Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Int J Mol Med ; 53(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38186319

ABSTRACT

Although some clinical studies have reported increased mitochondrial respiration in patients with fatty liver and early non­alcoholic steatohepatitis (NASH), there is a lack of in vitro models of non­alcoholic fatty liver disease (NAFLD) with similar findings. Despite being the most commonly used immortalized cell line for in vitro models of NAFLD, HepG2 cells exposed to free fatty acids (FFAs) exhibit a decreased mitochondrial respiration. On the other hand, the use of HepaRG cells to study mitochondrial respiratory changes following exposure to FFAs has not yet been fully explored. Therefore, the present study aimed to assess cellular energy metabolism, particularly mitochondrial respiration, and lipotoxicity in FFA­treated HepaRG and HepG2 cells. HepaRG and HepG2 cells were exposed to FFAs, followed by comparative analyses that examained cellular metabolism, mitochondrial respiratory enzyme activities, mitochondrial morphology, lipotoxicity, the mRNA expression of selected genes and triacylglycerol (TAG) accumulation. FFAs stimulated mitochondrial respiration and glycolysis in HepaRG cells, but not in HepG2 cells. Stimulated complex I, II­driven respiration and ß­oxidation were linked to increased complex I and II activities in FFA­treated HepaRG cells, but not in FFA­treated HepG2 cells. Exposure to FFAs disrupted mitochondrial morphology in both HepaRG and HepG2 cells. Lipotoxicity was induced to a greater extent in FFA­treated HepaRG cells than in FFA­treated HepG2 cells. TAG accumulation was less prominent in HepaRG cells than in HepG2 cells. On the whole, the present study demonstrates that stimulated mitochondrial respiration is associated with lipotoxicity in FFA­treated HepaRG cells, but not in FFA­treated HepG2 cells. These findings suggest that HepaRG cells are more suitable for assessing mitochondrial respiratory adaptations in the developed in vitro model of early­stage NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Hep G2 Cells , Mitochondria , Respiration , Cell Line , Fatty Acids, Nonesterified , Triglycerides
2.
Int J Mol Sci ; 21(3)2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32046101

ABSTRACT

Maladaptation of mitochondrial oxidative flux seems to be a considerable feature of nonalcoholic fatty liver disease (NAFLD). The aim of this work was to induce NAFLD in mice fed a Western-style diet (WD) and to evaluate liver mitochondrial functions. Experiments were performed on male C57BL/6J mice fed with a control diet or a WD for 24 weeks. Histological changes in liver and adipose tissue as well as hepatic expression of fibrotic and inflammatory genes and proteins were evaluated. The mitochondrial respiration was assessed by high-resolution respirometry. Oxidative stress was evaluated by measuring lipoperoxidation, glutathione, and reactive oxygen species level. Feeding mice a WD induced adipose tissue inflammation and massive liver steatosis accompanied by mild inflammation and fibrosis. We found decreased succinate-activated mitochondrial respiration and decreased succinate dehydrogenase (SDH) activity in the mice fed a WD. The oxidative flux with other substrates was not affected. We observed increased ketogenic capacity, but no impact on the capacity for fatty acid oxidation. We did not confirm the presence of oxidative stress. Mitochondria in this stage of the disease are adapted to increased substrate flux. However, inhibition of SDH can lead to the accumulation of succinate, an important signaling molecule associated with inflammation, fibrosis, and carcinogenesis.


Subject(s)
Lipid Peroxidation , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Diet, High-Fat/adverse effects , Glutathione/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Succinate Dehydrogenase/metabolism
3.
Drug Chem Toxicol ; 40(4): 448-456, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27960556

ABSTRACT

CONTEXT: Acetaminophen (APAP) hepatotoxicity is often studied in primary cultures of hepatocytes of various species, but there are only few works comparing interspecies differences in susceptibility of hepatocytes to APAP in vitro. OBJECTIVES: The aim of our work was to compare hepatotoxicity of APAP in rat and mouse hepatocytes in primary cultures. MATERIALS AND METHODS: Hepatocytes isolated from male Wistar rats and C57Bl/6J mice were exposed to APAP for up to 24 h. We determined lactate dehydrogenase (LDH) activity in culture medium, activity of cellular dehydrogenases (WST-1) and activity of caspases 3 in cell lysate as markers of cell damage/death. We assessed content of intracellular reduced glutathione, production of reactive oxygen species (ROS) and malondialdehyde (MDA). Respiration of digitonin-permeabilized hepatocytes was measured by high resolution respirometry and mitochondrial membrane potential (MMP) was visualized (JC-1). RESULTS: APAP from concentrations of 2.5 and 0.75 mmol/L induced a decrease in viability of rat (p < 0.001) and mouse (p < 0.001) hepatocytes (WST-1), respectively. In contrast to rat hepatocytes, there was no activation of caspase-3 in mouse hepatocytes after APAP treatment. Earlier damage to plasma membrane and faster depletion of reduced glutathione were detected in mouse hepatocytes. Mouse hepatocytes showed increased glutamate + malate-driven respiration in state 4 and higher susceptibility of the outer mitochondrial membrane (OMM) to APAP-induced injury. CONCLUSION: APAP displayed dose-dependent toxicity in hepatocytes of both species. Mouse hepatocytes in primary culture however had approximately three-fold higher susceptibility to the toxic effect of APAP when compared to rat hepatocytes.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Cell Membrane/drug effects , Hepatocytes/drug effects , Lipid Peroxidation/drug effects , Mitochondria, Liver/drug effects , Oxidative Stress/drug effects , Animals , Biomarkers/metabolism , Cell Membrane/metabolism , Cell Survival/drug effects , Cells, Cultured , Glutathione/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria, Liver/enzymology , Mitochondria, Liver/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Oxidation-Reduction , Rats, Wistar , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Species Specificity
4.
Acta Medica (Hradec Kralove) ; 59(2): 35-42, 2016.
Article in English | MEDLINE | ID: mdl-27526303

ABSTRACT

AIM: The aim of our study was to assess whether simple steatosis impairs liver regeneration after partial hepatectomy (PHx) in rats. METHODS: Male Sprague-Dawley rats were fed a standard diet (ST-1, 10% kcal fat) and high-fat diet (HFD, 71% kcal fat) for 6 weeks. Then the rats were submitted to 2/3 PHx and animals were sacrificed 24, 48 or 72 h after PHx. Serum biochemistry, respiration of mitochondria in liver homogenate, hepatic oxidative stress markers, selected cytokines and DNA content were measured, and histopathological samples were prepared. Liver regeneration was evaluated by incorporation of bromodeoxyuridine (BrdU) to hepatocyte DNA. RESULTS: HFD induced simple microvesicular liver steatosis. PHx caused elevation of serum markers of liver injury in both groups; however, an increase in these parameters was delayed in HFD group. Hepatic content of reduced glutathione was significantly increased in both groups after PHx. There were no significant changes in activities of respiratory complexes I and II (state 3). Relative and absolute liver weights, total DNA content, and DNA synthesis exerted very similar changes in both ST-1 and HFD groups after PHx. CONCLUSION: PHx-induced regeneration of the rat liver with simple steatosis was not significantly affected when compared to the lean liver.


Subject(s)
Fatty Liver/pathology , Fatty Liver/physiopathology , Hepatectomy , Liver Regeneration/physiology , Animals , Liver/pathology , Liver/physiopathology , Liver Function Tests , Male , Mitochondria, Liver/pathology , Mitochondria, Liver/physiology , Rats , Rats, Sprague-Dawley
5.
Oxid Med Cell Longev ; 2015: 476180, 2015.
Article in English | MEDLINE | ID: mdl-25918582

ABSTRACT

Epigallocatechin-3-gallate (EGCG) is the main compound of green tea with well-described antioxidant, anti-inflammatory, and tumor-suppressing properties. However, EGCG at high doses was reported to cause liver injury. In this study, we evaluated the effect of EGCG on primary culture of rat hepatocytes and on rat liver mitochondria in permeabilized hepatocytes. The 24-hour incubation with EGCG in concentrations of 10 µmol/L and higher led to signs of cellular injury and to a decrease in hepatocyte functions. The effect of EGCG on the formation of reactive oxygen species (ROS) was biphasic. While low doses of EGCG decreased ROS production, the highest tested dose induced a significant increase in ROS formation. Furthermore, we observed a decline in mitochondrial membrane potential in cells exposed to EGCG when compared to control cells. In permeabilized hepatocytes, EGCG caused damage of the outer mitochondrial membrane and an uncoupling of oxidative phosphorylation. EGCG in concentrations lower than 10 µmol/L was recognized as safe for hepatocytes in vitro.


Subject(s)
Catechin/analogs & derivatives , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Animals , Caspase 3/metabolism , Catechin/toxicity , Cell Survival/drug effects , Cells, Cultured , Hepatocytes/cytology , Hepatocytes/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Microscopy, Fluorescence , Mitochondria, Liver/metabolism , Oxidative Phosphorylation/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Tea/chemistry , Tea/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Acta Medica (Hradec Kralove) ; 57(1): 3-8, 2014.
Article in English | MEDLINE | ID: mdl-25006656

ABSTRACT

Literature data support that green tea and its major component epigallocatechin gallate (EGCG) have powerful antioxidant effects. Contrary, hepatotoxicity can be induced by high-dose EGCG. The timing of exposure to green tea in relation to administration of hepatotoxic agent plays an import role too. The aim of our work was a verification of antioxidative effect of EGCG on D-galactosamine-induced injury in primary culture of rat hepatocytes. Hepatocytes were incubated with EGCG at concentrations of 1.25-10 µM and toxic D-galactosamine (GalN) for 24 hrs. Alternatively, hepatocytes were pretreated with EGCG for 24 hrs, and then incubated with EGCG and GalN for further 24 hrs. Cytotoxicity was analysed by lactate dehydrogenase activity, functional capacity by albumin production. Oxidative stress was evaluated from a production of malondialdehyde and glutathione content in the cells. EGCG protected hepatocytes against GalN-induced cytotoxicity but preventive treatment of intact hepatocytes with EGCG was required to diminish the development of hepatocyte injury. Oxidative stress induced in our study seems to overcome the ability of hepatocytes to improve GSH depletion and albumin production. Prolongation of the pretreatment with EGCG could be a promising strategy leading to amelioration of its hepatoprotective effect.


Subject(s)
Antioxidants/pharmacology , Catechin/analogs & derivatives , Galactosamine/pharmacology , Glutathione/pharmacology , Hepatocytes/drug effects , Animals , Catechin/pharmacology , Cell Culture Techniques , Hepatocytes/pathology , Rats
7.
Oxid Med Cell Longev ; 2014: 752506, 2014.
Article in English | MEDLINE | ID: mdl-24847414

ABSTRACT

Oxidative stress and mitochondrial dysfunction play an important role in the pathogenesis of nonalcoholic fatty liver disease and toxic liver injury. The present study was designed to evaluate the effect of exogenous inducer of oxidative stress (tert-butyl hydroperoxide, tBHP) on nonfatty and steatotic hepatocytes isolated from the liver of rats fed by standard and high-fat diet, respectively. In control steatotic hepatocytes, we found higher generation of ROS, increased lipoperoxidation, an altered redox state of glutathione, and decreased ADP-stimulated respiration using NADH-linked substrates, as compared to intact lean hepatocytes. Fatty hepatocytes exposed to tBHP exert more severe damage, lower reduced glutathione to total glutathione ratio, and higher formation of ROS and production of malondialdehyde and are more susceptible to tBHP-induced decrease in mitochondrial membrane potential. Respiratory control ratio of complex I was significantly reduced by tBHP in both lean and steatotic hepatocytes, but reduction in NADH-dependent state 3 respiration was more severe in fatty cells. In summary, our results collectively indicate that steatotic rat hepatocytes occur under conditions of enhanced oxidative stress and are more sensitive to the exogenous source of oxidative injury. This confirms the hypothesis of steatosis being the first hit sensitizing hepatocytes to further damage.


Subject(s)
Hepatocytes/drug effects , Oxidative Stress/drug effects , tert-Butylhydroperoxide/toxicity , Animals , Cells, Cultured , Diet, High-Fat , Glutathione/metabolism , Hepatocytes/metabolism , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
8.
Anal Biochem ; 423(2): 236-40, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22386808

ABSTRACT

Glutathione is an important tripeptide involved in a variety of cellular processes. Thus, precise knowledge of its levels is essential. Glutathione exists in two free forms-reduced and oxidized-and a number of methods exist to measure its levels. The aim of our work was to optimize a spectrofluorometric assay for reduced glutathione based on the reaction between glutathione and o-phthalaldehyde. We found that a change of excitation wavelength to 340 nm and modification of pH to 6.0 enhance sensitivity and specificity of the method (intraassay coefficient of variation CV < 3%, interassay CV = 5.1%, recovery = 98-102%, linearity = 0-1000 µM GSH, calibration R2 = 1.00). We also anticipated possible effect of various amino acids on the fluorescence signal, but no interference was found. We compared the optimized fluorometric method with a popular enzymatic recycling glutathione assay and found very strong correlation of results (r = 0.99, n = 45). We introduce here an optimized fluorometric method possessing sufficient sensitivity and specificity that is comparable to the enzymatic glutathione assay. Because the fluorometric assay procedure is faster and lower in cost, it could be ideal for routine analysis of reduced glutathione levels in a large number of samples.


Subject(s)
Fluorometry , Glutathione/analysis , Enzymes/metabolism , Hydrogen-Ion Concentration , Oxidation-Reduction , o-Phthalaldehyde/chemistry
9.
J Gastroenterol Hepatol ; 27(2): 323-30, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21649732

ABSTRACT

BACKGROUND AND AIM: Acetaminophen overdose is the most frequent cause of acute liver failure. Non-alcoholic fatty liver disease is the most common chronic condition of the liver. The aim was to assess whether non-alcoholic steatosis sensitizes rat liver to acute toxic effect of acetaminophen. METHODS: Male Sprague-Dawley rats were fed a standard diet (ST-1, 10% kcal fat) and high-fat gelled diet (HFGD, 71% kcal fat) for 6 weeks and then acetaminophen was applied in a single dose (1 g/kg body weight). Animals were killed 24, 48 and 72 h after acetaminophen administration. Serum biochemistry, activities of mitochondrial complexes, hepatic malondialdehyde, reduced and oxidized glutathione, triacylglycerol and cholesterol contents, and concentrations of serum and liver cytokines (TNF-α, TGF-ß1) were measured and histopathological samples were prepared. RESULTS: The degree of liver inflammation and hepatocellular necrosis were significantly higher in HFGD fed animals after acetaminophen administration. Serum markers of liver injury were elevated only in acetaminophen treated HFGD fed animals. Concentration of hepatic reduced glutathione and ratio of reduced/oxidized glutathione were decreased in both ST-1 and HFGD groups at 24 h after acetaminophen application. Mild oxidative stress induced by acetaminophen was confirmed by measurement of malondialdehyde. Liver content of TNF-α was not significantly altered, but hepatic TGF-ß1 was elevated in acetaminophen treated HFGD rats. We did not observe acetaminophen-induced changes in activities of respiratory complexes I, II, and IV and activity of caspase-3. CONCLUSION: Liver from rats fed HFGD is more susceptible to acute toxic effect of acetaminophen, compared to non-steatotic liver.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/etiology , Fatty Liver/complications , Liver/drug effects , Animals , Biomarkers/blood , Caspase 3/metabolism , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/pathology , Cholesterol/metabolism , Disease Models, Animal , Disease Susceptibility , Electron Transport Chain Complex Proteins/metabolism , Fatty Liver/blood , Fatty Liver/pathology , Glutathione/metabolism , Glutathione Disulfide/metabolism , Liver/metabolism , Liver/pathology , Male , Malondialdehyde/blood , Necrosis , Non-alcoholic Fatty Liver Disease , Rats , Rats, Sprague-Dawley , Risk Factors , Severity of Illness Index , Time Factors , Transforming Growth Factor beta1/blood , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/blood
10.
Life Sci ; 89(23-24): 827-33, 2011 Dec 05.
Article in English | MEDLINE | ID: mdl-21871903

ABSTRACT

AIMS: Several groups found different impact of erythropoietin (EPO) on liver regeneration. Both pro-proliferative as well as anti-proliferative and non-proliferative activities have been reported using high dosage of EPO. Systemic administration of high doses of this cytokine is a clinical concern due to risk of thrombosis. Herein, we applied EPO in low dosages and investigated whether it can stimulate liver regeneration after liver resection. MAIN METHODS: Parameters of liver regeneration were assessed 3 days after 70% hepatectomy by means of immunochemistry and proteomics. EPO was given twice in low dosages (200 and 600 IU/kg BW). KEY FINDINGS: We showed that EPO facilitated hepatic regeneration in rats. Enhanced hepatocyte proliferation (Ki67, BrdU-positive cells) was observed in all EPO-treated groups. By performing Differential Proteomic analysis, we identified two proteins which resulted sensitive to EPO treatment after hepatectomy: Peroxiredoxin-1 and glutathione S-transferase Mu 1. SIGNIFICANCE: Based on our results, low doses of rhEPO increase the hepatic regenerative capacity after partial hepatectomy in rats by enhancing hepatocyte proliferation and acting on antioxidant enzymes. Both proteins identified by proteomic analysis have not previously been associated with liver regeneration and will aid in the understanding of EPO's regenerative response having clinical implications to treat liver failure.


Subject(s)
Erythropoietin/pharmacology , Liver Regeneration/drug effects , Proteomics , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Electrophoresis, Gel, Two-Dimensional , Erythropoietin/administration & dosage , Hepatectomy , Liver/drug effects , Liver/metabolism , Liver Regeneration/genetics , Male , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Gen Physiol Biophys ; 30(1): 66-74, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21460414

ABSTRACT

Antiinflammatory effect of statins mediated by the reduction of cytokine IL-6 in hepatocytes have been reported. Contrary to beneficial effect, statins can increase susceptibility to mitochondrial dysfunction. Extrahepatic biliary obstruction is associated with oxidative stress, pro-inflammatory response and hepatocyte mitochondrial dysfunction. The aim of our study was to verify the effect of fluvastatin on cholestatic liver injury. Cholestasis was induced in Wistar rats by bile duct ligation. Fluvastatin (1 or 5 mg/kg) was administered after surgery and then daily for 7 days. The dose of 5 mg/kg led to the deterioration of hepatocellular injury. Despite lower production of IL-6, decrease in GSH content, rise of TGFß and inhibition of respiratory complex I in mitochondria were determined. The mRNA expressions of canalicular transporter Mdr1b and basolateral transporter Mrp3 increased in cholestatic liver. Fluvastatin administration then led to the attenuation of this change. Analogously, mRNA expression of conjugative enzyme Ugt1a1 was diminished by fluvastatin administration to cholestatic rats. We can conclude that decrease in the antioxidative status and mitochondrial dysfunction could at least in part participate on the deteriorating effect of fluvastatin. Whether these processes can be a consequence of the alteration in metabolism and transport of potentially toxic substances remains to verify.


Subject(s)
Cholestasis, Intrahepatic/drug therapy , Cholestasis, Intrahepatic/metabolism , Fatty Acids, Monounsaturated/adverse effects , Indoles/adverse effects , Interleukin-6/metabolism , Alanine Transaminase/blood , Alanine Transaminase/drug effects , Alanine Transaminase/metabolism , Alkaline Phosphatase/blood , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/drug effects , Aspartate Aminotransferases/metabolism , Bilirubin/blood , Bilirubin/metabolism , Fluvastatin , Glucuronosyltransferase/drug effects , Glucuronosyltransferase/metabolism , Glutathione/drug effects , Glutathione/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Ligation , Liver/drug effects , Liver/pathology , Male , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism , gamma-Glutamyltransferase/blood , gamma-Glutamyltransferase/drug effects , gamma-Glutamyltransferase/metabolism
12.
J Gastroenterol Hepatol ; 26(10): 1544-51, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21501227

ABSTRACT

BACKGROUND AND AIM: The administration of pravastatin to patients with cholestatic liver disease has suggested the potential of the drug with regard to reducing raised plasma cholesterol and bile acid levels. Information about the mechanisms associated with this effect is lacking. Thus, the aim of the present study is to evaluate pravastatin effects on the liver bile acid and cholesterol homeostasis in healthy and cholestatic rats. METHODS: Control sham-operated and reversibly bile duct-obstructed (BDO) rats were treated with pravastatin (1 or 5 mg/kg) or the vehicle alone for 7 days after surgery. RESULTS: Lower doses of pravastatin reduced bile acid plasma concentrations in cholestatic animals. The effect was associated with reduced liver mRNA expression of Cyp7a1, Cyp8b1, Mrp2, Ugt1a1 and the increased expression of Bsep. In addition, BDO-induced increase in the liver content of cholesterol was normalized by pravastatin. The change was accompanied by the reduced liver expression of Hmg-CoA reductase, LDL receptor, and Acat2, and induced the expression of Abca1 and Mdr2. These changes corresponded with the upregulation of nuclear receptors LXRα and PPARα, and the downregulation of FXR, CAR, SREBP-2 and HNF1α. High doses of pravastatin lacked any positive effects on bile acids and cholesterol homeostasis, and blocked bile formation through the reduction of the biliary excretion of bile acids. CONCLUSIONS: Pravastatin rendered a positive reduction in BDO-induced increases in plasma bile acid concentrations and cholesterol liver content, mainly through the transcriptionally-mediated downregulation of genes involved in the synthesis of these compounds in the liver.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/drug therapy , Cholesterol/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/drug effects , Pravastatin/pharmacology , Animals , Cholestasis/genetics , Cholestasis/metabolism , Chronic Disease , Disease Models, Animal , Gene Expression Regulation, Enzymologic/drug effects , Homeostasis , Liver/metabolism , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Permeability , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Int J Exp Pathol ; 92(4): 281-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21410800

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic condition of the liver in the western world. There is only little evidence about altered sensitivity of steatotic liver to acute toxic injury. The aim of this project was to test whether hepatic steatosis sensitizes rat liver to acute toxic injury induced by thioacetamide (TAA). Male Sprague-Dawley rats were fed ad libitum a standard pelleted diet (ST-1, 10% energy fat) and high-fat gelled diet (HFGD, 71% energy fat) for 6 weeks and then TAA was applied intraperitoneally in one dose of 100 mg/kg. Animals were sacrificed in 24-, 48- and 72-h interval after TAA administration. We assessed the serum biochemistry, the hepatic reduced glutathione, thiobarbituric acid reactive substances, cytokine concentration, the respiration of isolated liver mitochondria and histopathological samples (H+E, Sudan III, bromodeoxyuridine [BrdU] incorporation). Activities of alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase and concentration of serum bilirubin were significantly higher in HFGD groups after application of TAA, compared to ST-1. There were no differences in activities of respiratory complexes I and II. Serum tumour necrosis factor alpha at 24 and 48 h, liver tissue interleukin-6 at 72 h and transforming growth factor ß1 at 24 and 48 h were elevated in TAA-administrated rats fed with HFGD, but not ST-1. TAA-induced centrilobular necrosis and subsequent regenerative response of the liver were higher in HFGD-fed rats in comparison with ST-1. Liver affected by NAFLD, compared to non-steatotic liver, is more sensitive to toxic effect of TAA.


Subject(s)
Carcinogens/toxicity , Fatty Liver/pathology , Liver/drug effects , Liver/pathology , Thioacetamide/toxicity , Animals , Cell Proliferation/drug effects , Cholesterol/metabolism , Cytokines/blood , Dietary Fats/adverse effects , Disease Models, Animal , Electron Transport Complex I/drug effects , Electron Transport Complex I/physiology , Electron Transport Complex II/drug effects , Electron Transport Complex II/physiology , Fatty Liver/blood , Fatty Liver/chemically induced , Liver/metabolism , Male , Non-alcoholic Fatty Liver Disease , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/metabolism , Triglycerides/metabolism
14.
Toxicol In Vitro ; 24(8): 2097-103, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20600801

ABSTRACT

Thioacetamide (TAA) is a hepatotoxin frequently used for experimental purposes which produces centrilobular necrosis after a single dose administration. In spite of the fact that oxidative stress seems to play a very important role in the mechanism of TAA-induced injury, the effect of TAA on hepatocytes in primary culture with respect to the influence on mitochondria has yet to be verified. Hepatocytes were incubated for 24h in a medium containing TAA (0-70 mmol/l). Glutathione content (GSH/GSSG), reactive oxygen species and malondialdehyde formation were assessed as markers of cell redox state. Toxicity was determined by lactate dehydrogenase leakage and WST-1 assay. The functional capacity of hepatocytes was evaluated from albumin and urea production. Mitochondrial metabolism was assessed by measuring mitochondrial membrane potential and oxygen consumption. Our results show that a profound decrease in the GSH level in hepatocytes precedes a sharp rise in endogenous ROS production. ROS production correlates with an increase in lipoperoxidation. Mitochondria are affected by TAA secondarily as a consequence of oxidative stress. Oxidation of the NADH-dependent substrates of respiratory Complex I is significantly more sensitive to the toxic action of TAA than oxidation of the flavoprotein-dependent substrate of Complex II. Mitochondria can also maintain their membrane potential better when they utilize succinate as a respiratory substrate. It appears that GSH should be depleted below a certain critical level in order to cause a marked increase in lipid peroxidation. Mitochondrial injury can then occur and cell death develops.


Subject(s)
Hazardous Substances/toxicity , Liver/drug effects , Thioacetamide/toxicity , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Hepatocytes/metabolism , L-Lactate Dehydrogenase/metabolism , Liver/metabolism , Male , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
15.
Gen Physiol Biophys ; 29(1): 72-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20371883

ABSTRACT

S-adenosylmethionine (SAMe) is a key metabolite regulating growth, differentiation and death of hepatocytes. Experimentally, exogenous SAMe has been documented to attenuate hepatocarcinogenesis. The aim of our study was to evaluate the effect of SAMe on proliferation of hepatocytes that are not cancerously transformed. Partial 2/3 hepatectomy (PH) was performed in rats, control animals underwent laparotomy. SAMe was injected immediately after the surgery and then at 24 h intervals for two days at 10 or 40 mg/kg. The animals were sacrificed 24, 48 and 72 h after operation and the intensity of liver regeneration was evaluated. SAMe treatment at 10 mg/kg was associated with decrease in the synthesis of liver DNA 48 h after PH, however, it was not reflected in DNA content. SAMe treatment at 40 mg/kg led to the reduction of DNA synthesis 72 h after PH followed by the diminution of DNA content. The results have documented the inhibition of the liver regeneration by SAMe that may be mediated by the suppression of liver fat accumulation. Cell GSH level correlating with the growth rate was not affected by SAMe. Prevention from the decrease in the intracellular content of SAMe, as a factor attenuating regeneration remains to be verified.


Subject(s)
Liver Regeneration/drug effects , Liver Regeneration/physiology , Liver/drug effects , Liver/surgery , S-Adenosylmethionine/administration & dosage , Animals , Hepatectomy , Liver/pathology , Male , Rats , Rats, Wistar
16.
Acta Medica (Hradec Kralove) ; 52(2): 69-72, 2009.
Article in English | MEDLINE | ID: mdl-19777870

ABSTRACT

Ca(2+)-induced opening of the mitochondrial permeability transition pore (MPTP) is involved in induction of apoptotic and necrotic processes. We studied sensitivity of MPTP to calcium using the model of Ca(2+)-induced, cyclosporine A-sensitive mitochondrial swelling. Presented data indicate that the extent of mitochondrial swelling (dA520/4 min) induced by addition of 25 microM Ca2+ is seven-fold higher in liver than in heart mitochondria (0.564 +/- 0.08/0.077 +/- 0.01). The extent of swelling induced by 100 microM Ca2+ was in liver tree times higher than in heart mitochondria (0.508 +/- 0.05/ 0.173 +/- 0.02). Cyclosporine A sensitivity showed that opening of the MPTP is involved. We may thus conclude that especially at low Ca2+ concentration heart mitochondria are more resistant to damaging effect of Ca2+ than liver mitochondria. These finding thus support hypothesis that there exist tissue specific strategies of cell protection against induction of the apoptotic and necrotic processes.


Subject(s)
Calcium/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Calcium/pharmacology , Cyclosporine/pharmacology , Ion Channel Gating/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Rats , Rats, Wistar
17.
Arch Toxicol ; 83(4): 363-72, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19020865

ABSTRACT

The aim of the study was to evaluate time course and dose dependence of peroxidative damage induced by tert-butyl hydroperoxide (tBHP) in rat hepatocytes cultured in suspension and in monolayer. At the lowest (0.1 mM) concentration, decrease of cytosolic glutathione and discharge of mitochondrial membrane potential (MMP) could be detected. Significant increases in leakage of lactate dehydrogenase and in malondialdehyde concentrations together with decrease of pyruvate-dependent respiration were detected at higher tBHP concentrations (above 0.5 mM) and after longer periods of incubation. Changes in plasma membrane integrity were observed at 1 mM concentration of tBHP. Succinate-dependent oxidation was most resistant to peroxidative damages. Opening of the mitochondrial permeability transition pore was responsible for the discharge of mitochondria membrane potential. In the presence of cyclosporine A and succinate, the membrane potential could be restored. Our data showed that the most sensitive indicators of the peroxidative damage are changes of cytosolic glutathione concentration and MMP.


Subject(s)
Cytosol/drug effects , Hepatocytes/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/drug effects , Oxidants/toxicity , Oxidative Stress/drug effects , tert-Butylhydroperoxide/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Cytosol/metabolism , Dose-Response Relationship, Drug , Glutathione/metabolism , Hepatocytes/metabolism , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Male , Malondialdehyde/metabolism , Mitochondria, Liver/enzymology , Mitochondria, Liver/physiology , Oxidative Stress/physiology , Oxygen Consumption/drug effects , Rats , Rats, Wistar
18.
Altern Lab Anim ; 35(3): 353-61, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17650955

ABSTRACT

The majority of toxic agents act either fully or partially via oxidative stress, the liver, specifically the mitochondria in hepatocytes, being the main target. Maintenance of mitochondrial function is essential for the survival and normal performance of hepatocytes, which have a high energy requirement. Therefore, greater understanding of the role of mitochondria in hepatocytes is of fundamental importance. Mitochondrial function can be analysed in several basic models: hepatocytes cultured in vitro; mitochondria in permeabilised hepatocytes; and isolated mitochondria. The aim of our study was to use all of these approaches to evaluate changes in mitochondria exposed in vitro to a potent non-specific peroxidating agent, tert-butylhydroperoxide (tBHP), which is known to induce oxidative stress. A decrease in the mitochondrial membrane potential (MMP) was observed in cultured hepatocytes treated with tBHP, as illustrated by a significant reduction in Rhodamine 123 accumulation and by a decrease in the fluorescence of the JC-1 molecular probe. Respiratory Complex I in the mitochondria of permeabilised hepatocytes showed high sensitivity to tBHP, as documented by high-resolution respirometry. This could be caused by the oxidation of NADH and NADPH by tBHP, followed by the disruption of mitochondrial calcium homeostasis, leading to the collapse of the MMP. A substantial decrease in the MMP, as determined by tetraphenylphosphonium ion-selective electrode measurements, also confirmed the dramatic impact of tBHP-induced oxidative stress on mitochondria. Swelling was observed in isolated mitochondria exposed to tBHP, which could be prevented by cyclosporin A, which is evidence for the role of mitochondrial permeability transition. Our results demonstrate that all of the above-mentioned models can be used for toxicity assessment, and the data obtained are complementary.


Subject(s)
Hepatocytes/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondria, Liver/metabolism , Oxidative Stress , Animal Testing Alternatives , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Hepatocytes/pathology , Male , Manometry , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/drug effects , Mitochondrial Swelling/drug effects , Mitochondrial Swelling/physiology , Oxidants/toxicity , Oxygen/analysis , Oxygen/metabolism , Oxygen Consumption , Rats , Rats, Wistar , tert-Butylhydroperoxide/toxicity
19.
Altern Lab Anim ; 35(3): 363-71, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17650956

ABSTRACT

S-adenosylmethionine (SAMe) has been shown to protect hepatocytes from toxic injury, both experimentally-induced in animals and in isolated hepatocytes. The mechanisms by which SAMe protects hepatocytes from injury can result from the pathways of SAMe metabolism. Unfortunately, data documenting the protective effect of SAMe against mitochondrial damage from toxic injury are not widely available. Thioacetamide is frequently used as a model hepatotoxin, which causes in vivo centrilobular necrosis. Even though thioacetamide-induced liver necrosis in rats was alleviated by SAMe, the mechanisms of this protective effect remain to be verified. The aim of our study was to determine the protective mechanisms of SAMe on thioacetamide-induced hepatocyte injury by using primary hepatocyte cultures. The release of lactate dehydrogenase (LDH) from cells incubated with thioacetamide for 24 hours, was lowered by simultaneous treatment with SAMe, in a dose-dependent manner. The inhibitory effect of SAMe on thioacetamide-induced lipid peroxidation paralleled the effect on cytotoxicity. A decrease in the mitochondrial membrane potential, as determined by Rhodamine 123 accumulation, was also prevented. The attenuation by SAMe of thioacetamide-induced glutathione depletion was determined after subsequent incubation periods of 48 and 72 hours. SAMe protects both cytoplasmic and mitochondrial membranes. This effect was more pronounced during the development of thioacetamide-induced hepatocyte injury that was mediated by lipid peroxidation. Continuation of the SAMe treatment then led to a reduction in glutathione depletion, as a potential consequence of an increase in glutathione production, for which SAMe is a precursor.


Subject(s)
Carcinogens/toxicity , Hepatocytes/drug effects , Protective Agents/pharmacology , S-Adenosylmethionine/pharmacology , Thioacetamide/toxicity , Animal Testing Alternatives , Animals , Cell Survival/drug effects , Cells, Cultured , Drug Antagonism , Glutathione/metabolism , Hepatocytes/enzymology , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Necrosis/chemically induced , Necrosis/prevention & control , Rats , Rats, Wistar
20.
J Pharm Biomed Anal ; 43(4): 1382-7, 2007 Mar 12.
Article in English | MEDLINE | ID: mdl-17182211

ABSTRACT

A HPLC method for determination of both reduced (GSH) and oxidized (GSSG) glutathione in plasma, whole blood and rat hepatocytes has been developed and evaluated. Reduced glutathione reacts with orthophthaldehyde (OPA) to form a stable, highly fluorescent tricyclic derivate at pH 8, while GSSG reacts with OPA at pH 12. At measurement of GSSG, GSH was complexed to N-ethylmaleimide. For the separation, reverse phase column Discovery C(18), 150 mm x 4 mm, 5 microm, was used. The mixture of methanol and 25 mM sodium hydrogenphosphate (15:85, v/v), pH 6.0, was used as mobile phase. The analytical performance of this method is satisfactory for both GSH and GSSG. The intra-assay coefficients of variation were 1.8 and 2.1% for whole blood, 2.0 and 1.9% for rat hepatocytes, 4.3 and 5.2% for plasma. The inter-assay coefficients of variation were 5.8 and 6.2% for whole blood, 6.6 and 7.1% for rat hepatocytes, 6.9 and 7.8% for plasma. The recoveries were as follows: 98.2% (CV 3.5%) and 101.5% (CV 4.2%) for whole blood, 99.1% (2.5%) and 102.3 (4.4%) for rat hepatocytes, 94.1% (CV 7.5%) and 103.5 (CV 8.5%) for plasma. The calibration curve was linear in the whole range tested. The limit of detection was 14.0 and 5.6 fmol, respectively. The preliminary reference ranges of reduced and oxidized glutathione in a group of blood donors are (4.69+/-0.93) and (0.28+/-0.12)micromol/gHb for whole blood, (1.82+/-0.55) and (0.154+/-0.044)microM for plasma.


Subject(s)
Chromatography, Liquid/methods , Glutathione Disulfide/blood , Glutathione/blood , Hepatocytes/chemistry , Adult , Aldehydes/chemistry , Animals , Ethylmaleimide/chemistry , Female , Glutathione/chemistry , Glutathione Disulfide/chemistry , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Rats , Rats, Wistar , Spectrometry, Fluorescence , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL