal a prominent role for H(2)O(2) as a mediator in the cytotoxicity observed after depletion of GSH in HepG2 cells overexpressing CYP2E1. Damage to mitochondria may be a critical step for cellular toxicity by CYP2E1-derived reactive oxygen species.


Subject(s)
Adenoviridae/genetics , Catalase/biosynthesis , Cytochrome P-450 CYP2E1/biosynthesis , Cytosol/enzymology , Glutathione/physiology , Mitochondria/enzymology , Apoptosis/physiology , Blotting, Western , Buthionine Sulfoximine/pharmacology , Catalase/genetics , Cytochrome P-450 CYP2E1/genetics , Enzyme Inhibitors/pharmacology , Flow Cytometry , Genetic Vectors , Humans , Lipid Peroxidation/drug effects , Membrane Potentials/drug effects , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism , Tetrazolium Salts , Thiazoles , Tumor Cells, Cultured
14.
Free Radic Biol Med ; 32(1): 73-83, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11755319

ABSTRACT

Several studies have shown that pyruvate can scavenge H(2)O(2) and protect from H(2)O(2)-mediated cell injury. Mitochondria are critical participants in the control of apoptotic and necrotic cell death. Mitochondrial GSH plays an important role in the maintenance of cell functions and viability by metabolism of oxygen free radicals generated by the respiratory chain. Since loss of GSH, especially mitochondrial GSH, is associated with increased production of reactive oxygen species and cell toxicity, the ability of pyruvate to protect against these actions was evaluated. Adding pyruvate to HepG2 cells depleted of GSH by treatment with l-buthionine sulfoximine (BSO) surprisingly caused loss of viability after 24 and 48 h of incubation. Anoxia, treatment with antioxidants, and infection with cytosolic catalase, and interestingly, catalase expressed in the mitochondrial compartment were able to rescue the HepG2 cells from this pyruvate plus BSO injury, suggesting a key role for H(2)O(2), and lipid peroxides as mediators in the cytotoxicity. This toxicity and cell death observed was linked to damage to the mitochondria as evidenced by the increased lipid peroxidation in total homogenate and mitochondrial fraction, loss of mitochondrial membrane potential, and a decrease in protein-sulfhydryl groups. The type of cell death observed under these conditions was a mixture of apoptosis and necrosis. These results suggest that the protective ability of pyruvate against oxidant damage requires a functional GSH pool, especially in the mitochondrial compartment, and that in the absence of GSH, pyruvate increases cell injury by damaging the mitochondria, presumably as a consequence of enhanced electron flow and reactive oxygen production by the respiratory chain.


Subject(s)
Apoptosis , Glutathione/metabolism , Mitochondria/drug effects , Pyruvates/toxicity , Antioxidants/pharmacology , Buthionine Sulfoximine/toxicity , Carcinoma, Hepatocellular , Catalase/pharmacology , Glutathione/deficiency , Humans , Lipid Peroxidation/drug effects , Mitochondria/metabolism , Necrosis , Oxygen/metabolism , Sulfhydryl Compounds/antagonists & inhibitors , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL