Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
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.
Cells ; 12(9)2023 04 27.
Article in English | MEDLINE | ID: mdl-37174672

ABSTRACT

The mitochondrial permeability transition pore (MPTP) is a calcium-dependent, ion non-selective membrane pore with a wide range of functions. Although the MPTP has been studied for more than 50 years, its molecular structure remains unclear. Short-term (reversible) opening of the MPTP protects cells from oxidative damage and enables the efflux of Ca2+ ions from the mitochondrial matrix and cell signaling. However, long-term (irreversible) opening induces processes leading to cell death. Ca2+ ions, reactive oxygen species, and changes in mitochondrial membrane potential regulate pore opening. The sensitivity of the pore to Ca2+ ions changes as an organism ages, and MPTP opening plays a key role in the pathogenesis of many diseases. Most studies of the MPTP have focused on elucidating its molecular structure. However, understanding the mechanisms that will inhibit the MPTP may improve the treatment of diseases associated with its opening. To evaluate the functional state of the MPTP and its inhibitors, it is therefore necessary to use appropriate methods that provide reproducible results across laboratories. This review summarizes our current knowledge of the function and regulation of the MPTP. The latter part of the review introduces two optimized methods for evaluating the functional state of the pore under standardized conditions.


Subject(s)
Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Calcium/metabolism , Mitochondria/metabolism , Cell Death
4.
Arch Physiol Biochem ; 128(4): 985-992, 2022 Aug.
Article in English | MEDLINE | ID: mdl-32208934

ABSTRACT

CONTEXT: Monoestolides belonging to the fatty acid-hydroxy fatty acid (FAHFA) family have recently emerged as promising insulin sensitizers. OBJECTIVE: To investigate and compare impact of two selected FAHFA isomers, namely 9-hexadecanoyloxy-octadecanoic acid [9-PAHSA] and 9-(9Z-octadecenoyloxy)-octadecanoic acid [9-OAHSA], on intact livers in C57BL/6J mice. MATERIALS AND METHODS: Short-term in vivo study with intragastric gavage of 13 mg/kg of substances. Morphological, biochemical and high-resolution respirometric assessment of plasma and liver tissue or homogenates thereof. RESULTS: The 9-OAHSA-gavaged mice had the highest final total body weight, the lowest free fatty acid circulating levels and the highest plasma activities of both ALT and AST. No significant changes of ambient glycaemia were found, however 9-PAHSA-gavaged mice tended to have lower glycaemia than other animals. Respirometry proved no substance-dependent differences. DISCUSSION AND CONCLUSION: 9-PAHSA was more metabolically beneficial and less hepatotoxic than 9-OAHSA. Bioenergetic machinery of liver homogenates seemed unaffected at our FAHFA dose.


Subject(s)
Fatty Acids , Insulin , Animals , Liver , Mice , Mice, Inbred C57BL
5.
J Vis Exp ; (174)2021 08 13.
Article in English | MEDLINE | ID: mdl-34459820

ABSTRACT

Mitochondrial substrate flux is a distinguishing characteristic of each cell type, and changes in its components such as transporters, channels, or enzymes are involved in the pathogenesis of several diseases. Mitochondrial substrate flux can be studied using intact cells, permeabilized cells, or isolated mitochondria. Investigating intact cells encounters several problems due to simultaneous oxidation of different substrates. Besides, several cell types contain internal stores of different substrates that complicate results interpretation. Methods such as mitochondrial isolation or using permeabilizing agents are not easily reproducible. Isolating pure mitochondria with intact membranes in sufficient amounts from small samples is problematic. Using non-selective permeabilizers causes various degrees of unavoidable mitochondrial membrane damage. Recombinant perfringolysin O (rPFO) was offered as a more appropriate permeabilizer, thanks to its ability to selectively permeabilize plasma membrane without affecting mitochondrial integrity. When used in combination with microplate respirometry, it allows testing the flux of several mitochondrial substrates with enough replicates within one experiment while using a minimal number of cells. In this work, the protocol describes a method to compare mitochondrial substrate flux of two different cellular phenotypes or genotypes and can be customized to test various mitochondrial substrates or inhibitors.


Subject(s)
Bacterial Toxins , Cell Respiration , Bacterial Toxins/metabolism , Hemolysin Proteins/metabolism , Mitochondria/metabolism , Oxygen Consumption
6.
Int J Mol Sci ; 22(13)2021 Jun 27.
Article in English | MEDLINE | ID: mdl-34199098

ABSTRACT

Mitochondria play an essential role in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Previously, we found that succinate-activated respiration was the most affected mitochondrial parameter in mice with mild NAFLD. In this study, we focused on the role of succinate dehydrogenase (SDH) in NAFLD pathogenesis. To induce the progression of NAFLD to nonalcoholic steatohepatitis (NASH), C57BL/6J mice were fed a Western-style diet (WD) or control diet for 30 weeks. NAFLD severity was evaluated histologically and the expression of selected proteins and genes was assessed. Mitochondrial respiration was measured by high-resolution respirometry. Liver redox status was assessed using glutathione, malondialdehyde, and mitochondrial production of reactive oxygen species (ROS). Metabolomic analysis was performed by GC/MS. WD consumption for 30 weeks led to reduced succinate-activated respiration. We also observed decreased SDH activity, decreased expression of the SDH activator sirtuin 3, decreased gene expression of SDH subunits, and increased levels of hepatic succinate, an important signaling molecule. Succinate receptor 1 (SUCNR1) gene and protein expression were reduced in the livers of WD-fed mice. We did not observe signs of oxidative damage compared to the control group. The changes observed in WD-fed mice appear to be adaptive to prevent mitochondrial respiratory chain overload and massive ROS production.


Subject(s)
Diet, Western , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Oxidation-Reduction , Oxidative Stress , Succinic Acid/metabolism , Animals , Apoptosis , Biomarkers , Cell Respiration , Disease Models, Animal , Disease Susceptibility , Fibrosis , Metabolome , Metabolomics/methods , Mice , Non-alcoholic Fatty Liver Disease/pathology , Succinate Dehydrogenase/metabolism
7.
Acta Medica (Hradec Kralove) ; 64(1): 1-7, 2021.
Article in English | MEDLINE | ID: mdl-33855952

ABSTRACT

Non-Alcoholic Fatty Liver Disease (NAFLD) is one of the most important causes of liver disease worldwide leading the foreground cause of liver transplantation. Recently miRNAs, small non-coding molecules were identified as an important player in the negative translational regulation of many protein-coding genes involved in hepatic metabolism. Visceral adipose tissue was found to take part in lipid and glucose metabolism and to release many inflammatory mediators that may contribute to progression of NAFLD from simple steatosis to Non-Alcoholic SteatoHepatitis. Since visceral adipose tissue enlargement and dysregulated levels of miRNAs were observed in patients with NAFLD, the aim of this paper is to reflect the current knowledge of the role of miRNAs released from visceral adipose tissue and NAFLD.


Subject(s)
Intra-Abdominal Fat/metabolism , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Humans
8.
Biochem Pharmacol ; 176: 113902, 2020 06.
Article in English | MEDLINE | ID: mdl-32156660

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and the fourth most frequent cause of cancer-related death worldwide. Sorafenib is the first line recommended therapy for patients with locally advanced/metastatic HCC. The low response rate is attributed to intrinsic resistance of HCC cells to Sorafenib. The potential resistance to Sorafenib-induced cell death is multifactorial and involves all hallmarks of cancer. However, the presence of sub-therapeutic dose can negatively influence the antitumoral properties of the drug. In this sense, the present study showed that the sub-optimal Sorafenib concentration (10 nM) was associated with activation of caspase-9, AMP-activated protein kinase (AMPK), sustained autophagy, peroxisome proliferator-activated receptor-coactivator 1α (PGC-1α) and mitochondrial function in HepG2 cells. The increased mitochondrial respiration by Sorafenib (10 nM) was also observed in permeabilized HepG2 cells, but not in isolated rat mitochondria, which suggests the involvement of an upstream component in this regulatory mechanism. The basal glycolysis was dose dependently increased at early time point studied (6 h). Interestingly, Sorafenib increased nitric oxide (NO) generation that played an inhibitory role in mitochondrial respiration in sub-therapeutic dose of Sorafenib. The administration of sustained therapeutic dose of Sorafenib (10 µM, 24 h) induced mitochondrial dysfunction and dropped basal glycolysis derived acidification, as well as increased oxidative stress and apoptosis in HepG2. In conclusion, the accurate control of the administered dose of Sorafenib is relevant for the potential prosurvival or proapoptotic properties induced by the drug in liver cancer cells.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Mitochondria, Liver/drug effects , Signal Transduction/drug effects , Sorafenib/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Hepatocellular/pathology , Caspase 9/metabolism , Cell Death/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Male , Mitochondria, Liver/metabolism , Nitric Oxide/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats, Wistar
9.
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
10.
Gen Physiol Biophys ; 38(4): 343-352, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31241045

ABSTRACT

Non-alcoholic fatty liver disease and its complications are frequent causes of liver-related morbidity and mortality. Incretin glucagon-like peptide-1 (GLP-1) affects liver functions and metabolism. Although GLP-1 analogues are widely used in clinical practice, information regarding their potential toxic effect on hepatocytes in vitro is missing. Therefore, we evaluated the effect of GLP-1 analogue liraglutide on activity of caspases 3/7, cell viability and oxidative stress in primary cultures of hepatocytes. Primary cultures isolated from male Wistar rats fed a standard (ST1-group, 10% energy from fat) or a high-fat diet (HF-group, 71% fat) for 10 weeks were incubated with liraglutide (0.1-1000 nmol/l) for 24 h. Activities of caspases 3/7 and cellular dehydrogenases (WST-1), lactate dehydrogenase (LDH) leakage and oxidative stress (malondialdehyde concentration and DCFDA assay) were evaluated. HF-groups vs. ST1-groups showed higher caspases activity, LDH leakage and MDA production (p < 0.001) and lower cellular dehydrogenases activity (p < 0.01). Liraglutide induced a dose-dependent decrease of caspases activity in both groups, reduction of oxidative stress in HF-animals and exerted no negative effects on other parameters. In conclusion, GLP-1 analogue liraglutide decreased activity of caspases 3/7, reduced ROS production and didn't exhibit negative effects on cell viability and oxidative stress in primary cultures of hepatocytes isolated from lean and steatotic livers.


Subject(s)
Cell Separation , Fatty Liver/pathology , Hepatocytes/cytology , Hepatocytes/drug effects , Liraglutide/pharmacology , Liver/cytology , Animals , Cells, Cultured , Male , Rats , Rats, Wistar
11.
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
12.
J Bioenerg Biomembr ; 48(4): 363-73, 2016 08.
Article in English | MEDLINE | ID: mdl-27530389

ABSTRACT

A compound with promising anticancer properties, 3-bromopyruvate (3-BP) is a synthetic derivative of a pyruvate molecule; however, its toxicity in non-malignant cells has not yet been fully elucidated. Therefore, we elected to study the effects of 3-BP on primary hepatocytes in monolayer cultures, permeabilized hepatocytes and isolated mitochondria. After a 1-h treatment with 100 µM 3-BP cell viability of rat hepatocytes was decreased by 30 % as measured by the WST-1 test (p < 0.001); after 3-h exposure to ≥200 µM 3-BP lactate dehydrogenase leakage was increased (p < 0.001). Reactive oxygen species production was increased in the cell cultures after a 1-h treatment at concentrations ≥100 µmol/l (p < 0.01), and caspase 3 activity was increased after a 20-h incubation with 150 µM and 200 µM 3-BP (p < 0.001). This toxic effect of 3-BP was also proved using primary mouse hepatocytes. In isolated mitochondria, 3-BP induced a dose- and time-dependent decrease of mitochondrial membrane potential during a 10-min incubation both with Complex I substrates glutamate + malate or Complex II substrate succinate, although this decrease was more pronounced with the latter. We also measured the effect of 3-BP on respiration of isolated mitochondria. ADP-activated respiration was inhibited by 20 µM 3-BP within 10 min. Similar effects were also found in permeabilized hepatocytes of both species.


Subject(s)
Hepatocytes/drug effects , Mitochondrial Diseases/chemically induced , Pyruvates/toxicity , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Hepatocytes/cytology , Hepatocytes/ultrastructure , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria, Liver/drug effects , Mitochondrial Diseases/physiopathology , Pyruvates/pharmacology , Rats , Reactive Oxygen Species/metabolism , Time Factors
13.
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
14.
Oxid Med Cell Longev ; 2016: 7573131, 2016.
Article in English | MEDLINE | ID: mdl-28074116

ABSTRACT

Epigallocatechin gallate (EGCG) is a green tea antioxidant with adverse effects on rat liver mitochondria and hepatocytes at high doses. Here, we assessed whether low doses of EGCG would protect these systems from damage induced by tert-butyl hydroperoxide (tBHP). Rat liver mitochondria or permeabilized rat hepatocytes were pretreated with EGCG and then exposed to tBHP. Oxygen consumption, mitochondrial membrane potential (MMP), and mitochondrial retention capacity for calcium were measured. First, 50 µM EGCG or 0.25 mM tBHP alone increased State 4 Complex I-driven respiration, thus demonstrating uncoupling effects; tBHP also inhibited State 3 ADP-stimulated respiration. Then, the coexposure to 0.25 mM tBHP and 50 µM EGCG induced a trend of further decline in the respiratory control ratio beyond that observed upon tBHP exposure alone. EGCG had no effect on MMP and no effect, in concentrations up to 50 µM, on mitochondrial calcium retention capacity. tBHP led to a decline in both MMP and mitochondrial retention capacity for calcium; these effects were not changed by pretreatment with EGCG. In addition, EGCG dose-dependently enhanced hydrogen peroxide formation in a cell- and mitochondria-free medium. Conclusion. Moderate nontoxic doses of EGCG were not able to protect rat liver mitochondria and hepatocytes from tBHP-induced mitochondrial dysfunction.


Subject(s)
Catechin/analogs & derivatives , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , tert-Butylhydroperoxide/toxicity , Animals , Calcium/metabolism , Catechin/pharmacology , Cells, Cultured , Hepatocytes/cytology , Hepatocytes/metabolism , Hydrogen Peroxide/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Oxygen Consumption/drug effects , Rats , Rats, Wistar
15.
Am J Physiol Gastrointest Liver Physiol ; 309(2): G100-11, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26045616

ABSTRACT

Nonalcoholic fatty liver disease is associated with chronic oxidative stress. In our study, we explored the antioxidant effect of antidiabetic metformin on chronic [high-fat diet (HFD)-induced] and acute oxidative stress induced by short-term warm partial ischemia-reperfusion (I/R) or on a combination of both in the liver. Wistar rats were fed a standard diet (SD) or HFD for 10 wk, half of them being administered metformin (150 mg·kg body wt(-1)·day(-1)). Metformin treatment prevented acute stress-induced necroinflammatory reaction, reduced alanine aminotransferase and aspartate aminotransferase serum activity, and diminished lipoperoxidation. The effect was more pronounced in the HFD than in the SD group. The metformin-treated groups exhibited less severe mitochondrial damage (markers: cytochrome c release, citrate synthase activity, mtDNA copy number, mitochondrial respiration) and apoptosis (caspase 9 and caspase 3 activation). Metformin-treated HFD-fed rats subjected to I/R exhibited increased antioxidant enzyme activity as well as attenuated mitochondrial respiratory capacity and ATP resynthesis. The exposure to I/R significantly increased NADH- and succinate-related reactive oxygen species (ROS) mitochondrial production in vitro. The effect of I/R was significantly alleviated by previous metformin treatment. Metformin downregulated the I/R-induced expression of proinflammatory (TNF-α, TLR4, IL-1ß, Ccr2) and infiltrating monocyte (Ly6c) and macrophage (CD11b) markers. Our data indicate that metformin reduces mitochondrial performance but concomitantly protects the liver from I/R-induced injury. We propose that the beneficial effect of metformin action is based on a combination of three contributory mechanisms: increased antioxidant enzyme activity, lower mitochondrial ROS production, and reduction of postischemic inflammation.


Subject(s)
Antioxidants/pharmacology , Liver/drug effects , Metformin/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Reperfusion Injury/prevention & control , Adenosine Triphosphate/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cytoprotection , Diet, High-Fat , Disease Models, Animal , Energy Metabolism/drug effects , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Male , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Rats, Wistar , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Time Factors
16.
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
17.
World J Gastroenterol ; 20(26): 8364-76, 2014 Jul 14.
Article in English | MEDLINE | ID: mdl-25024595

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the Western world, and it persists at a high prevalence. NAFLD is characterised by the accumulation of triglycerides in the liver and includes a spectrum of histopathological findings, ranging from simple fatty liver through non-alcoholic steatohepatitis (NASH) to fibrosis and ultimately cirrhosis, which may progress to hepatocellular carcinoma. The pathogenesis of NAFLD is closely related to the metabolic syndrome and insulin resistance. Understanding the pathophysiology and treatment of NAFLD in humans has currently been limited by the lack of satisfactory animal models. The ideal animal model for NAFLD should reflect all aspects of the intricate etiopathogenesis of human NAFLD and the typical histological findings of its different stages. Within the past several years, great emphasis has been placed on the development of an appropriate model for human NASH. This paper reviews the widely used experimental models of NAFLD in rats. We discuss nutritional, genetic and combined models of NAFLD and their pros and cons. The choice of a suitable animal model for this disease while respecting its limitations may help to improve the understanding of its complex pathogenesis and to discover appropriate therapeutic strategies. Considering the legislative, ethical, economical and health factors of NAFLD, animal models are essential tools for the research of this disease.


Subject(s)
Disease Models, Animal , Non-alcoholic Fatty Liver Disease , Animals , Disease Progression , Genetic Predisposition to Disease , Humans , Liver/metabolism , Liver/pathology , Liver/physiopathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/physiopathology , Nutritional Status , Phenotype , Rats , Species Specificity
18.
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
19.
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
20.
Can J Physiol Pharmacol ; 92(6): 512-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24853265

ABSTRACT

Epigallocatechin gallate (EGCG) is an antioxidant found in green tea. In this study, male Wistar rats were subjected either to partial hepatectomy (PHx), or a sham operation (LAP). Twenty-four hours after surgery, hepatocytes were isolated and treated with various concentrations of EGCG for up to 72 h. We then measured markers of cell viability, oxidative stress, DNA synthesis, and caspase activity. Morphological criteria, cell viability tests, and albumin synthesis revealed toxicity starting at 10 µmol/L. DNA synthesis was higher in hepatocytes isolated from rats after PHx and inhibited by EGCG. Furthermore, EGCG increased the activity of caspases 3 and 7, seen more in hepatocytes from PHx rats. In conclusion, EGCG at a concentration of 10 µmol/L was toxic for hepatocytes isolated from both PHx and LAP rats.


Subject(s)
Antioxidants/toxicity , Catechin/analogs & derivatives , Hepatectomy , Hepatocytes/drug effects , Animals , Antioxidants/administration & dosage , Caspase 3/metabolism , Caspase 7/metabolism , Catechin/administration & dosage , Catechin/toxicity , Cell Survival/drug effects , DNA/biosynthesis , Dose-Response Relationship, Drug , Hepatocytes/enzymology , Hepatocytes/metabolism , Male , Oxidative Stress/drug effects , Primary Cell Culture , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...