Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
CNS Neurol Disord Drug Targets ; 9(4): 439-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20522012

ABSTRACT

Oxidative stress has been consistently linked to ageing-related neurodegenerative diseases leading to the generation of lipid peroxides, carbonyl proteins and oxidative DNA damage in tissue samples from affected brains. Studies from mouse models that express disease-specific mutant proteins associated to the major neurodegenerative processes have underscored a critical role of mitochondria in the pathogenesis of these diseases. There is strong evidence that mitochondrial dysfunction is an early event in neurodegeneration. Mitochondria are the main cellular source of reactive oxygen species and key regulators of cell death. Moreover, mitochondria are highly dynamic organelles that divide, fuse and move along axons and dendrites to supply cellular energetic demands; therefore, impairment of any of these processes would directly impact on neuronal viability. Most of the disease-specific pathogenic mutant proteins have been shown to target mitochondria, promoting oxidative stress and the mitochondrial apoptotic pathway. In addition, disease-specific mutant proteins may also impair mitochondrial dynamics and recycling of damaged mitochondria via autophagy. Collectively, these data suggest that ROS-mediated defective mitochondria may accumulate during and contribute to disease progression. Strategies aimed to improve mitochondrial function or ROS scavenging may thus be of potential clinical relevance.


Subject(s)
Disease Models, Animal , Mitochondria/physiology , Neurodegenerative Diseases/physiopathology , Oxidative Stress/physiology , Animals , Brain/metabolism , Brain/physiopathology , Humans , Mice , Mitochondria/genetics , Neurodegenerative Diseases/genetics , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase/physiology
3.
Cell Death Differ ; 16(12): 1573-81, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19779498

ABSTRACT

Growing evidence points to the fact that glucose metabolism has a central role in carcinogenesis. Among the enzymes controlling this energy production pathway, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is of particular interest. Initially identified as a glycolytic enzyme and considered as a housekeeping gene, this enzyme is actually tightly regulated and is involved in numerous cellular functions. Particularly intriguing are recent reports describing GAPDH as a regulator of cell death. However, its role in cell death is unclear; whereas some studies point toward a proapoptotic function, others describe a protective role and suggest its participation in tumor progression. In this study, we highlight recent findings and discuss potential mechanisms through which cells regulate GAPDH to fulfill its diverse functions to influence cell fate.


Subject(s)
Cell Death , Cell Transformation, Neoplastic/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Reactive Oxygen Species/metabolism
4.
Histol Histopathol ; 24(1): 117-32, 2009 01.
Article in English | MEDLINE | ID: mdl-19012251

ABSTRACT

Cholesterol is a critical component of biological membranes, which not only plays an essential role in determining membrane physical properties, but also in the regulation of multiple signaling pathways. Cells satisfy their need for cholesterol either by uptake from nutrients and lipoproteins or de novo synthesis from acetyl-CoA. The latter process occurs in the endoplasmic reticulum, where transcription factors that regulate the expression of enzymes involved in the de novo cholesterol synthesis reside. Cholesterol is distributed to different membranes most prominently to plasma membrane, where it participates in the physical organization of specific membrane domains. Mitochondria, however, are considered cholesterol-poor organelles, and obtain their cholesterol load by the action of specialized proteins involved in its delivery from extramitochondrial sources and trafficking within mitochondrial membranes. Although mitochondrial cholesterol fulfills vital physiological functions, such as the synthesis of bile acids in the liver or the formation of steroid hormones in specialized tissues, recent evidence indicates that the accumulation of cholesterol in mitochondria may be a key step in disease progression, including steatohepatitis, carcinogenesis or Alzheimer disease.


Subject(s)
Cholesterol/metabolism , Mitochondria/metabolism , Alzheimer Disease/metabolism , Animals , Humans , Liver Diseases/metabolism , Oxidative Stress/physiology
5.
An. pediatr. (2003, Ed. impr.) ; 59(6): 529-534, dic. 2003.
Article in Es | IBECS | ID: ibc-25521

ABSTRACT

Objetivo: Comunicar la experiencia de 5 años de un servicio pediátrico de transporte en helicóptero, describir las características del medio, los equipos, sus indicaciones y ventajas respecto al transporte terrestre. Métodos: Se revisan retrospectivamente 224 vuelos efectuados durante 5 años. El equipo está formado por un pediatra y una enfermera especialistas en pacientes críticos del Servicio de Pediatría del Hospital de Sant Pau de Barcelona, disponibles 365 días al año, de orto a ocaso, y opera en helicópteros del Real Automóvil Club de Cataluña coordinados por el Sistema de Emergencias Médicas. Su ámbito de actuación es Cataluña y Andorra. Se cuantifican el número de pacientes, edad, sexo, patología y tiempos de respuesta y estabilización. Resultados: El número de pacientes fue de 220, 139 varones y 81 mujeres; 6 fallecieron en el hospital emisor, 7 servicios se anularon por mala climatología, avería o negativa familiar, y se realizaron 3 transportes dobles de gemelos. Se efectuaron 224 vuelos en los que se transportaron 214 pacientes. Los tiempos medios en minutos fueron: entre alerta y despegue, 15; tiempo de vuelo, 29; desde el aterrizaje hasta la cabecera del enfermo, 10. El total fue de 54. El tiempo medio de estabilización fue de 42 min. Conclusiones: El transporte de niños críticos en helicóptero realizado por equipos especializados de pediatras y enfermeras acorta el tiempo de respuesta en las zonas alejadas y mal comunicadas. El menor número de aceleraciones y vibraciones del helicóptero aporta, sobre todo en los pacientes con traumatismos, una mayor estabilidad durante el transporte. Ambos modelos, terrestre y aéreo, deben ser complementarios (AU)


Subject(s)
Child, Preschool , Child , Adolescent , Male , Infant, Newborn , Infant , Female , Humans , Air Ambulances , Critical Care , Steroids , Age of Onset , Retrospective Studies , Prospective Studies , Arthritis, Juvenile , Anti-Inflammatory Agents , Anthropometry , Chronic Disease , Cross-Sectional Studies , Growth Disorders , Follow-Up Studies , Bone Density , Adrenocorticotropic Hormone
6.
An Pediatr (Barc) ; 59(6): 529-34, 2003 Dec.
Article in Spanish | MEDLINE | ID: mdl-14636516

ABSTRACT

OBJECTIVE: To report a 5-year experience of pediatric helicopter transport and describe its characteristics, the composition of the team, its indications and the advantages of air versus ground transport. METHODS: A total of 224 flights over a 5-year period were retrospectively analyzed. The team was composed of a pediatrician and a pediatric nurse from the Pediatric Department of Hospital Sant Pau and was available 365 days per year from sunrise to sunset. The helicopters belonged to the Royal Automobile Club of Catalonia and were coordinated by the Emergency Medical Service. The area covered was Catalonia and Andorra. The number of patients, age, sex, diagnosis, and response and stabilization times were recorded. RESULTS: There were 220 patients (139 males and 81 females). Six patients died in the primary hospital before transport. Seven flights were canceled because of adverse weather, engine breakdown, or family refusal. Three twin transportations were performed. A total of 214 patients were transported in 224 flights. The mean times (in minutes) were: from emergency call to takeoff: 15; flight time: 39; between landing to the emergency room: 10. The mean stabilization time was 42 min. CONCLUSIONS: Helicopter transportation of critically-ill children by specialist teams of pediatricians and nurses shortens response time in isolated areas with poor transport. The lower number of accelerations and vibrations of the helicopter provides greater stability during transport, especially in trauma patients. Both transport models, air and ground, should be complementary.


Subject(s)
Air Ambulances , Critical Care , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Retrospective Studies
7.
Free Radic Res ; 36(3): 345-55, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12071354

ABSTRACT

D-galactosamine (D-GalN) toxicity is a useful experimental model of liver failure in human. It has been previously observed that PGE1 treatment reduced necrosis and apoptosis induced by D-GalN in rats. Primary cultured rat hepatocytes were used to evaluate if intracellular oxidative stress was involved during the induction of apoptosis and necrosis by D-GalN (0-40mM). Also, the present study investigated if PGE1 (1 microM) was equally potent reducing both types of cell death. The presence of hypodiploid cells, DNA fragmentation and caspase-3 activation were used as a marker of hepatocyte apoptosis. Necrosis was measured by lactate dehydrogenase (LDH) release. Oxidative stress was evaluated by the intracellular production of hydrogen peroxide (H2O2), the disturbances on the mitochondrial transmembrane potential (MTP), thiobarbituric-reacting substances (TBARS) release and the GSH/GSSG ratio. Data showed that intermediate range of D-GalN concentrations (2.5-10mM) induced apoptosis in association with a moderate oxidative stress. High D-GalN concentration (40 mM) induced a reduction of all parameters associated with apoptosis and enhanced all those related to necrosis and intracellular oxidative stress, including a reduction of GSH/GSSG ratio and MTP in comparison with D-GalN (2.5-10 mM)-treated cells. Although PGE1 reduced apoptosis induced by D-GalN, it was not able to reduce the oxidative stress and cell necrosis induced by the hepatotoxin in spite to its ability to abolish the GSH depletion.


Subject(s)
Alprostadil/pharmacology , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/pathology , Galactosamine , Hepatocytes/drug effects , Liver/drug effects , Platelet Aggregation Inhibitors/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , DNA Fragmentation , Flow Cytometry , Free Radicals , Glutathione/metabolism , Hepatocytes/pathology , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation , Liver/pathology , Male , Membrane Potentials , Mitochondria/metabolism , Necrosis , Ploidies , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
8.
Hepatology ; 34(5): 964-71, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11679967

ABSTRACT

Mitochondrial glutathione (GSH) plays a key role against tumor necrosis factor alpha (TNF)-induced apoptosis because its depletion is known to sensitize hepatocytes to TNF. The present study examined the role of tauroursodeoxycholic acid (TUDCA) administration to chronic ethanol-fed rats on mitochondrial GSH levels and kinetics, mitochondrial membrane physical properties, TNF-induced peroxide formation, and subsequent hepatocyte survival. TUDCA selectively increased the levels of GSH in mitochondria without an effect on cytosolic GSH. This outcome was accompanied by improved initial rate of GSH transport examined at low (1 mmol/L) and high (10 mmol/L) GSH concentrations both in intact mitochondria and mitoplasts prepared from ethanol-fed livers. Assessment of membrane fluidity revealed an increased order parameter in mitochondria and mitoplasts from ethanol-fed rats compared with pair-fed controls, which was prevented by TUDCA administration. Compared with hepatocytes from pair-fed rats, TNF stimulated peroxide generation in hepatocytes from ethanol-fed rats, preceding TNF-induced cell death. Administration of TUDCA to ethanol-fed rats prevented TNF-induced peroxide formation and cell death, an effect that was reversed on depletion of the recovered mitochondrial GSH levels by (R,S)-3-hydroxy-4-pentenoate before TNF treatment. The protective effect of TUDCA against TNF was not because of activation of phosphatidylinositol 3-kinase, discarding a role for a survival-dependent pathway. Thus, these findings reveal a novel role of TUDCA in protecting hepatocytes in long-term ethanol-fed rats through modulation of mitochondrial membrane fluidity and subsequent normalization of mitochondrial GSH levels.


Subject(s)
Ethanol/pharmacology , Glutathione/metabolism , Hepatocytes/drug effects , Mitochondria, Liver/metabolism , Taurochenodeoxycholic Acid/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Biological Transport/drug effects , Cell Death/drug effects , Drug Synergism , Enzyme Activation , Glutathione/antagonists & inhibitors , Glutathione/deficiency , Male , Pentanoic Acids/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Sprague-Dawley
9.
Alcohol Clin Exp Res ; 25(5 Suppl ISBRA): 171S-181S, 2001 May.
Article in English | MEDLINE | ID: mdl-11391068

ABSTRACT

This article represents the proceedings of a symposium at the 2000 ISBRA Meeting in Yokohama, Japan. The chairs were Hidekazu Tsukamoto and Yoshiyuki Takei. The presentations were (1) Tribute to Professor Rajendar K. Chawla, by Craig J. McClain; (2) Dysregulated TNF signaling in alcoholic liver disease, by Craig J. McClain, S. Joshi-Barve, D. Hill, J Schmidt, I. Deaciuc, and S. Barve; (3) The role of mitochondria in ethanol-mediated sensitization of the liver, by Anna Colell, Carmen Garcia-Ruiz, Neil Kaplowitz, and Jose C. Fernandez-Checa; (4) A peroxisome proliferator (bezafibrate) can prevent superoxide anion release into hepatic sinusoid after acute ethanol administration, by Hirokazu Yokoyama, Yukishige Okamura, Yuji Nakamura, and Hiromasa Ishii; (5) S-adenosylmethionine affects tumor necrosis factor-alpha gene expression in macrophages, by Rajendar K. Chawla, S. Barve, S. Joshi-Barve, W. Watson, W. Nelson, and C. McClain; (6) Iron, retinoic acid and hepatic macrophage TNFalpha gene expression in ALD, by Hidekazu Tsukamoto, Min Lin, Mitsuru Ohata, and Kenta Motomura; and (7) Role of Kupffer cells and gut-derived endotoxin in alcoholic liver injury, by N. Enomoto, K. Ikejima, T. Kitamura, H. Oide, Y. Takei, M. Hirose, B. U. Bradford, C. A. Rivera, H. Kono, S. Peter, S. Yamashina, A. Konno, M. Ishikawa, H. Shimizu, N. Sato, and R. Thurman.


Subject(s)
Gene Expression/physiology , Liver Diseases, Alcoholic/etiology , Liver/drug effects , Mitochondria, Liver/drug effects , Superoxides/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Bezafibrate/pharmacology , Endotoxins/metabolism , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypolipidemic Agents/pharmacology , Iron/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/metabolism , Liver Diseases, Alcoholic/metabolism , Mitochondria, Liver/metabolism , Peroxisome Proliferators/pharmacology , S-Adenosylmethionine/metabolism , Tretinoin/metabolism
11.
Hepatology ; 32(1): 56-65, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10869289

ABSTRACT

Ceramide has been identified as a putative lipid messenger that mediates diverse cellular processes including cell death. Since glutathione (GSH) depletion is known to sensitize cells to many cytotoxic agents and as a result of the reported regulation of neutral sphyngomyelinase (NSMase) by GSH, the present study compared the role of individual SMases in the induction of oxidative stress, regulation of cellular GSH, and apoptosis of rat hepatocytes. Exposure of cultured rat hepatocytes to exogenous Bacillus cereus sphingomyelinase (bSMase), a neutral SMase, or human placenta sphingomyelinase (hSMase), an acidic SMase (ASMase), generated similar ceramide levels in a dose-dependent manner. However, whereas bSMase increased hepatocellular GSH levels, hSMase depleted GSH stores, an effect that was prevented by monensin and mannose 6-phosphate (M-6-P), suggesting that exogenous hSMase enters hepatocytes by endocytosis and is delivered to an endosomal/lysosomal acidic compartment. Interestingly, despite the differential effect of either SMases on cell GSH levels, both bSMase and hSMase increased gamma-glutamylcysteine synthetase heavy-subunit chain (gamma-GCS-HS) mRNA levels. Consistent with these findings on GSH regulation, hSMase, but not bSMase, generated reactive oxygen species (ROS), being accompanied by mitochondrial depolarization, suggesting that hSMase targeted mitochondria, leading to oxidative stress. Accordingly, hepatocytes displayed a selective sensitivity to hSMase in contrast to bSMase exposure, and depletion of GSH stores enhanced susceptibility to hSMase as a result of potentiation of ROS formation and caspase 3 activation. Thus, these findings reveal the ability of ASMase to induce oxidative stress as a result of the targeting of mitochondria, and that GSH depletion sensitizes hepatocytes to the ASMase-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Glutathione/metabolism , Liver/metabolism , Oxidative Stress/drug effects , Placenta/enzymology , Sphingomyelin Phosphodiesterase/pharmacology , Animals , Cells, Cultured , Ceramides/analysis , Glutamate-Cysteine Ligase/metabolism , Humans , Liver/cytology , Magnesium/pharmacology , Rats , Reactive Oxygen Species/metabolism
12.
FASEB J ; 14(7): 847-58, 2000 May.
Article in English | MEDLINE | ID: mdl-10783138

ABSTRACT

Glycosphingolipids, including gangliosides, are emerging as signaling intermediates of extracellular stimuli. Because mitochondria play a key role in the orchestration of death signals, we assessed the interaction of GD3 ganglioside (GD3) with mitochondria and the subsequent cascade of events that culminate in cell death. In vitro studies with isolated mitochondria from rat liver demonstrate that GD3 elicited a burst of peroxide production within 15-30 min, which preceded the opening of the mitochondrial permeability transition, followed by cytochrome c (cyt c) release. These effects were mimicked by lactosylceramide and N-acetyl-sphingosine but not by sphinganine or sphingosine and were prevented by cyclosporin A and butylated hydroxytoluene (BHT). Reconstitution of mitochondria pre-exposed to GD3 with cytosol from rat liver in a cell-free system resulted in the proteolytic processing of procaspase 3 and subsequent caspase 3 activation. Intact hepatocytes or U937 cells selectively depleted of glutathione in mitochondria by 3-hydroxyl-4-pentenoate (HP) with the sparing of cytosol reduced glutathione (GSH) were sensitized to GD3, manifested as an apoptotic death. Inhibition of caspase 3 prevented the apoptotic phenotype of HP-treated cells caused by GD3 without affecting cell survival; in contrast, BHT fully protected HP-treated cells to GD3 treatment. Treatment of cells with tumor necrosis factor increased the level of GD3, whereas blockers of mitochondrial respiration at complex I and II protected sensitized cells to GD3 treatment. Thus, the effect of GD3 as a lipid death effector is determined by its interaction with mitochondria leading to oxidant-dependent caspase activation. Mitochondrial glutathione plays a key role in controlling cell survival through modulation of the oxidative stress induced by glycosphingolipids.


Subject(s)
Caspases/metabolism , Cell Membrane Permeability , Cytochrome c Group/metabolism , Gangliosides/metabolism , Mitochondria, Liver/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , Enzyme Activation , Glutathione/physiology , Glycosphingolipids/metabolism , Hydrogen Peroxide/metabolism , Intracellular Membranes/metabolism , Intracellular Membranes/physiology , Membrane Potentials , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Mitochondria, Liver/physiology , Oxidative Stress , Rats , Tumor Necrosis Factor-alpha/pharmacology
13.
Hepatology ; 30(6): 1473-80, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10573527

ABSTRACT

The oxidative metabolism of ethanol by the cytochrome P450 2E1 (CYP2E1) has been recognized to contribute to the ethanol-induced deleterious effects through the induction of oxidative stress. This study compared the effect of ethanol and acetaldehyde in the induction of oxidative stress and activation of transcription factors nuclear factor-kappaB (NF-kappaB) and activating protein 1 (AP-1) in HepG2 cells, which do not express CYP2E1, and HepG2 cells transfected with CYP2E1 (E47 cells). Neither ethanol (80 mmol/L) nor acetaldehyde (25-200 micromol/L) caused oxidative stress in HepG2 cells, an effect that was independent of blocking reduced glutathione (GSH) synthesis with buthionine-L-sulfoximine (BSO). However, BSO preincubation caused an overproduction of peroxides and activation of NF-kappaB and AP-1 in E47 cells even in the absence of ethanol. Furthermore, the incubation of E47 cells with ethanol (80 mmol/L for up to 5 days) depleted cellular GSH stores in both cytosol and mitochondria, reflecting the induction of oxidative stress. Ethanol activated NF-kappaB and AP-1 in E47 cells, an effect that was prevented by 4-methylpyrazole, potentiated by cyanamide, and attenuated by trolox C. Interestingly, however, despite the inability of acetaldehyde to induce oxidative stress in HepG2, acetaldehyde activated NF-kappaB and AP-1; in contrast, ethanol failed to activate these transcription factors in HepG2. Thus, our findings indicate that activation of NF-kappaB and AP-1 by ethanol and acetaldehyde occurs through distinct mechanisms. CYP2E1 is indispensable in the induction of oxidative stress from ethanol, whereas the activation of NF-kappaB and AP-1 by acetaldehyde is independent of oxidative stress.


Subject(s)
Acetaldehyde/pharmacology , Ethanol/pharmacology , Liver/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Transcription Factor AP-1/metabolism , Antioxidants/pharmacology , Binding, Competitive , Buthionine Sulfoximine/pharmacology , Cyanamide/pharmacology , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , DNA/genetics , DNA/metabolism , Gene Expression Regulation/drug effects , Glutathione/metabolism , Humans , Liver/cytology , Liver/drug effects , Liver/enzymology , Peroxides/metabolism , Reactive Oxygen Species/metabolism , Transfection , Tumor Cells, Cultured
14.
Gastroenterology ; 115(6): 1541-51, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9834283

ABSTRACT

BACKGROUND & AIMS: Tumor necrosis factor (TNF)-alpha induces cell injury by generating oxidative stress from mitochondria. The purpose of this study was to determine the effect of ethanol on the sensitization of hepatocytes to TNF-alpha. METHODS: Cultured hepatocytes from ethanol-fed (ethanol hepatocytes) or pair-fed (control hepatocytes) rats were exposed to TNF-alpha, and the extent of oxidative stress, gene expression, and viability were evaluated. RESULTS: Ethanol hepatocytes, which develop a selective deficiency of mitochondrial glutathione (mGSH), showed marked susceptibility to TNF-alpha. The susceptibility to TNF-alpha, manifested as necrosis rather than apoptosis, was accompanied by a progressive increase in hydrogen peroxide that correlated inversely with cell survival. Nuclear factor kappaB activation by TNF-alpha was significantly greater in ethanol hepatocytes than in control hepatocytes, an effect paralleled by the expression of cytokine-induced neutrophil chemoattractant. Similar sensitization of normal hepatocytes to TNF-alpha was obtained by depleting the mitochondrial pool of GSH with 3-hydroxyl-4-pentenoate. Restoration of mGSH by S-adenosyl-L-methionine or by GSH-ethyl ester prevented the increased susceptibility of ethanol hepatocytes to TNF-alpha. CONCLUSIONS: These results indicate that mGSH controls the fate of hepatocytes in response to TNF-alpha. Its depletion caused by alcohol consumption amplifies the power of TNF-alpha to generate reactive oxygen species, compromising mitochondrial and cellular functions that culminate in cell death.


Subject(s)
Ethanol/metabolism , Glutathione/metabolism , Liver/cytology , Liver/physiology , Mitochondria/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Liver/drug effects , Male , Rats , Rats, Sprague-Dawley , S-Adenosylmethionine/pharmacology , Time Factors
15.
Biofactors ; 8(1-2): 7-11, 1998.
Article in English | MEDLINE | ID: mdl-9699001

ABSTRACT

Increasing evidence has unraveled a dual functional role of mitochondria as suppliers of the energy required for cell viability, and critical players in the pathway leading to cell death. Consequence of their physiological role in the oxidative phosphorylation is the generation of reactive oxygen species (ROS) as byproducts of the consumption of molecular oxygen in the electron transport chain. Superoxide anion and hydrogen peroxide produced during aerobic respiration are precursors of hydroxyl radical by the participation of transition metals. Glutathione (GSH) in mitochondria is the only defense available to metabolize hydrogen peroxide. A small fraction of the total cellular pool of GSH is sequestered in mitochondria by the action of a carrier that transports GSH from cytosol to the mitochondrial matrix. Recent evidence position mitochondria as subcellular targets of cytokines leading to overproduction of ROS induced by ceramide, a lipid intermediate of cytokine action. Chronic ethanol-fed cells are selectively depleted of GSH in mitochondria due to a defective operation of the carrier responsible for the transport of GSH from cytosol into the mitochondrial matrix. Its limitation sensitizes alcohol hepatocytes to the prooxidant effects of cytokines and prooxidants generated by the oxidative metabolism of ethanol. One of the mechanisms leading to the onset of selective defect in the mitochondrial transport of GSH induced by chronic ethanol exposure is mediated by decreased fluidity of the mitochondrial inner membrane. Its fluidization by SAM treatment normalizes the steady state levels of GSH in mitochondria contributing to withstand the oxidative stress derived by the oxidative metabolism of ethanol.


Subject(s)
Glutathione/metabolism , Mitochondria/physiology , Oxidative Stress/physiology , Animals , Apoptosis/drug effects , Cell Death , Ceramides/physiology , Humans , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/toxicity
16.
Free Radic Biol Med ; 25(3): 365-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9680183

ABSTRACT

Chronic ethanol feeding promotes oxidative stress in rat peripheral nerve. Malondialdehyde, a lipid peroxidation product, content increases in sciatic nerves of rats fed an ethanol-containing diet, when compared with pair-fed animals. Moreover, glutathione content and glutathione peroxidase activity in this same tissue decrease in ethanol-fed vs. pair-fed rats. S-Adenosyl-L-methionine and N-acetyl-L-cysteine, both with possible therapeutic action on alcoholism, were tested in this animal model. Only N-acetyl-L-cysteine was able to normalize malondialdehyde content and to restore glutathione content and glutathione peroxidase activity, to values not significantly different from those of sciatic nerves from pair-fed animals. The reasons for the different effect of both substances tested is also discussed.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/metabolism , Ethanol/administration & dosage , Oxidative Stress , Peripheral Nerves/metabolism , S-Adenosylmethionine/pharmacology , Animals , Ethanol/pharmacology , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Lipid Peroxidation , Male , Malondialdehyde/metabolism , Microscopy, Electron , Peripheral Nerves/drug effects , Rats , Rats, Sprague-Dawley
17.
FEBS Lett ; 427(1): 15-20, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9613591

ABSTRACT

Since glutathione (GSH) protects against oxidative stress, we determined the regulation of cellular GSH by ionizing radiation in human hepatoblastoma cells, HepG2. The levels of GSH increased in irradiated HepG2 due to a greater gamma-glutamylcysteine synthetase (gamma-GCS) activity, which was paralleled by gamma-GCS heavy subunit chain (gamma-GCS-HS) mRNA levels. Transcription of deletion constructs of the gamma-GCS-HS promoter cloned in a reporter vector was associated with activator protein-1 (AP-1), consistent with the DNA binding of AP-1 in nuclear extracts of irradiated HepG2. Hence, the transcriptional regulation of gamma-GCS by ionizing radiation emerges as an adaptive mechanism, which may be of significance to control the consequences of the oxidative stress induced by radiation.


Subject(s)
Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Gene Expression Regulation, Enzymologic/radiation effects , Glutamate-Cysteine Ligase/radiation effects , Glutathione/pharmacology , Hepatoblastoma/metabolism , Humans , Liver Neoplasms/metabolism , Oxidative Stress/drug effects , RNA, Messenger/metabolism , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured/radiation effects
18.
Semin Liver Dis ; 18(4): 389-401, 1998.
Article in English | MEDLINE | ID: mdl-9875556

ABSTRACT

Accumulating evidence pointing to mitochondria as critical participants in the control of apoptotic and necrotic cell death and in the development of specific disease states has led to a renaissance on the study of these organelles. Because mitochondria are the major consumers of molecular oxygen within cells, they stand as one of the most important generators of reactive oxygen species and therefore constitute potential targets of therapeutic intervention in pathologic states in which oxidative stress originates from these organelles. In this regard, mitochondria are specific targets of ethanol intoxication, thereby leading to reported morphologic and functional alterations of mitochondria. Because mitochondria are also indispensable for the maintenance of cell functions, their dysfunction induced by ethanol may be a key event in the development of alcoholic liver disease. Indeed, chronic ethanol feeding in experimental animals has been reported to cause a selective deficiency in the availability of reduced glutathione (GSH) in mitochondria due to the impaired functioning of the specific mitochondrial carrier that translocates GSH from cytosol into the mitochondrial matrix. Such a selective depletion sensitizes hepatocytes from chronic ethanol-fed animals to the oxidative effects of cytokines, e.g., tumor necrosis factor (TNF). Restoration of mitochondrial GSH by the in vivo administration of S-adenosyl-L-methionine or the in vitro use of GSH ethyl ester prevents the susceptibility of hepatocytes to TNF. Although the nature of this specific carrier has not yet been uncovered, the elucidation of the mechanisms whereby ethanol leads to its impaired activity may provide important clues as to its function and mechanism of action, which in turn may be useful toward the definitive characterization and identification of this important carrier.


Subject(s)
Glutathione/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Biological Transport , Ethanol/pharmacology , Humans , Liver Diseases, Alcoholic/metabolism , Mitochondria, Liver/metabolism , Oxidative Stress
19.
J Biol Chem ; 272(48): 30371-9, 1997 Nov 28.
Article in English | MEDLINE | ID: mdl-9374527

ABSTRACT

Tumor necrosis factor (TNF) is an inflammatory cytokine that causes cell injury by generation of oxidative stress. Since glutathione (GSH) is a key cellular antioxidant that detoxifies reactive oxygen species, the purpose of our work was to examine the regulation of cellular GSH, the expression of heavy subunit chain of gamma-glutamylcysteine synthetase (gamma-GCS-HS), and control of intracellular generation of reactive oxygen species in cultured rat hepatocytes treated with TNF. Exposure of cells to TNF (10,000 units/ml) resulted in depletion of cellular GSH levels (50-70%) and overproduction of hydrogen peroxide (2-3-fold) and lipid peroxidation. However, cells treated with lower doses of TNF (250-500 units/ml) exhibited increased levels of GSH (60-80% over control). TNF treatment increased (70-100%) the levels of gamma-GCS-HS mRNA, the catalytic subunit of the regulating enzyme in GSH biosynthesis. Furthermore, intact nuclei isolated from hepatocytes treated with TNF transcribed the gamma-GCS-HS gene to a greater extent than control cells, indicating that TNF regulates gamma-GCS-HS at the transcriptional level. The capacity to synthesize GSH de novo determined in cell-free extracts incubated with GSH precursors was greater (50-70%) in hepatocytes that were treated with TNF; however, the activity of GSH synthetase remained unaltered by TNF treatment indicating that TNF selectively increased the activity of gamma-GCS. Despite activation of nuclear factor-kappaB (NF-kappaB) by TNF, this transcription factor was not required for TNF-induced transcription of gamma-GCS-HS as revealed by deletion constructs of the gamma-GCS-HS promoter subcloned in a chloramphenicol acetyltransferase reporter vector and transfected into HepG2 cells. In contrast, a construct containing AP-1 like/metal response regulatory elements increased chloramphenicol acetyltransferase activity upon exposure to TNF. Thus, TNF increases hepatocellular GSH levels by transcriptional regulation of gamma-GCS-HS gene, probably through AP-1/metal response element-like binding site(s) in its promoter, which may constitute a protective mechanism in the control of oxidative stress induced by inflammatory cytokines.


Subject(s)
Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Liver/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , NF-kappa B/metabolism , Oxidation-Reduction , Promoter Regions, Genetic , RNA, Messenger/genetics , Rats , Reactive Oxygen Species/metabolism , Rotenone/pharmacology , Thenoyltrifluoroacetone/pharmacology , Transcription, Genetic/drug effects
20.
Hepatology ; 26(3): 699-708, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9303501

ABSTRACT

Ethanol intake depletes the mitochondrial pool of reduced glutathione (GSH) by impairing the transport of GSH from cytosol into mitochondria. S-Adenosyl-L-methionine (SAM) supplementation of ethanol-fed rats restores the mitochondrial pool of GSH. The purpose of the current study was to determine the effect of ethanol feeding on the kinetic parameters of mitochondrial GSH transport, the fluidity of mitochondria, and the effect of SAM on these changes. Male Sprague-Dawley rats were fed ethanol-liquid diet for 4 weeks supplemented with either SAM or N-acetylcysteine (NAC). SAM-supplementation of ethanol-fed rats restored the mitochondrial GSH pool but NAC administration did not. Kinetic studies of GSH transport in isolated mitochondria revealed two saturable, adenosine triphosphate (ATP)-stimulated components that were affected significantly by chronic ethanol feeding: lowering Vmax (0.22 and 1.6 in ethanol case vs. 0.44 and 2.7 nmol/15 sec/mg protein in controls) for both low and high affinity components with the latter showing an increased Km (15.5 vs. 8.9, mmol/L in ethanol vs. control). Mitochondria from SAM-supplemented ethanol-fed rats showed kinetic features of GSH transport similar to control mitochondria. Determination of membrane fluidity revealed an increased order parameter in ethanol compared with control mitochondria, which was restricted to the polar head groups of the bilayer and was prevented by SAM but not NAC supplementation of ethanol-fed rats. The changes elicited in mitochondria by ethanol were confined to the inner membrane; mitoplasts from ethanol-fed rats showed features similar to those of intact mitochondria such as impaired transport of GSH and increased order parameter. A different mitochondrial transporter, adenosine diphosphate (ADP)/ATP translocator, was unaffected by ethanol feeding. Furthermore, fluidization of mitochondria or mitoplasts from ethanol-fed rats by treatment with a fatty acid derivative restored their ability to transport GSH to control levels. Thus, ethanol-induced impaired transport of GSH into mitochondria is selective, mediated by decreased fluidity of the mitochondrial inner membrane, and prevented by SAM treatment.


Subject(s)
Alcoholism/metabolism , Ethanol/pharmacology , Glutathione/metabolism , Intracellular Membranes/metabolism , Mitochondria, Liver/metabolism , S-Adenosylmethionine/pharmacology , Acetylcysteine/pharmacology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Fluorescence Polarization , Intracellular Membranes/drug effects , Kinetics , Male , Membrane Fluidity/drug effects , Mitochondria, Liver/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...