Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
J Nutr Biochem ; 63: 35-43, 2019 01.
Article in English | MEDLINE | ID: mdl-30321750

ABSTRACT

High-fat diet (HFD)-fed mice show obesity with development of liver steatosis and a proinflammatory state without establishing an inflammatory reaction. The aim of this work was to assess the hypothesis that eicosapentaenoic acid (EPA) plus hydroxytyrosol (HT) supplementation prevents the inflammatory reaction through enhancement in the hepatic resolvin content in HFD-fed mice. Male C57BL/6J mice were fed an HFD or a control diet and supplemented with EPA (50 mg/kg/day) and HT (5 mg/kg/day) or their respective vehicles for 12 weeks. Measurements include liver levels of EPA, DHA and palmitate (gas chromatography), liver resolvins and triglyceride (TG) and serum aspartate transaminase (AST) (specific kits) and hepatic and serum inflammatory markers (quantitative polymerase chain reaction and enzyme-linked immunosorbent assay). Compared to CD, HFD induced body weight gain, liver steatosis and TG accumulation, with up-regulation of proinflammatory markers in the absence of histological inflammation or serum AST changes; these results were accompanied by higher hepatic levels of resolvins RvE1, RvE2, RvD1 and RvD2, with decreases in EPA and DHA contents. EPA+HT supplementation in HFD feeding synergistically reduced the steatosis score over individual treatments and increased the hepatic levels of EPA, DHA and resolvins, with attenuation of proinflammatory markers. Lack of progression of HFD-induced proinflammatory state into overt inflammation is associated with resolvin up-regulation, which is further increased by EPA+HT supplementation eliciting steatosis attenuation. These findings point to the importance of combined protocols in hepatoprotection due to the involvement of cross-talk mechanisms, which increase effectiveness and diminish dosages, avoiding undesirable effects.


Subject(s)
Diet, High-Fat/adverse effects , Eicosapentaenoic Acid/pharmacology , Hepatitis/diet therapy , Liver/drug effects , Phenylethyl Alcohol/analogs & derivatives , Animals , Dietary Supplements , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/metabolism , Fatty Acids/metabolism , Hepatitis/etiology , Hepatitis/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Phenylethyl Alcohol/pharmacology
2.
Food Funct ; 8(11): 3980-3988, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28990606

ABSTRACT

Liver preconditioning by a docosahexaenoic acid (DHA) and triiodothyronine (T3) combined protocol underlies peroxisome-proliferator activated receptor α (PPARα)-fibroblast growth factor 21 (FGF21) upregulation, the study of the regulatory mechanisms involved being the aim of this work. Combined DHA (daily doses of 300 mg kg-1 for 3 days)-T3 (0.05 mg kg-1 at the fourth day) administration elicited higher levels of liver DHA and serum T3, with enhanced hepatic nuclear/cytosolic PPARα ratios, upregulation of FGF21 and ß-Klotho expression, and a small reduction in that of FGF receptor 1 (FGFR1), compared with the respective controls. Concomitantly, the components of the FGF21 cascade extracellular-signal-regulated kinase 1/2 (ERK1/2), FGF receptor substrate 2α (FRS2α), cFos, ribosomal S6 kinase 1 (RSK1), liver kinase B1 (LKB1), and AMP-activated protein kinase (AMPK) were activated. The upregulation of liver PPARα-FGF21-AMPK signaling by the combined DHA-T3 protocol resulted in values significantly higher than those elicited by the addition of the data obtained for DHA and T3 alone. It is concluded that combined DHA-T3 supplementation achieves synergistic effects on liver PPARα-FGF21-AMPK signaling, which may result in significant metabolic changes associated with energy expenditure that are of importance in the treatment of obesity and other metabolic disorders.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Fibroblast Growth Factors/metabolism , Glucuronidase/metabolism , Liver/metabolism , Metabolic Diseases/drug therapy , Triiodothyronine/administration & dosage , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Energy Metabolism/drug effects , Fibroblast Growth Factors/genetics , Glucuronidase/genetics , Humans , Klotho Proteins , Liver/drug effects , Liver/enzymology , Male , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/physiopathology , PPAR alpha/genetics , PPAR alpha/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stress, Physiological/drug effects , Up-Regulation/drug effects
3.
J Biol Regul Homeost Agents ; 27(4): 989-99, 2013.
Article in English | MEDLINE | ID: mdl-24382180

ABSTRACT

AMP-activated protein kinase (AMPK) is a sensor of energy status supporting cellular energy homeostasis that may represent the metabolic basis for 3,3,,5-triiodo-L-thyronine (T3) liver preconditioning. Functionally transient hyperthyroid state induced by T3 (single dose of 0.1 mg/kg) in fed rats led to upregulation of mRNA expression (RT-PCR) and protein phosphorylation (Western blot) of hepatic AMPK at 8 to 36 h after treatment. AMPK Thr 172 phosphorylation induced by T3 is associated with enhanced mRNA expression of the upstream kinases Ca2+ -calmodulin-dependent protein kinase kinase-beta (CaMKKbeta) and transforming growth-factor-beta-activated kinase-1 (TAK1), with increased protein levels of CaMKKbeta and higher TAK1 phosphorylation, without changes in those of the liver kinase B1 (LKB1) signaling pathway. Liver contents of AMP and ADP were augmented by 291 percent and 44 percent by T3 compared to control values (p less than 0.05), respectively, whereas those of ATP decreased by 64% (p less than 0.05), with no significant changes in the total content of adenine nucleotides (AMP + ADP + ATP) at 24 h after T3 administration. Consequently, hepatic ATP/ADP content ratios exhibited 64 percent diminution (p less than 0.05) and those of AMP/ATP increased by 425 percent (p less than 0.05) in T3-treated rats over controls. It is concluded that in vivoT3 administration triggers liver AMPK upregulation in association with significant enhancements in AMPK mRNA expression, AMPK phosphorylation coupled to CaMKKbeta and TAK1 activation, and in AMP/ATP ratios, which may promote enhanced AMPK activity to support T3-induced energy consuming processes such as those of liver preconditioning.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Liver/metabolism , MAP Kinase Kinase Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Triiodothyronine/pharmacology , AMP-Activated Protein Kinase Kinases , Animals , Energy Metabolism , Male , Phosphorylation , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
4.
Nutr Hosp ; 26(3): 441-50, 2011.
Article in English | MEDLINE | ID: mdl-21892559

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most important cause of chronic liver disease and is considered the hepatic manifestation of the metabolic syndrome associated with diabetes mellitus type 2. The prevalence of NAFLD in the general population reaches 15-20%. It is also estimated that nonalcoholic steatohepatitis (NASH) affects 3% of the population. NAFLD refers to a wide spectrum of liver damage, which ranges from simple steatosis or intracellular triglyceride accumulation, to inflammation (NASH), fibrosis and cirrhosis. The mechanisms involved in the accumulation of triglycerides in the liver and subsequent hepatocellular damage are multifactorial and are not completely understood. However, metabolic changes such as insulin resistance (IR) are developed, being a common factor in the retention of fatty acids (FA) within the hepatocytes with oxidation and production of free radicals at the mitochondrial level, which are capable of causing lipid peroxidation, cytokine production, and necrosis. In addition, there are alterations in the hepatic bioavailability of long chain n-3 polyunsaturated fatty acids, conditions that alter the expression of a series of transcriptional factors involved in lipolytic and lipogenic processes in the liver. A greater knowledge of the etiopathogenic mechanisms of NAFLD is fundamental for the development of future effective therapeutic strategies. The pathophysiological fundamentals of liver steatosis are analyzed in this study.


Subject(s)
Fatty Liver/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Fatty Acids/metabolism , Fatty Liver/complications , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/physiopathology , Humans , Insulin Resistance , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Non-alcoholic Fatty Liver Disease , Transcription Factors
5.
Transplant Proc ; 42(5): 1569-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20620476

ABSTRACT

OBJECTIVES: Ischemic preconditioning (IP) affords resistance to liver ischemia-reperfusion (IR) injury, providing an early phase of protection. Development of delayed IP against IR injury was assessed using partial IR in rat liver. METHODS: The IP manuver (10 minutes of ischemia and up to 72 hours of reperfusion) was induced before 1 hour of ischemia and 20 hours of reperfusion. At the end of the reperfusion period, blood and liver samples were analyzed for serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), haptoglobin and tumor necrosis factor-alpha (TNF-alpha) levels, hepatic histology, protein carbonyl and glutathione (GSH) contents as well as nuclear factor-kappaB (NF-kappaB), and activating protein-1 (AP-1) DNA binding. RESULTS: The IP manuver significantly increased protein carbonyl/GSH ratios (275%), serum ALT (42%), and AST (58%); these changes normalized after 12 hours. Serum AST, ALT, and LDH levels were significantly increased by IR (4-, 5.6-, and 7.0-fold, respectively), with significant changes in liver histology, protein carbonyl/GSH ratio (481% enhancement), and serum TNF-alpha (6.1-fold increase). Delayed IP in IR animals reduced serum AST (66%), ALT (57%), and LDH (90%) and liver GSH depletion (89%), with normalization of protein carbonyl content, serum TNF-alpha levels, and liver histology. Enhanced AP-1/NF-kappaB DNA binding ratios and diminished haptoglobin expression induced by IR were normalized by IP. CONCLUSION: These data support that delayed IP suppresses IR-induced liver injury, oxidative stress, and TNF-alpha response, which coincide with recovery of IR-altered signaling functions represented by normal AP-1/NF-kappaB DNA binding ratios and acute phase responses.


Subject(s)
Ischemic Preconditioning/methods , Liver/pathology , Reperfusion Injury/prevention & control , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/metabolism , Glutathione/metabolism , Haptoglobins/metabolism , Inflammation/prevention & control , L-Lactate Dehydrogenase/blood , L-Lactate Dehydrogenase/metabolism , Liver/metabolism , Liver/physiopathology , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/metabolism
6.
Mol Cell Endocrinol ; 323(2): 292-7, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20303386

ABSTRACT

We studied the role of Kupffer cell functioning in T3 liver preconditioning against ischemia-reperfusion (IR) injury using the macrophage inactivator gadolinium chloride (GdCl3) previous to T3 treatment. Male Sprague-Dawley rats given a single i.p. dose of 0.1 mg T3/kg were subjected to 1 h ischemia followed by 20 h reperfusion, in groups of animals pretreated with 10 mg GdCl3/kg i.v. 72 h before T(3) or with the respective vehicles. IR resulted in significant enhancement of serum aspartate aminotransferase (3.3-fold increase) and tumor necrosis factor-alpha (93% increase) levels, development of liver damage, and diminished nuclear factor-kappaB DNA binding over control values. These changes, which were suppressed by the T3 administration prior to IR, persisted in animals given GdCl3 before T3 treatment, under conditions of complete elimination of ED2+ Kupffer cells achieved in a time window of 72 h. It is concluded that Kupffer cell functioning is essential for T3 liver preconditioning, assessed in a warm IR injury model by hepatic macrophage inactivation.


Subject(s)
Ischemic Preconditioning , Kupffer Cells/physiology , Liver/drug effects , Liver/physiopathology , Reperfusion Injury/physiopathology , Triiodothyronine/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Aspartate Aminotransferases/blood , DNA/metabolism , Gadolinium/pharmacology , Liver/cytology , Liver/metabolism , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/blood
7.
Free Radic Res ; 36(7): 741-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12180124

ABSTRACT

beta 2-Glycoprotein I (beta 2 GPI) is known to influence macrophage uptake of particles with phosphatidylserine containing surfaces, as apoptotic thymocytes and unilamellar vesicles in vitro. Nevertheless, effects upon macrophage activation induced by this interaction are still unknown. beta 2 GPI influence upon the reactive species production by Kupffer cells was evaluated in order to investigate whether beta 2 GPI modulates the macrophage response to negatively charged surfaces. Chemiluminescence of isolated non-parenchymal rat liver cells was measured after phagocytosis of opsonized zymosan or phorbolymristate acetate (PMA) stimulation, in the presence and absence of large unilamellar vesicles (LUVs) containing 25 mol% phosphatidylserine (PS) or 50 mol% cardiolipin (CL) and complementary molar ratio of phosphatidylcholine (PC). beta 2 GPI decreased by 50% the chemiluminescence response induced by opsonized zymosan, with a 66% reduction of the initial light emission rate. PMA stimulated Kupffer cell chemiluminescence was insensitive to human or rat beta 2 GPI. Albumin (500 micrograms/ml) showed no effect upon chemiluminescence. beta 2 GPI increased PS/PC LUV uptake and degradation by Kupffer cells in a concentration-dependent manner, without leakage of the internal contents of the LUVs, as shown by fluorescence intensity enhancement. LUVs opsonized with antiphospholipid antibodies (aPL) from syphilitic patients increased light emission by Kupffer cells. Addition of beta 2 GPI to the assay reduced chemiluminescence due to opsonization with purified IgG antibodies from systemic lupus erythematosus (SLE or syphilis (Sy) patient sera. A marked net increase in chemiluminescence is observed in the presence of Sy aPL antibodies, whereas a decrease was found when SLE aPL were added to the assay, in the presence or absence of beta 2 GPI. At a concentration of 125 micrograms/ml, beta 2 GPI significantly reduced Kupffer cell Candida albicans phagocytosis index and killing score by 50 and 10%, respectively. The present data strongly suggest that particle uptake in the presence of beta 2 GPI is coupled to an inhibition of reactive species production by liver macrophages during the respiratory burst, supporting the role of beta 2 GPI as a mediator of senescent cell removal.


Subject(s)
Endocytosis/drug effects , Glycoproteins/pharmacology , Kupffer Cells/drug effects , Liver/drug effects , Animals , Apolipoproteins/pharmacology , Liver/cytology , Luminescent Measurements , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Oxidation-Reduction , Oxidative Stress/drug effects , Phagocytes/drug effects , Phosphatidylserines/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Respiratory Burst , beta 2-Glycoprotein I
8.
Inflamm Res ; 51(7): 351-6, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12146726

ABSTRACT

OBJECTIVES: Kupffer cells, liver macrophages involved in immunomodulation, phagocytosis, and biochemical attack, can induce cytotoxicity and inflammation when their activity is exacerbated. The aim of this study was to evaluate the effects of C-phycocyanin on Kupffer cell functioning considering its antioxidant and anti-inflammatory properties. MATERIALS AND METHODS: Actions of C-phycocyanin on colloidal carbon phagocytosis, carbon-induced respiratory burst activity, and sinusoidal lactate dehydrogenase (LDH) release were studied in isolated perfused mouse liver. The influence of C-phycocyanin on tumor necrosis factor-a (TNF-alpha) and nitrite levels in serum and liver nitric oxide synthase (NOS) activity was assessed in rats subjected to thyroid hormone (T3) administration, a condition known to underlie hepatic oxidative stress comprising an increased Kupffer cell activity. RESULTS: C-phycocyanin elicited a concentration-dependent inhibition of carbon phagocytosis and carbon-induced O2 uptake (IC50 = 0.2 mg/ml) by perfused livers, with a 52% diminution in the carbon-induced sinusoidal release of LDH being found at a concentration of 0.25 mg/ml. Thyroid calorigenesis induced an 82-fold increase in serum TNF-alpha levels, an effect that was suppressed by pretreatment with C-phycocyanin, the antioxidant alpha-tocopherol, and by the Kupffer cell inactivator gadolinium chloride. C-phycocyanin also suppressed the T3-induced increases in serum nitrite levels (234%) and in the activity of hepatic NOS (75%). CONCLUSIONS: C-phycocyanin significantly decreases Kupffer cell phagocytosis and the associated respiratory burst activity, effects that may contribute to the abolition of oxidative stress-induced TNF-alpha response and NO production by hyperthyroid state.


Subject(s)
Hepatocytes/metabolism , Kupffer Cells/metabolism , Liver/metabolism , Oxidative Stress/drug effects , Phycocyanin/pharmacology , Animals , Carbon/metabolism , Enzyme-Linked Immunosorbent Assay , Hepatocytes/drug effects , In Vitro Techniques , Kupffer Cells/drug effects , L-Lactate Dehydrogenase/metabolism , Liver/drug effects , Mice , Nitric Oxide Synthase/metabolism , Nitrites/blood , Oxygen Consumption/drug effects , Rats , Respiratory Burst/drug effects , Triiodothyronine/pharmacology , Tumor Necrosis Factor-alpha/metabolism
9.
Redox Rep ; 6(3): 155-9, 2001.
Article in English | MEDLINE | ID: mdl-11523590

ABSTRACT

The influence of aging on the respiratory activity of stimulated Kupffer cells was investigated in the isolated perfused mouse liver in relation to colloidal carbon phagocytosis, and the content of glutathione (GSH) and protein carbonyls as parameters related to oxidative stress. Livers from aged (22 months) mice exhibited significant 35% and 65% decreases in the carbon uptake and in the carbon-induced O2 consumption compared to young (3 months) animals, respectively, with a concomitant 46% diminution in the carbon-induced O2 consumption/carbon uptake ratio. Hepatic GSH depletion was observed in aged mice compared to young animals, whereas protein oxidation was enhanced. It is concluded that aging leads to an impairment in the functional capacity of Kupffer cells reflected by a substantial reduction in their respiratory burst activity, lessened endocytic capacity and enhanced oxidative stress, that may contribute to increased susceptibility of the liver to noxious challenges.


Subject(s)
Aging/metabolism , Kupffer Cells/physiology , Liver/cytology , Oxygen Consumption , Phagocytosis , Animals , Carbon , Colloids , Glutathione/metabolism , Kupffer Cells/cytology , Male , Mice , Organ Size , Oxidative Stress , Perfusion , Respiratory Burst , Superoxides/metabolism
10.
Toxicol Lett ; 119(2): 87-93, 2001 Feb 28.
Article in English | MEDLINE | ID: mdl-11311569

ABSTRACT

The involvement of cytosolic nitric oxide (NO) and mitochondrial superoxide radical (O2(.-)) production was evaluated as a mechanism triggering liver oxidative stress in lindane (40 mg/kg) or L-3,3',5-triiodothyronine (T3, 0.1 mg/kg for 2 consecutive days) treated animals (male Sprague-Dawley rats) subjected to iron overload (200 mg/kg). Lindane and iron led to 504 and 210% increases in the content of hepatic protein carbonyls as an index of oxidative stress, with a 706% enhancement being produced by their combined administration. T3 did not alter this parameter, whereas iron overload increased the content of protein carbonyls by 116% in hyperthyroid rats. Lindane increased NO generation by 106% without changes in generation of O2(.-), whereas iron enhanced both parameters by 109 and 80% over control values, respectively, with a net 33 and 46% decrease, respectively, being elicited by the combined treatment related to iron overload alone. Hyperthyroidism increased liver NO (69%) and O2(.-) (110%) generation compared to controls, effects that were either synergistically augmented or suppressed by iron overload, respectively. The in vitro addition of iron (1 micromol/mg protein) to liver cytosolic fractions from euthyroid (97%) and hyperthyroid (173%) rats also enhanced NO generation. The effects of iron overload on mitochondrial O2(.-) production by hyperthyroid rats were reproduced by the in vitro addition of 1 micromol iron/mg protein and abolished by the in vivo pretreatment with the iron chelator desferrioxamine (500 mg/kg). It is concluded that liver oxidative stress induced by iron overload is independent of NO and O2(.-) production in lindane-treated rats, whereas in hyperthyroid animals NO generation is a major factor contributing to this redox imbalance.


Subject(s)
Hexachlorocyclohexane/toxicity , Iron/pharmacology , Liver/drug effects , Nitric Oxide/metabolism , Superoxides/metabolism , Thyroid Hormones/toxicity , Animals , Liver/metabolism , Male , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
11.
Toxicol Appl Pharmacol ; 170(1): 23-8, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11141352

ABSTRACT

Parameters related to liver oxidative stress, Kupffer cell function, and hepatocellular injury were assessed in control rats and in animals subjected to lindane (40 mg/kg; 24 h) and/or iron (200 mg/kg; 4 h) administration. Independently of lindane treatment, iron overload enhanced the levels of iron in serum and liver. Biliary efflux of glutathione disulfide increased by 140, 160, or 335% by lindane, iron, or their combined administration, respectively, and the hepatic content of protein carbonyls was elevated by 5.84-, 2.95-, and 10-fold. Colloidal carbon uptake by perfused livers was not modified by lindane and/or iron, whereas gadolinium chloride (GdCl(3)) pretreatment diminished uptake by 60-72%. Carbon-induced liver O(2) uptake was not altered by lindane, whereas iron produced a 61% increase and the combined treatment led to a 72% decrease over control values. Pretreatment with GdCl(3) abolished these effects in all groups. Lindane-treated rats showed acidophilic hepatocytes in periportal areas and some hepatic cells with nuclear pyknosis, whereas iron overload led to moderate hyperplasia and hypertrophy of Kupffer cells and moderate inflammatory cell infiltration. Combined lindane-iron treatment led to hepatocytes with pyknotic nuclei, significant acidophilia, and extensive lymphatic and neutrophil infiltration in the portal space. Hepatic myeloperoxidase activity increased by 1.1-, 2.1-, or 6.7-fold by lindane, iron, or their combined administration, respectively. Liver sinusoidal lactate dehydrogenase efflux increased by 2.2-fold (basal conditions) and 9.7-fold (carbon infusion) in the lindane-iron treated rats, effects that were diminished by 35 and 78% by GdCl(3) pretreatment, respectively. These data support the contention that lindane sensitizes the liver to the damaging effects of iron overload by providing an added enhancement to the oxidative stress status in the tissue, and this may contribute to the alteration of the respiratory activity of Kupffer cells and the development of an inflammatory response.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Hexachlorocyclohexane/toxicity , Insecticides/toxicity , Iron Overload/pathology , Kupffer Cells/drug effects , Oxidative Stress/physiology , Animals , Carbon/pharmacology , Iron/metabolism , Iron/pharmacokinetics , Iron Overload/metabolism , L-Lactate Dehydrogenase/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Peroxidase/metabolism , Phagocytosis/drug effects , Rats , Rats, Sprague-Dawley
12.
Free Radic Res ; 33(3): 313-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10993485

ABSTRACT

To assess the effect of chronic ethanol ingestion in the content of the reduced forms of coenzymes Q9 (ubiquinol-9) and Q10 (ubiquinol-10) as a factor contributing to oxidative stress in liver and brain, male Wistar rats were fed ad libitum a basal diet containing either 10 or 2.5 mg alpha-tocopherol/100 g diet (controls), or the same basal diet plus a 32% ethanol-25% sucrose solution. After three months treatment, ethanol chronically-treated rats showed identical growth rates to the isocalorically pair-fed controls, irrespectively of alpha-tocopherol dietary level. Lowering dietary alpha-tocopherol led to a decreased content of this vitamin in the liver and brain of control rats, without changes in that of ubiquinol-9, and increased levels of hepatic ubiquinol-10 and total glutathione (tGSH), accompanied by a decrease in brain tGSH. At the two levels of dietary alpha-tocopherol, ethanol treatment significantly decreased the content of hepatic alpha-tocopherol and ubiquinols 9 and 10. This effect was significantly greater at 10 mg alpha-tocopherol/100 g diet than at 2.5, whereas those of tGSH were significantly elevated by 43% and 9%, respectively. Chronic ethanol intake did not alter the content of brain alpha-tocopherol and tGSH, whereas those of ubiquinol-9 were significantly lowered by 20% and 14% in rats subjected to 10 and 2.5 mg alpha-tocopherol/100 g diet, respectively. It is concluded that chronic ethanol intake at two levels of dietary alpha-tocopherol induces a depletion of hepatic alpha-tocopherol and ubiquinols 9 and 10, thus contributing to ethanol-induced oxidative stress in the liver tissue. This effect of ethanol is dependent upon the dietary level of alpha-tocopherol, involves a compensatory enhancement in hepatic tGSH availability, and is not observed in the brain tissue, probably due to its limited capacity for ethanol biotransformation and glutathione synthesis.


Subject(s)
Brain/drug effects , Ethanol/administration & dosage , Liver/drug effects , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism , Vitamin E/administration & dosage , Animals , Brain/metabolism , Diet , Glutathione/metabolism , Liver/metabolism , Male , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Wistar
13.
Toxicol Lett ; 115(1): 45-51, 2000 Apr 10.
Article in English | MEDLINE | ID: mdl-10817630

ABSTRACT

Lindane administration to rats (60 mg/kg b.w.) led to an enhancement in the oxidative stress status of the liver at 4 h after treatment, characterized by increases in hepatic thiobarbituric acid reactants (TBARS) formation and chemiluminescence, reduced glutathione (GSH) depletion, and diminution in the biliary content and release of GSH. These changes were observed in the absence of changes in either microsomal functions (cytochrome P450 content, NADPH-dependent superoxide radical production, and NADPH-cytochrome P450 reductase or NADPH oxidase activities) or in oxidative stress-related enzymatic activities (superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase, glucose-6-phosphate dehydrogenase, and glutathione-S-transferases), over control values. Phenobarbital (PB) administration (0.1% in drinking water; 15 days) elicited an enhancement in liver microsomal functions, lipid peroxidation, and GSH content, without changes in oxidative stress-related enzymatic activities, except for the elevation in those of glutathione reductase and glutathione-S-transferase, compared to control rats. Lindane given to PB-pretreated rats did not alter liver microsomal functions, lipid peroxidation, glutathione status, or oxidative stress-related enzymatic activities, as compared to PB-pretreated animals. In addition, lindane induced periportal necrosis with hemorrhagic foci in untreated rats, but not in PB-pretreated animals. It is concluded that the early oxidative stress response of the liver to lindane and hepatic injury are suppressed by PB pretreatment via induction of microsomal enzymes in all zones of the hepatic acinus. reserved.


Subject(s)
Hexachlorocyclohexane/poisoning , Liver Diseases/prevention & control , Oxidative Stress/drug effects , Phenobarbital/pharmacology , Acute Disease , Animals , Biotransformation , Chemical and Drug Induced Liver Injury , Excitatory Amino Acid Antagonists/pharmacology , Hexachlorocyclohexane/antagonists & inhibitors , Hexachlorocyclohexane/blood , Lipid Peroxidation/drug effects , Liver/drug effects , Male , Rats , Rats, Wistar
15.
Free Radic Res ; 30(6): 489-98, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10400461

ABSTRACT

Formation of reactive O2 species in biological systems can be accomplished by copper-(II) (Cu2+) catalysis, with the consequent cytotoxic response. We have evaluated the influence of Cu2+ on the respiratory activity of Kupffer cells in the perfused liver after colloidal carbon infusion. Studies were carried out in untreated rats and in animals pretreated with the Kupffer cell inactivator gadolinium chloride (GdCl3) or with the metallothionein (MT) inducing agent zinc sulphate, and results were correlated with changes in liver sinusoidal efflux of lactate dehydrogenase (LDH) as an index of hepatotoxicity. In the concentration range of 0.1-1 microM, Cu2+ did not modify carbon phagocytosis by Kupffer cells, whereas the carbon-induced liver O2 uptake showed a sigmoidal-type kinetics with a half-maximal concentration of 0.23 microM. Carbon-induced O2 uptake occurred concomitantly with an increased LDH efflux, effects that were significantly correlated and abolished by GdCl3 pretreatment or by MT induction. It is hypothesized that Cu2+ increases Kupffer cell-dependent O2 utilization by promotion of the free radical processes related to the respiratory burst of activated liver macrophages, which may contribute to the concomitant development of hepatocellular injury.


Subject(s)
Carbon/pharmacology , Copper/pharmacology , Kupffer Cells/metabolism , Liver/metabolism , Oxygen Consumption , Povidone/pharmacology , Animals , Carbon/metabolism , Carbon/toxicity , Cell Survival/drug effects , Copper/antagonists & inhibitors , Copper/toxicity , Dose-Response Relationship, Drug , Drug Combinations , Gadolinium/pharmacology , Glutathione/analysis , Kinetics , Kupffer Cells/cytology , Kupffer Cells/drug effects , L-Lactate Dehydrogenase/metabolism , Liver/chemistry , Liver/cytology , Liver/drug effects , Male , Metallothionein/analysis , Perfusion , Phagocytosis/drug effects , Povidone/metabolism , Povidone/toxicity , Rats , Rats, Sprague-Dawley , Respiratory Burst , Zinc Sulfate/pharmacology
16.
Toxicol Lett ; 106(2-3): 209-14, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10403665

ABSTRACT

The influence of thyroid hormone administration (daily doses of 0.1 mg of 3,3',5-triiodothyranine (T3)/kg for 1-3 consecutive days) on rat liver protein oxidation was investigated in relation to the calorigenic and lipid peroxidative actions of the hormone. T3 treatment elicited a progressive enhancement in the serum levels of the hormone, the rectal temperature of the animals, and in the rate of O2 uptake of the liver, changes that are significantly correlated and evidence the development of thyroid calorigenesis. Liver lipid peroxidation was augmented by T3 administration as determined by the tissue content of thiobarbituric acid reactants, with a maximal effect (3.1-fold increase) being found at 2 days after treatment, whereas protein oxidation measured by the content of protein hydrazone derivatives exhibited a maximal 88% increase at 3 days. Maximal rates of lipid peroxidation occur at 1 day after the administration of T3, whereas those of protein oxidation are attained after treatment with three daily doses of T3, time at which the former levels off. It is concluded that T3 administration induces a substantial enhancement in hepatic protein oxidation, in addition to lipid peroxidation, that seems to be due to the higher oxidative stress status conditioned in the liver by thyroid calorigenesis. Both processes exhibit a differential time course of changes, that may represent differences in the susceptibility of target molecules to free radical attack and/or in the efficiency of repair mechanisms.


Subject(s)
Lipid Peroxidation/drug effects , Liver/drug effects , Oxidative Stress/drug effects , Proteins/metabolism , Triiodothyronine/pharmacology , Animals , Female , Liver/metabolism , Oxidation-Reduction , Oxygen Consumption/drug effects , Rats , Rats, Sprague-Dawley
17.
J Neurosci Res ; 56(6): 620-31, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10374817

ABSTRACT

This work addresses the differential effects of several oxidative insults on two neuronal cell lines, PC12 and Neuro 2a cells, extensively used as neuronal models in vitro. We measured cellular damage using the cytotoxic assays for MTT reduction and LDH release and found that acetylcholinesterase (AChE)-amyloid-beta-peptide (Abeta) complexes, Abeta25-35 fragment, glutamate and H2O2 were over 200-fold more toxic to PC12 than to Neuro 2a cells. 17alpha and 17beta estradiol were able to protect both cell types from damage caused by H2O2 or glutamate. By contrast, other insults not related to oxidative stress, such as those caused by the nonionic detergent Triton X-100 and serum deprivation, induced a similar level of damage in both PC12 and Neuro 2a cells. Considering that the Abeta peptide, H2O2 and glutamate are cellular insults that cause an increase in reactive oxygen species (ROS), the intracellular levels of the antioxidant compound, glutathione were verified. Neuro 2a cells were found to have 4- to 5-fold more glutathione than PC12 cells. Our results suggest that Neuro 2a cells are less susceptible to exposure to AChE-Abeta complexes, Abeta25-35 fragment, glutamate and H2O2 than PC12 cells, due to higher intracellular levels of antioxidant defense factors.


Subject(s)
Acetylcholinesterase/toxicity , Amyloid beta-Peptides/toxicity , Cell Survival/drug effects , Neurons/drug effects , Peptide Fragments/toxicity , Acetylcholinesterase/isolation & purification , Animals , Brain/enzymology , Cattle , Culture Media, Serum-Free , Estradiol/pharmacology , Glutamic Acid/toxicity , Glutathione/metabolism , Hydrogen Peroxide/toxicity , Kinetics , Mice , Neuroblastoma , Neurons/cytology , Neurons/metabolism , Octoxynol/pharmacology , PC12 Cells , Rats , Tumor Cells, Cultured
18.
Arch Med Res ; 30(1): 14-8, 1999.
Article in English | MEDLINE | ID: mdl-10071419

ABSTRACT

BACKGROUND: The effect of bromoethylamine (BEA) administration on lipid peroxidation and on the activities of antioxidant enzymes was studied. METHODS: Adult rats received BEA at 1.2 mmol/kg, a dose that produces renal papillary necrosis. Lipid peroxidation assessed by maximal rate in MDA formation, the activities of catalase, superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), and the levels of non-protein sulfhydryls (NPSH) were measured in renal cortex and papilla of control and BEA-treated animals. RESULTS: After BEA treatment, an increment in lipid peroxidation in papilla and cortex was found after 1.5 and 24 hours of treatment. Catalase activity decreased in both regions, but earlier in cortex. CONCLUSION: These data suggest some role of oxidative stress in the mechanism of BEA-induced papillary necrosis.


Subject(s)
Antioxidants/metabolism , Catalase/metabolism , Ethylamines/toxicity , Glutathione Peroxidase/metabolism , Kidney Papillary Necrosis/chemically induced , Lipid Peroxidation , Superoxide Dismutase/metabolism , Animals , Female , Kidney Cortex/enzymology , Kidney Medulla/enzymology , Kidney Papillary Necrosis/enzymology , Malondialdehyde/analysis , Organ Specificity , Oxidative Stress , Rats , Rats, Sprague-Dawley , Sulfhydryl Compounds/analysis
19.
J Biochem Mol Toxicol ; 13(1): 47-52, 1999.
Article in English | MEDLINE | ID: mdl-9890447

ABSTRACT

Administration of bromoethylamine (BEA, 1.2 mmol/kg) to fed rats induced a significant diminution in the activity of hepatic superoxide dismutase (at 1 h after treatment), catalase, and glutathione peroxidase and in the content of nonprotein sulfhydryls (at 15 h after treatment). The content of thiobarbituric acid reactants by the liver was enhanced by 1.9 times over control values (at 3 h). Light microscopy studies revealed that BEA (72 h after treatment) induced periportal fatty accumulation, focal liver cell necrosis, and diffuse inflammatory infiltrates, in addition to hypertrophic Kupffer cells and mitotic hepatocytes. Also, hypertrophic middle tunic or hypertrophic smooth muscle layers of arterioles was observed in the periportal space, with dilated sinusoidal capillaries and free macrophage infiltration. It is concluded that BEA induces a derangement in the antioxidant status of the liver with the consequent lipid peroxidation response, which may constitute a significant hepatotoxic mechanism of the haloaklylamine.


Subject(s)
Catalase/metabolism , Ethylamines/toxicity , Glutathione Peroxidase/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Superoxide Dismutase/metabolism , Animals , Antioxidants/metabolism , Arterioles/drug effects , Arterioles/pathology , Fatty Acids/metabolism , Female , Hypertrophy , Inflammation , Kupffer Cells/drug effects , Kupffer Cells/pathology , Liver/pathology , Liver Circulation/drug effects , Mitosis/drug effects , Necrosis , Rats , Rats, Sprague-Dawley , Time Factors
20.
Redox Rep ; 4(5): 243-50, 1999.
Article in English | MEDLINE | ID: mdl-10731099

ABSTRACT

Liver oxidative stress, Kupffer cell functioning, and cell injury were studied in control rats and in animals subjected to L-3,3',5-tri-iodothyronine (T3) and/or acute iron overload. Thyroid calorigenesis with increased rates of hepatic O2 uptake was not altered by iron treatment, whereas iron enhanced serum and liver iron levels independently of T3. Liver thiobarbituric acid reactants formation increased by 5.8-, 5.7-, or 11.0-fold by T3, iron, or their combined treatment, respectively. Iron enhanced the content of protein carbonyls independently of T3 administration, whereas glutathione levels decreased in T3- and iron-treated rats (54%) and in T3Fe-treated animals (71%). Colloidal carbon infusion into perfused livers elicited a 109% and 68% increase in O2 uptake in T3 and iron-treated rats over controls. This parameter was decreased (78%) by the joint T3Fe administration and abolished by gadolinium chloride (GdCl3) pretreatment in all experimental groups. Hyperthyroidism and iron overload did not modify the sinusoidal efflux of lactate dehydrogenase, whereas T3Fe-treated rats exhibited a 35-fold increase over control values, with a 54% reduction by GdCl3 pretreatment. Histological studies showed a slight increase in the number or size of Kupffer cells in hyperthyroid rats or in iron overloaded animals, respectively. Kupffer cell hypertrophy and hyperplasia with presence of inflammatory cells and increased hepatic myeloperoxidase activity were found in T3Fe-treated rats. It is concluded that hyperthyroidism increases the susceptibility of the liver to the toxic effects of iron, which seems to be related to the development of a severe oxidative stress status in the tissue, thus contributing to the concomitant liver injury and impairment of Kupffer cell phagocytosis and particle-induced respiratory burst activity.


Subject(s)
Hyperthyroidism/metabolism , Iron Overload/metabolism , Kupffer Cells/metabolism , Oxidative Stress , Animals , Hyperthyroidism/pathology , Iron Overload/pathology , Kupffer Cells/pathology , Male , Oxygen Consumption , Rats , Rats, Sprague-Dawley , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...