Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Transplant Proc ; 41(10): 4094-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20005347

ABSTRACT

We have previously shown that treatment of steatotic livers with vitamin E succinate decreases liver injury and increases survival after ischemia/reperfusion (I/R). It is now understood that compromised energy status is associated with increased injury following liver ischemia in the setting of hepatic steatosis at least partially as a result of increased reactive oxygen species (ROS) and induction of mitochondrial uncoupling protein-2 (UCP2). Given the association between ROS, mitochondrial function, and UCP2, it was our goal to determine whether the protective effects of vitamin E succinate were associated with decreased ROS injury, down-regulation of UCP2, or improvement of ATP levels following I/R. To test this, leptin deficient (ob/ob) mice with steatotic livers that had received other 50 IU of vitamin E succinate supplement per day or control chow for 7 days were subjected to total hepatic ischemia (15 minutes) followed by reperfusion. We measured liver expressions of ATP, glutathione (GSH), and UCP2 as well as mitochondrial DNA damage. Vitamin E treatment decreased hepatic UCP2 expression and increased ATP and GSH levels prior to I/R. These levels were maintained at 1 hour after I/R. At 24 hours, while hepatic UCP2 expression, ATP, and GSH levels were similar to those of mice not receiving vitamin E, mitochondrial DNA damage was blocked. These results revealed that vitamin E succinate decreased hepatic UCP2 expression, reduced oxidative stress, and improved mitochondrial function in mice with steatotic livers before and after I/R, identifying mechanisms of protection in this setting.


Subject(s)
Fatty Liver/metabolism , Reperfusion Injury/prevention & control , Vitamin E/pharmacology , Animals , Blotting, Northern , DNA Damage , DNA, Mitochondrial/genetics , Fatty Liver/pathology , Glutathione/metabolism , Ion Channels/genetics , Leptin/deficiency , Liver/drug effects , Liver/metabolism , Liver/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mitochondrial Proteins/genetics , RNA, Messenger/genetics , Succinates/pharmacology , Uncoupling Protein 2
2.
Transplant Proc ; 40(10): 3327-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19100382

ABSTRACT

Steatotic livers represent a growing proportion of marginal organs available for transplantation. These livers are highly prone to primary nonfunction following transplantation and are therefore routinely turned down for surgery. Given the elevated levels and sensitivity for reactive oxygen species (ROS) in these livers, we evaluated whether pretreatment with a targeted ROS scavenger, vitamin E succinate, increased survival and decreased injury after ischemia/reperfusion (I/R). For this study, ob/ob mice received 50 IU/d vitamin E succinate in supplemented vs control chow for 7 days, and were subjected to 15 minutes of total hepatic ischemia and 24 hours of reperfusion. Treatment resulted in a 5-fold decrease in serum alanine aminotransferase (ALT) levels after reperfusion, mirrored by significant decreases in hepatocellular necrosis. These results suggested that targeted antioxidants such as vitamin E succinate may prove to be highly applicable for the pretreatment of steatotic donor livers, increasing their tolerance for I/R and the transplantation process.


Subject(s)
Liver/pathology , Reperfusion Injury/prevention & control , Tocopherols/therapeutic use , Animals , Ion Channels/deficiency , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mitochondrial Proteins/deficiency , Necrosis , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Uncoupling Protein 2
3.
Cardiovasc Toxicol ; 5(3): 257-67, 2005.
Article in English | MEDLINE | ID: mdl-16244371

ABSTRACT

Doxorubicin (DOX) is a widely prescribed antineoplastic and although the precise mechanism(s) have yet to be identified, DOX-induced oxidative stress to mitochondrial membranes is implicated in the pathogenic process. Previous attempts to protect against DOX-induced cardiotoxicity with alpha-tocopherol (vitamin E) have met with limited success, possibly as a result of inadequate delivery to relevant subcellular targets such as mitochondrial membranes. The present investigation was designed to assess whether enrichment of cardiac membranes with alpha-ocopherol is sufficient to protect against DOX-induced mitochondrial cardiotoxicity. Adult male Sprague-Dawley rats received seven weekly subcutaneous injections of 2 mg/kg DOX and fed either standard diet or diet supplemented with alpha-tocopherol succinate. Treatment with a cumulative dose of 14 mg/kg DOX caused mitochondrial cardiomyopathy as evidenced by histology, accumulation of oxidized cardiac proteins, and a significant decrease in mitochondrial calcium loading capacity. Maintaining rats on the alpha-tocopherol supplemented diet resulted in a significant (two- to four-fold) enrichment of cardiac mitochondrial membranes with alpha-tocopherol and diminished the content of oxidized cardiac proteins associated with DOX treatment. However, dietary alpha-tocopherol succinate failed to protect against mitochondrial dysfunction and cardiac histopathology. From this we conclude that although dietary vitamin E supplementation enriches cardiac mitochondrial membranes with alpha-tocopherol, either (1) this tocopherol enrichment is not sufficient to protect cardiac mitochondrial membranes from DOX toxicity or (2) oxidative stress alone is not responsible for the persistent mitochondrial cardiomyopathy caused by long-term DOX therapy.


Subject(s)
Antibiotics, Antineoplastic/antagonists & inhibitors , Antibiotics, Antineoplastic/toxicity , Diet , Doxorubicin/antagonists & inhibitors , Doxorubicin/toxicity , Mitochondria, Heart/drug effects , Oxidative Stress/drug effects , Vitamin E/pharmacology , Vitamins/pharmacology , Animals , Body Weight/drug effects , Calcium/metabolism , Heart Diseases/chemically induced , Heart Diseases/pathology , Indicators and Reagents , Mitochondria, Heart/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Organ Size/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley
4.
Chem Biol Interact ; 138(3): 267-84, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11714483

ABSTRACT

The mechanism of alpha-tocopheryl succinate (TS) cytoprotection against mitochondria-derived oxidative stress was investigated. Incubation of isolated rat hepatocytes with ethyl methanesulfonate (EMS), a mitochondrial alkylating toxicant caused mitochondrial dysfunction and necrotic cell death that was dependent on the production of reactive oxygen species (ROS) and lipid peroxidation. Mitochondria isolated from these cells showed a 3-fold increase in lipid hydroperoxides and a selective depletion of alpha-tocopherol (T), which preceded cell death. The pretreatment of hepatocytes with TS dramatically enriched cells and mitochondria with alpha-tocopherol and provided these membranes with complete protection against EMS-induced oxidative damage. TS pretreatment suppressed EMS-induced cellular ROS production, generated from mitochondrial complex I and III sites. In addition, the treatment with either rotenone (ROT, a complex I inhibitor) or antimycin A (AA, a complex III inhibitor) potentiated EMS-induced lipid peroxidation and necrotic cell death which were again completely prevented by TS treatment. Surprisingly, TS did not protect hepatocytes against thenoyltrifluoroacetone (TTFA), a complex II inhibitor-induced enhancement of EMS-induced toxic oxidative damage. We conclude that the inhibition of mitochondrial ROS production and lipid peroxidation by T released from TS, are the critical events responsible for TS-mediated cytoprotection against toxic oxidative stress derived from both mitochondrial complexes I and III. Our findings suggest that TS treatment may prove useful in combating diseases associated with mitochondrial-derived oxidative stress.


Subject(s)
Antioxidants/pharmacology , Electron Transport/drug effects , Ethyl Methanesulfonate/toxicity , Hepatocytes/drug effects , Mitochondria/drug effects , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Animals , Antimycin A/pharmacology , Cell Survival/drug effects , Cells, Cultured , Drug Antagonism , Enzyme Inhibitors/pharmacology , Hepatocytes/metabolism , Hepatocytes/pathology , Lipid Peroxidation , Male , Mitochondria/metabolism , Necrosis , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Rotenone/pharmacology , Thenoyltrifluoroacetone/pharmacology , Tocopherols/metabolism
5.
Free Radic Biol Med ; 31(4): 530-41, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11498286

ABSTRACT

Numerous in vitro studies attest to the enhanced ability of vitamin E succinate (TS), as compared with conventional vitamin E compounds such as unesterified d-alpha-tocopherol (T) and d-alpha-tocopheryl acetate (TA), to protect hepatocytes from toxic oxidative stress. In the present study we tested the hypothesis that this unique protective ability is related to an enhanced cellular accumulation of TS. The results of this study indicate, using both in vitro and in vivo model systems, that acute TS administration results in a rapid increase in T and TS content and antioxidant protection of hepatocytes and mitochondria. In contrast, conventional vitamin E compounds such as T and TA lack these same protective properties. We suggest that TS acts as a unique delivery system for T, rapidly accumulating in cellular and mitochondrial membranes and gradually releasing active T to prevent membrane oxidative damage. We propose that TS administration may prove useful for the prevention and treatment of oxidative stress-mediated diseases, especially those of mitochondrial origin.


Subject(s)
Antioxidants/pharmacology , Cytoprotection/drug effects , Hepatocytes/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Animals , Cell Survival/drug effects , Free Radicals , Hepatocytes/drug effects , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation , Liver/drug effects , Male , Mitochondria, Liver/drug effects , Oxidative Stress , Rats , Rats, Sprague-Dawley , Subcellular Fractions , Thiobarbituric Acid Reactive Substances/metabolism , Tocopherols
6.
Arch Biochem Biophys ; 393(1): 87-96, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11516165

ABSTRACT

Mitochondrial electron transport inhibitors induced two distinct pathways for acute cell death: lipid peroxidation-dependent and -independent in isolated rat hepatocytes. The toxic effects of mitochondrial complex I and II inhibitors, rotenone (ROT) and thenoyltrifluoroacetone (TTFA), respectively, were dependent on oxidative stress and lipid peroxidation, while cell death induced by inhibitors of complexes III and IV, antimycin A (AA) and cyanide (CN), respectively, was caused by MMP collapse and loss of cellular ATP. Accordingly, cellular and mitochondrial antioxidant depletion or supplementation, in general, resulted in a dramatic potentiation or prevention, respectively, of toxic injury induced by complex I and II inhibitors, with little or no effect on complex III and IV inhibitor-induced toxicity. ROT-induced oxidative stress was prevented by the addition of d-alpha-tocopheryl succinate (TS) but surprisingly TS did not afford hepatocytes protection against TTFA-induced oxidative damage. TS treatment prevented ROT-induced mitochondrial lipid hydroperoxide formation but had no effect on the loss of mitochondrial GSH or cellular ATP, suggesting a mitochondrial lipid peroxidation-mediated mechanism for ROT-induced acute cell death. In contrast, only fructose treatment provided excellent cytoprotection against AA- and CN-induced toxicity. Our findings indicate that complex III and IV inhibitors cause a rapid and severe depletion of cellular ATP content resulting in acute cell death that is dependent on cellular energy impairment but not lipid peroxidation. In contrast, inhibitors of mitochondrial complex I or II moderately deplete cellular ATP levels and thus cause acute cell death via a lipid peroxidation pathway.


Subject(s)
Antioxidants/pharmacology , Cell Death/drug effects , Electron Transport/drug effects , Lipid Peroxidation/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Vitamin E/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Antimycin A/toxicity , Cyanides/toxicity , Glutathione/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , In Vitro Techniques , Male , Maleates/toxicity , Oxidative Stress/drug effects , Rats , Rotenone/toxicity , Thenoyltrifluoroacetone/toxicity , Tocopherols , Vitamin E/pharmacology
7.
Chem Biol Interact ; 127(3): 201-17, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10967318

ABSTRACT

Diethyl maleate (DEM) (5 mM) and ethyl methanesulfonate (EMS) (35 mM) treatments rapidly depleted cellular reduced glutathione (GSH) below detectable levels (1 nmol/10(6) cells), and induced lipid peroxidation and necrotic cell death in freshly isolated rat hepatocytes. In hepatocytes incubated with 2.5 mM DEM and 10 mM EMS, however, the complete depletion of cellular GSH observed was not sufficient to induce lipid peroxidation or cell death. Instead, DEM- and EMS-induced lipid peroxidation and cell death were dependent on increased reactive oxygen species (ROS) production as measured by increases in dichlorofluorescein fluorescence. The addition of antioxidants (vitamin E succinate and deferoxamine) prevented lipid peroxidation and cell death, suggesting that lipid peroxidation is involved in the sequence of events leading to necrotic cell death induced by DEM and EMS. To investigate the subcellular site of ROS generation, the cytochrome P450 inhibitor, SKF525A, was found to reduce EMS-induced lipid peroxidation but did not protect against the loss of cell viability, suggesting a mitochondrial origin for the toxic lipid peroxidation event. In agreement with this conclusion, mitochondrial electron transport inhibitors (rotenone, thenoyltrifluoroacetone and antimycin A) increased EMS-induced lipid peroxidation and cell death, while the mitochondrial uncoupler, carbonyl cyanide m-chlorophenylhydrazone, blocked EMS- and DEM-mediated ROS production and lipid peroxidation. Furthermore, EMS treatment resulted in the significant loss of mitochondrial alpha-tocopherol shortly after its addition, and this loss preceded losses in cellular alpha-tocopherol levels. Treatment of hepatocytes with cyclosporin A, a mitochondrial permeability transition inhibitor, oxypurinol, a xanthine oxidase inhibitor, or BAPTA-AM, a calcium chelator, provided no protection against EMS-induced cell death or lipid peroxidation. Our results indicate that DEM and EMS induce cell death by a similar mechanism, which is dependent on the induction of ROS production and lipid peroxidation, and mitochondria are the major source for this toxic ROS generation. Cellular GSH depletion in itself does not appear to be responsible for the large increases in ROS production and lipid peroxidation observed.


Subject(s)
Glutathione/metabolism , Hepatocytes/metabolism , Reactive Oxygen Species/metabolism , Vitamin E/analogs & derivatives , Alkylating Agents/toxicity , Animals , Antioxidants/pharmacology , Cell Death/drug effects , Cell Membrane Permeability/drug effects , Cytochrome P-450 Enzyme Inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Ethyl Methanesulfonate/toxicity , Hepatocytes/drug effects , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lipid Peroxidation/drug effects , Male , Maleates/toxicity , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Proadifen/pharmacology , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism , Tocopherols , Vitamin E/metabolism , Vitamin E/pharmacology
8.
Lipids ; 35(2): 171-80, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10757548

ABSTRACT

Studies were conducted to determine the comparative effects of tocopherols and tocotrienols on normal mammary epithelial cell growth and viability. Cells isolated from midpregnant BALB/c mice were grown within collagen gels and maintained on serum-free media. Treatment with 0-120 microM alpha- and gamma-tocopherol had no effect, whereas 12.5-100m microM tocotrienol-rich fraction of palm oil (TRF), 100-120 microM delta-tocopherol, 50-60 microM alpha-tocotrienol, and 8-14 microM gamma- or delta-tocotrienol significantly inhibited cell growth in a dose-responsive manner. In acute studies, 24-h exposure to 0-250 microM alpha-, gamma-, and delta-tocopherol had no effect, whereas similar treatment with 100-250 microM TRF, 140-250 microM alpha-, 25-100 microM gamma- or delta-tocotrienol significantly reduced cell viability. Growth-inhibitory doses of TRF, delta-tocopherol, and alpha-, gamma-, and delta-tocotrienol were shown to induce apoptosis in these cells, as indicated by DNA fragmentation. Results also showed that mammary epithelial cells more easily or preferentially took up tocotrienols as compared to tocopherols, suggesting that at least part of the reason tocotrienols display greater biopotency than tocopherols is because of greater cellular accumulation. In summary, these findings suggest that the highly biopotent gamma- and delta-tocotrienol isoforms may play a physiological role in modulating normal mammary gland growth, function, and remodeling.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/drug effects , Mammary Glands, Animal/drug effects , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Chromans/pharmacology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Inhibitory Concentration 50 , Isomerism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred BALB C , Palm Oil , Plant Oils/pharmacology , Tocotrienols
9.
Toxicol Sci ; 53(1): 56-62, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10653521

ABSTRACT

Freshly isolated suspensions of rat parenchymal liver cells (hepatocytes) produce large amounts of nitrite following isolation. Nitrite production was inhibited by the inducible nitric oxide synthase (iNOS) inhibitor aminoguanidine, as well as the transcription inhibitor actinomycin D. Increases in iNOS mRNA, protein, and activity levels correlated with the formation of nitrite. iNOS mRNA was first detectable 2 h after the onset of hepatocyte incubations and peaked at 4 h. These results indicate that nitrite formation is a result of the large scale production of nitric oxide (NO) by hepatocytes in response to the time-dependent induction of iNOS. NO production by hepatocytes was attenuated by pretreatment of rats with the Kupffer cell inhibitor, gadolinium chloride. Also, the addition of the endotoxin neutralizing agent, polymyxin B; the protein kinase inhibitor, staurosporine, and antioxidants to perfusion buffers and hepatocyte suspensions also decreased nitrite formation. Collectively, our results suggest that iNOS is induced in hepatocytes in response to the stresses generated during collagenase isolation procedures. The response appears to be triggered by a complex interaction between several different factors including Kupffer cell activation, reactive oxygen species generation, and endotoxin contamination of collagenase preparations.


Subject(s)
Liver/metabolism , Nitric Oxide/biosynthesis , Animals , Antioxidants/pharmacology , Cell Separation , Enzyme Inhibitors/pharmacology , Gadolinium/pharmacology , Guanidines/pharmacology , In Vitro Techniques , Liver/cytology , Liver/drug effects , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitrites/metabolism , RNA/chemistry , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
10.
Antioxid Redox Signal ; 2(4): 767-77, 2000.
Article in English | MEDLINE | ID: mdl-11213481

ABSTRACT

Freshly isolated suspensions of rat parenchymal liver cells (hepatocytes) spontaneously produce large amounts of nitrite following collagenase isolation. Our previous studies indicate that nitrite production is associated with the expression of inducible nitric oxide synthase (iNOS) and reflects NO production. Depletion of glutathione (GSH) with diethylmaleate (DEM) inhibited nitrite production, and this inhibition was time-dependent. DEM was more effective in blocking nitrite production if it was added within the first 1 hr of the start of the incubation. The reducing agent dithiothreitol (DTT) and the alkylating agent ethyl methanesulfonate (EMS) also inhibited hepatocyte nitrite production, and this inhibition was also greatest if they were added within 1 hr of initiating the incubation. However, EMS added at 3 hr still reduced 6-hr nitrite production by about 70%. This reduction in nitrite production by EMS added at 3 hr may be due to the direct modification of thiol groups on the iNOS protein because we have determined that iNOS activity is inhibited by the sulfhydryl modifying reagent N-ethylmaleimide (NEM). Western blots also indicate that the iNOS protein is expressed when EMS is added at 3 hr. The addition of DEM, DTT, or EMS at 0 time greatly reduced the levels of cellular iNOS mRNA relative to controls as determined by quantitative RT-PCR. Based on our results with mRNA levels, both DTT and depletion of cellular GSH appear to inhibit the early signaling events leading to iNOS expression and suggest that the control of iNOS induction in hepatocytes is sensitive to the thiol redox status of the cell.


Subject(s)
Glutathione/metabolism , Hepatocytes/metabolism , Nitric Oxide/biosynthesis , Alkylating Agents/pharmacology , Animals , Base Sequence , DNA Primers/genetics , Dithiothreitol/pharmacology , Ethyl Methanesulfonate/pharmacology , Ethylmaleimide/pharmacology , Hepatocytes/drug effects , In Vitro Techniques , Male , Maleates/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Oxidation-Reduction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sulfhydryl Reagents/pharmacology
11.
Free Radic Biol Med ; 26(7-8): 825-35, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10232825

ABSTRACT

A series of tocopherol compounds were examined for their capacity to protect against carbon tetrachloride (CCl4)-induced hepatotoxicity in rats. Of the tocopherol compounds tested in our study, only the tris salt of d-alpha-tocopheryl hemisuccinate (TS-tris) protected against CCl4-induced hepatotoxicity. The administration of d-alpha-tocopherol (alpha-T) and the nonhydrolyzable tocopherol ether, d-alpha-tocopheryloxybutyrate tris salt (TSE-tris), failed to protect against CCl4-induced hepatotoxicity. TS-tris was the only tocopherol which significantly decreased CYP2E1 activity after 18 h. This decrease in CYP2E1 activity is likely to limit the activation of CCl4 and protect against CCl4-induced hepatotoxicity. Our results also suggest that TS-tris protection against CCl4-induced hepatotoxicity correlates with the enhanced capacity of TS-tris to deliver alpha-T and increase the antioxidant status of hepatocytes. TSE-tris did not increase cellular alpha-T levels, while administration of TS-tris produced large increases in alpha-T levels in liver homogenates as well as in liver nuclei, microsomes, mitochondria and plasma membranes. This enhanced ability to deliver tocopherol equivalents to parenchymal liver cells may be related in part to the ability of TS-tris to form liposomes in aqueous solutions. TS-tris administration protected against CCl4-induced microsomal lipid peroxide formation and inactivation of the microsomal enzyme glucose-6-phosphatase (G6Pase). Supplementation of animals with alpha-T protected against microsomal lipid peroxide formation but not against the inactivation of G6Pase. Based on our findings, we propose that high cellular levels of alpha-T protect against CCl4-induced hepatotoxicity by scavenging CCl4 radicals as well as protecting against lipid peroxidation. Our results do not support the importance of microsomal lipid peroxidation as an early event in acute CCl4-induced hepatic necrosis.


Subject(s)
Carbon Tetrachloride/toxicity , Cytochrome P-450 CYP2E1 Inhibitors , Liver/drug effects , Microsomes, Liver/metabolism , Mitochondria, Liver/metabolism , Vitamin E/analogs & derivatives , Vitamin E/metabolism , Alanine Transaminase/blood , Animals , Antioxidants/pharmacology , Glucose-6-Phosphatase/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Microsomes, Liver/drug effects , Mitochondria, Liver/drug effects , Rats , Rats, Sprague-Dawley , Tocopherols , Tromethamine/pharmacology , Vitamin E/pharmacology
12.
Biochem Pharmacol ; 57(11): 1223-6, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10230765

ABSTRACT

Isolated hepatocyte suspensions prepared by collagenase perfusion released high levels of nitrite into the extracellular medium during an 8-hr incubation. The release was time dependent, with increases first occurring by 4 hr and continuing throughout the remainder of the incubation period. Nitrite production was inhibited by the nitric oxide synthase (NOS) inhibitors aminoguanidine and N(G)-nitro-L-arginine methyl ester (L-NAME), indicating that the nitrite is derived from nitric oxide (NO) production from NOS activity. Nitrite production was not related to bacterial or Kupffer cell contamination. The protein synthesis inhibitor cycloheximide and the transcription inhibitor actinomycin D also prevented nitrite production by parenchymal hepatocytes. Calcium-independent NOS enzyme activity increased with incubation times, and this increase coincided with the observed increases in nitrite production. Our results suggest that NOS is induced following the isolation of hepatocytes, and this induction results in the formation of high levels of NO.


Subject(s)
Liver/metabolism , Nitric Oxide/metabolism , Animals , Cells, Cultured , Liver/cytology , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitrites/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
13.
J Chromatogr B Biomed Sci Appl ; 707(1-2): 308-11, 1998 Apr 10.
Article in English | MEDLINE | ID: mdl-9613964

ABSTRACT

The nonhydrolysable tocopherol ether analog, d-alpha-tocopheryloxybutyric acid (TSE), and its tocopherol ester counterpart, d-alpha-tocopheryl hemisuccinate (TS), have been shown to possess anti-tumor activity. In the present study, a sensitive high-performance liquid chromatography (HPLC) method using fluorometric detection is described for the simultaneous determination of TSE and alpha-T in biological specimens. Maximal sensitivity for the measurement of TSE and alpha-T was observed with the wavelengths, 210 nm excitation and 300 nm emission. Using an internal standard (I.S.) method, the amount of these tocopherol compounds was determined in standards, liver homogenates isolated from rats administered TSE-tris salt or vehicle (saline) and in HL-60 human leukaemia cells incubated with TSE-tris salt or saline. Treatment with TSE resulted in the significant accumulation of TSE, but not alpha-T, in the liver and HL-60 cells.


Subject(s)
Vitamin E/analogs & derivatives , Vitamin E/analysis , Animals , Chromatography, High Pressure Liquid , HL-60 Cells , Humans , Liver/chemistry , Male , Rats , Rats, Sprague-Dawley , Reference Standards , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Tocopherols , Vitamin E/pharmacokinetics
14.
Anal Biochem ; 265(2): 246-52, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9882399

ABSTRACT

The susceptibility of biological samples to lipid peroxidation can be determined by exposing samples to a lipid peroxidation initiator and measuring the length of time prior to the onset of lipid peroxidation. Previous studies have shown that aldehydes generated by lipid peroxidation can react with amines to produce fluorescent products. We have utilized this principle to develop a fluorescence plate reader assay for measuring susceptibility to lipid peroxidation. In this assay, samples are placed in glycine/phosphate buffer and loaded into a 96-well plate. Lipid peroxidation initiators are added, and fluorescence is monitored over time. Samples were assayed for susceptibility to lipid peroxidation by both the thiobarbituric acid reactive substances assay and the fluorescence plate reader assay. We found good agreement between these two methods in assessing relative susceptibility to lipid peroxidation in liver microsomes and mitochondria. The fluorescence assay was also used to monitor lipid peroxidation in liposomes and rat liver homogenates. Fluorescence was stable over an extended time period and could be induced by a variety of lipid peroxidation initiators. The fluorescence plate reader assay offers a rapid method for monitoring lipid peroxidation in a large number of samples.


Subject(s)
Lipid Peroxidation , Spectrometry, Fluorescence/instrumentation , Animals , Liposomes , Liver/metabolism , Male , Phosphatidylcholines/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism
15.
Toxicol Lett ; 92(1): 67-77, 1997 Jun 16.
Article in English | MEDLINE | ID: mdl-9242359

ABSTRACT

Rats were administered a series of tocopherol analogs 18 h prior to a hepatotoxic dose of carbon tetrachloride (CCl4). Of the compounds tested, only d-alpha-tocopheryl hemisuccinate (TS) provided significant protection against CCl4-induced hepatotoxicity. No protection was observed with either d-alpha-tocopherol (alpha-T) or a tocopherol succinate ether derivative, d-alpha-tocopheryloxybutyric acid (TSE). None of the tocopherol analogs significantly inhibited CYP2E1 activity as measured by oxidation of p-nitrophenol. Liver homogenates and subcellular fractions (cytosol, nuclei, plasma membranes, mitochondria and microsomes) were collected 18 h after tocopherol analog administration in the absence of CCl4. Homogenate and subcellular alpha-T levels were not significantly increased following TSE administration but were increased 2-3 fold following TS and alpha-T administration. Total tocopherol levels (alpha-T+ TS + TSE) in liver homogenates and subcellular fractions were highest in rats supplemented with TS. In these animals, TS was detected in all subcellular fractions and total tocopherol levels were increased from 6-23 fold over those seen in controls and 2-9 fold over alpha-T treated rats. In vitro studies in which liver homogenates and subcellular fractions were peroxidized with ascorbate and ADP/Fe suggest that increasing levels of alpha-T but not TS correlates with increased protection against lipid peroxidation. These results suggest that the ability of TS to protect against CCl4-induced hepatotoxicity relates to its enhanced hepatic accumulation and subsequent hydrolysis to alpha-T.


Subject(s)
Carbon Tetrachloride/toxicity , Liver Diseases/prevention & control , Liver/metabolism , Vitamin E/analogs & derivatives , Vitamin E/metabolism , Alanine Transaminase/metabolism , Animals , Chemical and Drug Induced Liver Injury , Cytochrome P-450 CYP2E1/drug effects , Cytochrome P-450 CYP2E1/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver Diseases/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Necrosis , Organelles/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Tocopherols , Vitamin E/pharmacology
16.
Biochem Pharmacol ; 53(5): 651-61, 1997 Mar 07.
Article in English | MEDLINE | ID: mdl-9113084

ABSTRACT

Suspensions of rat hepatocytes treated with the alkylating agent ethyl methanesulfonate (EMS) exhibited extensive lipid peroxidation as well as rapid and near complete depletion of cellular reduced glutathione (GSH) levels prior to cell death. Pretreatment of hepatocytes with medium deficient in sulfur amino acids accelerated cell death induced by EMS, confirming the previously reported cytoprotective role for GSH in this toxic event. Nearly all of the cellular GSH lost following 50 mM EMS treatment was accounted for as S-ethyl glutathione (GS-Et). No significant formation of glutathione disulfide was observed. The GS-Et formed was not exported from the cell but remained at high intracellular concentrations throughout the course of the experiment. In addition, EMS treatment inhibited the efflux of intracellular GSH and inhibited the cellular accumulation of glutamate (Glu). Supplementation of hepatocytes with 25 microM d-alpha-tocopheryl hemisuccinate (TS) protected these cells against EMS-induced lipid peroxidation and cell death. Cytoprotection with TS had no effect on EMS-induced depletion of intracellular GSH or intracellular levels of GS-Et or Glu. However, TS supplementation did prevent EMS-induced depletion of cellular protein thiols. Interestingly, the pretreatment of hepatocytes with 1 mM dithiothreitol promoted EMS toxicity. The results of this study suggest that the cytoprotective abilities of TS are related to the prevention of both EMS-induced lipid peroxidation and protein thiol depletion. Thus, the onset of lipid peroxidation and the loss of protein thiols in hepatocytes appear to be critical cellular events leading to EMS-induced cell death.


Subject(s)
Antioxidants/pharmacology , Ethyl Methanesulfonate/toxicity , Liver/drug effects , Sulfhydryl Compounds/physiology , Vitamin E/analogs & derivatives , Animals , Dithiothreitol/pharmacology , Glutathione/physiology , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Rats , Rats, Sprague-Dawley , Tocopherols , Vitamin E/pharmacology
17.
Toxicol Lett ; 90(2-3): 133-44, 1997 Feb 07.
Article in English | MEDLINE | ID: mdl-9067481

ABSTRACT

In addition to its use as a stabilizer/rigidifier of membranes, cholesteryl hemisuccinate, tris salt (CS) administration has also been shown to protect rats from the hepatotoxic effects of carbon tetrachloride (CCl4). To further our understanding of the mechanism of CS cytoprotection, we examined in rats and mice the protective abilities of CS and the non-hydrolyzable ether form of CS, gamma-cholesteryloxybutyric acid, tris salt (CSE) against acetaminophen-, adriamycin-, carbon tetrachloride-, chloroform- and galactosamine-induced toxicity. The results of these studies demonstrated that CS-mediated protection is not selective for a particular species, organ system or toxic chemical. A 24-h pretreatment of both rats and mice with a single dose of CS (100mg/kg, i.p.), resulted in significant protection against the hepatotoxic effects of CCl4, CHCl3, acetaminophen and galactosamine and against the lethal (and presumably cardiotoxic) effect of adriamycin administration. Maximal CS-mediated protection was observed in experimental animals pretreated 24 h prior to the toxic insult. These data suggest that CS intervenes in a critical cellular event that is an important common pathway to toxic cell death. The mechanism of CS protection does not appear to be dependent on the inhibition of chemical bioactivation to a toxic reactive intermediate (in light of the protection observed against galactosamine hepatotoxicity). However, based on the data presented, we can not exclude the possibility that CS administration inhibits chemical bioactivation. Our findings do suggest that CS-mediated protection is dependent on the action of the intact anionic CS molecule (non-hydrolyzable CSE was as protective as CS), whose mechanism has yet to be defined.


Subject(s)
Acetaminophen/toxicity , Carbon Tetrachloride/toxicity , Chloroform/toxicity , Cholesterol Esters/therapeutic use , Doxorubicin/toxicity , Galactosamine/toxicity , Liver Diseases/prevention & control , Animals , Chemical and Drug Induced Liver Injury/prevention & control , Heart Diseases/chemically induced , Heart Diseases/pathology , Heart Diseases/prevention & control , Liver Diseases/metabolism , Male , Mice , Mice, Inbred ICR , Rats , Rats, Sprague-Dawley
18.
Cancer Lett ; 111(1-2): 133-9, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-9022138

ABSTRACT

The growth inhibitory properties of alpha-tocopheryl hemisuccinate (vitamin E succinate) and related compounds were examined in MCF-7 human breast tumor cells and MCF-10A normal-like human breast cells since they have been suggested to be an effective antitumor compound. The data showed that both alpha-tocopherol hemisuccinate and a structurally-similar compound, cholesteryl hemisuccinate, inhibited the growth of MCF-7 and MCF-10A cells, while alpha-tocopherol, cholesterol, cholesteryl sulfate and Tris succinate had little effect on cell growth. The ether analogs of the succinate esters, alpha-tocopheryloxybutyric acid and cholesteryloxybutyric acid, also inhibited growth of MCF-7 and MCF-10A cells, indicating that hydrolysis of the succinate esters by esterases is not required for the antiproliferative effects. The antiproliferative effects of these succinate esters and ethers may be related to their physiochemical properties that allow incorporation into cell membranes.


Subject(s)
Breast Neoplasms/drug therapy , Cholesterol Esters/pharmacology , Vitamin E/analogs & derivatives , Breast Neoplasms/pathology , Cell Division/drug effects , Drug Screening Assays, Antitumor , Humans , Tocopherols , Tumor Cells, Cultured/drug effects , Vitamin E/pharmacology
19.
J Pharmacol Exp Ther ; 279(3): 1470-83, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8968373

ABSTRACT

This study of acetaminophen (AAP) hepatotoxicity examined whether some aspects of the highly integrated process of drug-induced toxicity involves apoptosis, in addition to necrosis in vivo; and if so, whether cholesteryl hemisuccinate (CS) pretreatment would selectively interfere with apoptotic or necrotic liver cell death. We have previously demonstrated that CS preexposure in vivo, protects hepatocellular necrosis and necrosis-related events induced by carbon tetrachloride (CCl4) administration. Our study demonstrates that administration of hepatotoxic doses of AAP (350-500 mg/kg, i.p.) to ICR mice (CD-1) results in severe liver injury leading to cell death both by necrosis and apoptosis. AAP-induced cell death was preceded by massive elevation in serum alanine aminotransferase coupled with rapid loss of large genomic DNA (2-24 hr), fragmentation of DNA in the form of a ladder (2-24 hr), apoptotic nuclear condensation at early hours (2-6 hr) followed by massive fragmentation and margination of heterochromatin at later (6-24) hours and a near total loss of glycogen in pericentral areas. Although CS (100 mg/kg, i.p.) alone had no noticeable biochemical or morphological effects, its administration before AAP (350-500 mg/kg, i.p.) abrogated histological and biochemical diagnostics of both apoptosis and necrosis. These include near total absence of loss of large genomic DNA and glycogen, and dramatic protection from escalating levels of liver injury. CS pretreatment also arrested AAP-induced ultrastructural changes typical of both apoptosis and necrosis. Histopathological examination of periodic acid-Schiff stained liver sections mirrored the biochemical and ultrastructural findings. In conclusion, this study for the first time establishes that apoptosis, in addition to necrosis, significantly contributes to AAP hepatotoxicity in vivo, and preexposure of mice to CS prevents AAP-induced hepatic apoptosis and necrosis.


Subject(s)
Acetaminophen/pharmacology , Apoptosis/drug effects , Cholesterol Esters/pharmacology , Liver/drug effects , Acetaminophen/antagonists & inhibitors , Animals , DNA Fragmentation , Liver/pathology , Liver/ultrastructure , Male , Mice , Microscopy, Electron , Necrosis
20.
Toxicol In Vitro ; 10(4): 383-93, 1996 Aug.
Article in English | MEDLINE | ID: mdl-20650219

ABSTRACT

Tacrine (tetrahydroaminoacridine, THA) is currently administered to thousands of patients for the treatment of Alzheimer's disease. Unfortunately, THA therapy is often limited by this drugs' propensity to induce reversible hepatotoxicity. In the present study we investigated the mechanism of THA cytotoxicity by measuring the effect of THA on cell viability, protein synthesis activity and the induction of apoptosis in suspensions of freshly isolated rat hepatocytes. Our experimental findings indicate that THA-mediated apoptosis is responsible for the acutein vitro hepatotoxicity observed with this aminoacridine derivative. We found that THA-treated hepatocytes (0.1, 0.25 and 0.5 mm) demonstrated a significant and dose-dependent reduction in cellular protein synthesis activity (84, 55 and 5% of control activity, respectively) after 1 hr of incubation. However, in hepatocytes exposed to 0.1 and 0.25 mm THA, the inhibition of protein synthesis was short-lived. In these treated cells, protein synthesis activity returned to control levels (100%) by the fifth hr of incubation without a significant increase in cellular lactate dehydrogenase (LDH) leakage or the induction of apoptosis. In hepatocytes exposed to 0.5 mm THA, the near complete inhibition of protein synthesis was not reversible and a dramatic increase in LDH leakage (necrosis) was observed after 6 hr of treatment. In 0.5 mm THA-treated hepatocytes the appearance of apoptotic nuclei and cells were observed with electron microscopy following 2 hr of treatment (12% of total hepatocytes analysed) and steadily increased to 42% by the fifth hr (compared with 4% for control cells). We speculate that THA's ability to inhibit hepatocyte protein synthesis (>50%) and induce apoptosis may have an important role in the hepatotoxic episodes experienced by Alzheimer's patients taking this drug. However, the role of apoptosis in clinical THA-induced hepatotoxicity and the relevance of using rat hepatocyte suspensions as anin vitro model for THA hepatotoxictyin vivo require additional investigation.

SELECTION OF CITATIONS
SEARCH DETAIL
...