Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Eur J Nutr ; 60(2): 961-973, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32556446

ABSTRACT

PURPOSE: To investigate the novel molecular mechanisms of the antioxidant and anti-inflammatory properties of S-allylmercaptocysteine (SAMC) based on a transcriptomic study in a nonalcoholic steatohepatitis (NASH) rat model METHODS: NASH was induced in Sprague-Dawley rats by feeding with a high fat diet (HFD) for 12 weeks. 200 mg/kg SAMC was fed by oral gavage for 4 weeks from 9 to 12 week. RESULTS: SAMC co-administration attenuated HFD-induced liver injury, including the increased serum ALT, hepatic oxidative stress and inflammation. Transcriptomic analysis revealed that SAMC dramatically induced the XRE- and ARE-driven drug metabolising enzymes (DMEs) including Akr7a3, Akr1b8, and Nqo1. The nuclear translocation of the upstream regulator of xenobiotics metabolism, AHR, and regulator of antioxidant responses, NRF2, were significantly increased by SAMC treatment. Furthermore, SAMC counteracted the effects of HFD on NF-κB/IκB and NLRP3/6 pathways with decreasing protein levels of ASC, cleaved caspase-1, IL-18, and IL-1ß. These results were further verified in another mice NASH model induced by an MCD diet with SAMC co-administration. CONCLUSION: We propose that SAMC triggers AHR/NRF2-mediated antioxidant responses which may further suppress the NLRP3/6 inflammasome pathway and NF-κB activation, contributing to the improvement of NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Pharmaceutical Preparations , Animals , Cysteine/analogs & derivatives , Inflammation/drug therapy , Liver , Mice , NF-E2-Related Factor 2/genetics , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Non-alcoholic Fatty Liver Disease/drug therapy , Rats , Rats, Sprague-Dawley
2.
Histol Histopathol ; 35(8): 833-847, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31975365

ABSTRACT

PURPOSE: This study aimed to investigate the role of cathepsin B dependent autophagy induced by chronic aerobic exercise on a high-fat diet (HFD)-induced nonalcoholic steatohepatitis (NASH) in rats. METHODS: Healthy female (Sprague-Dawley) SD rats (8-10 weeks old; 180g-200g; n=6 per group) were divided into: (1) control group; (2) HFD group; (3) Exercise group; (4) HFD + exercise group. Rats were fed with a normal chow or an HFD for 12 weeks. Rats with exercise ran on a rotarod for 30 min per day from weeks 9-12. RESULTS: Exercise training significantly (1) upregulated the levels of autophagy markers Beclin1, ATG5 and LC3II partly through inhibiting the p-AKT/mTOR pathway; (2) ameliorated HFD-mediated accumulation of fat mass by upregulating ß-oxidation regulator PPAR-α and downregulating fatty acid synthesis marker SREBP-1c via lipophagy; (3) diminished the HFD-induced hepatic pro-inflammatory mediators TNF-α and IL-1ß via NF-κB inactivation; (4) decreased the NASH-induced hepatic apoptotic marker caspase-3 activation caused by the upstream oxidative stress and by cytochrome P450 2E1 (CYP2E1); (5) mitigated the HFD-mediated lysosomal membrane permeabilisation and cathepsin B release partly via the reduction of reactive oxygen species (ROS). CONCLUSIONS: Chronic aerobic exercise reduces oxidative stress/ROS and ROS may cause lysosomal membrane destabilisation and disrupts the autophagic process. The beneficial effect of chronic exercise may further inhibit the process of lysosome membrane permeabilisation and facilitate lysosome fusion with autophagosomes to trigger autophagy. This process may possibly contribute to the inhibition of cathepsin B released into cytosol which further reduces inflammation and mitochondrial-dependent apoptosis.


Subject(s)
Autophagy/physiology , Cathepsin B/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Physical Conditioning, Animal/physiology , Animals , Diet, High-Fat/adverse effects , Female , Non-alcoholic Fatty Liver Disease/etiology , Rats , Rats, Sprague-Dawley
3.
Int J Biol Macromol ; 120(Pt B): 1480-1489, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30266645

ABSTRACT

Lycium barbarum polysaccharides (LBP) are major bioactive constituents of wolfberry which possess several pharmacological effects such as antioxidant and immunomodulatory activities. We aimed to evaluate how LBP attenuated the hepatic injury in a non-alcoholic steatohepatitis (NASH) methionine-choline deficient (MCD) mouse model. NASH was induced in C57BL/6N mice by feeding with MCD diet for 6 weeks. During the experiments, 1 mg/kg LBP was intragastrically fed on a daily basis with or without MCD diet lasting from the 4th to 6th week. Control and vehicle-control (LBP + PBS) were fed with a regular animal chow. LBP significantly ameliorated NASH-induced injuries, including the increase of serum ALT and AST levels, hepatic oxidative stress, fibrosis, inflammation, and apoptosis. The hepatoprotective effects of LBP were accompanied by the attenuation of thioredoxin interacting protein, nod-like receptor protein 3/6 (NLRP3/6) and reduced NF-κB (nuclear factor-kappa B) activity. Vehicle LBP fed mice showed no adverse effect on the liver. In conclusion, the suppression of the NLRP3/6 inflammasome pathway and NF-κB activation may partly contribute to the reduction of the hepatic injury during the progression of NASH by therapeutic LBP treatment.


Subject(s)
Diet/adverse effects , Drugs, Chinese Herbal/pharmacology , Liver/drug effects , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Cell Surface/metabolism , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Choline/analysis , Disease Models, Animal , Female , Fibrosis , Liver/injuries , Liver/metabolism , Liver/pathology , Male , Methionine/analysis , Mice , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects
4.
Sci Rep ; 6: 32447, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27580936

ABSTRACT

Acute liver disease is characterized by inflammation, oxidative stress and necrosis, which can greatly influence the long term clinical outcome and lead to liver failure or cancer. Here, we initially demonstrated the beneficial role of caspase-9-dependent autophagy in acute liver injury. Treatment with caspase-9 inhibitor z-LEHD-FMK in HepG2 cells, AML12 cells and C57BL/b6N mice exacerbated CCl4-induced acute hepatocellular damage, and also down-regulated autophagy markers expression levels, indicating that caspase-9 inhibition may aggravate acute liver damage by suppressing cytoprotective autophagy. CCl4 was used as an acute liver injury inducer which caused oxidative stress and apoptosis through up-regulation of HIF-1α, as well as triggered hepatic inflammation and necroptosis via TLR4/NF-κB pathway. Caspase-9 Thr125 site was firstly phosphorylated by ERK1/2 which subsequently activated the cytoprotective autophagy process to attenuate acute CCl4 injury. Caspase-9 inhibition further aggravated hepatic necroptosis through NF-κB expression, leading to increased pro-inflammatory mediators levels, suggesting a protective role of caspase-9-dependent autophagy in the inflammatory process as well as its possibility being a new therapeutic target for the treatment of acute liver injury.


Subject(s)
Autophagy/genetics , Caspase 9/genetics , Chemical and Drug Induced Liver Injury/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , NF-kappa B/genetics , Toll-Like Receptor 4/genetics , Animals , Autophagy/drug effects , Carbon Tetrachloride , Caspase 9/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Female , Gene Expression Regulation , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Oligopeptides/pharmacology , Oxidative Stress , Phosphorylation , Signal Transduction , Threonine/metabolism , Toll-Like Receptor 4/metabolism
5.
Eur J Nutr ; 55(4): 1465-77, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26133299

ABSTRACT

PURPOSE: We aim to examine whether honey ameliorates hepatic injury in non-alcoholic steatohepatitis (NASH) animal and cell line steatosis models. METHODS: NASH was induced in female Sprague-Dawley rat by 8-week feeding with a high-fat diet. During the experiment, 5 g/kg honey was intragastrically fed daily. Rat normal hepatocyte BRL-3A cell was treated with sodium palmitate (SP) to induce steatosis in the absence or presence of honey pre-treatment or specific siRNA/overexpress plasmid of thioredoxin-interacting protein (TXNIP) or antagonist/agonist of Nod-like receptor protein 3 (NLRP3). RESULTS: Honey significantly improved the high-fat-diet-induced hepatic injury, steatosis, fibrosis, oxidative stress, and inflammation in rats. Honey also inhibited the overexpression of TXNIP and the activation of NLRP3 inflammasome. These effects were replicated in BRL-3A cell line which showed that the down-regulation of TXNIP or inhibition of NLRP3 contributed to the suppression of NLRP3 inflammasome activation, inflammation, and re-balanced lipid metabolism. In contrast, overexpression of TXNIP or agonism of NLRP3 exacerbated the cellular damage induced by SP. CONCLUSION: Suppression of the TXNIP-NLRP3 inflammasome pathway may partly contribute to the amelioration of hepatic injury during the progression of NASH by honey. Targeting hepatic TXNIP-NLRP3 inflammasome pathway is a potential therapeutic way for the prevention and treatment of NASH.


Subject(s)
Carrier Proteins/metabolism , Honey , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/diet therapy , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line , Diet, High-Fat , Down-Regulation , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Inflammasomes/genetics , Inflammation/diet therapy , Lipid Metabolism/drug effects , Liver , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Non-alcoholic Fatty Liver Disease/chemically induced , Oxidative Stress/drug effects , Palmitic Acid/administration & dosage , Palmitic Acid/toxicity , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
6.
Hepatobiliary Pancreat Dis Int ; 14(2): 139-44, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25865685

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) refers to any fatty liver disease that is not due to excessive use of alcohol. NAFLD probably results from abnormal hepatic lipid metabolism and insulin resistance. Aerobic exercise is shown to improve NAFLD. This review aimed to evaluate the molecular mechanisms involved in the beneficial effects of aerobic exercise on NAFLD. DATA SOURCE: We searched articles in English on the role of aerobic exercise in NAFLD therapy in PubMed. RESULTS: The mechanisms of chronic aerobic exercise in regulating the outcome of NAFLD include: (i) reducing intrahepatic fat content by down-regulating sterol regulatory element-binding protein-1c and up-regulating peroxisome proliferator-activated receptor gamma expression levels; (ii) decreasing hepatic oxidative stress through modulating the reactive oxygen species, and enhancing antioxidant enzymes such as catalase and glutathione peroxidase; (iii) ameliorating hepatic inflammation via the inhibition of pro-inflammatory mediators such as tumor necrosis factor-alpha and interleukin-1 beta; (iv) attenuating mitochondrial dependent apoptosis by reducing cytochrome C released from the mitochondria to the cytosol; and (v) inducing hepato-protective autophagy. CONCLUSION: Aerobic exercise, via different mechanisms, significantly decreases the fat content of the liver and improves the outcomes of patients with NAFLD.


Subject(s)
Exercise/physiology , Lipid Metabolism/physiology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Apoptosis , Autophagy , Cytochromes c/metabolism , Down-Regulation , Hepatitis/prevention & control , Humans , Interleukin-1beta/antagonists & inhibitors , Intra-Abdominal Fat , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/enzymology , Oxidative Stress , Reactive Oxygen Species/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors
7.
Toxicol Sci ; 143(2): 430-40, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25432964

ABSTRACT

Cyclooxygenase-1 (COX-1) is the constitutive form of the COX enzyme family, which produces bioactive lipids called prostanoids. Although the role of COX-2 in liver diseases has been studied, little is known about the function of COX-1 in liver injury. We aimed to investigate the role and mechanism of COX-1 in acute liver injury. Carbon tetrachloride (CCl(4)) was administered to induce acute liver injury in wild-type or COX-1-deficient mice. Both genetic (partially or completely) deletion of COX-1 expression and pharmacological inhibition of COX-1 activity in mice exacerbated acute liver injury induced by CCl(4), revealing the (1) histopathological changes and increased serum levels of aminotransferases; (2) oxidative stress in the liver partly through the action of cytochrome P450 2E1-dependent pathway; (3) enhanced inflammatory and chemoattractive responses with increased number of activated macrophages; and (4) increased apoptosis through both intrinsic and extrinsic apoptotic pathways. These pathological changes were partly through the modulation of transcription factor-dependent pathways (eg, NF-κB and C/EBP-α). Pre-treatment with prostaglandin E2 (PGE(2)) or 5-lipoxygenase (5-LO) inhibitor in homozygous COX-1 knockout mice significantly ameliorated CCl(4)-induced hepatic injury. In addition, level of hepato-protective molecules (eg, OSM and OSMR) and associated liver regeneration pathway were significantly inhibited by the deficiency of COX-1 but restored by the addition of PGE(2) or the inhibition of 5-LO. Furthermore, the alternative arachidonic acid metabolism pathway of 5-LO, which induced additional inflammation in the liver, was activated in response to the deficiency of COX-1. In conclusion, basal expression of COX-1 is essential for the protection of liver against chemical-induced hepatotoxicity and required for hepatic homeostatic maintenance.


Subject(s)
Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/prevention & control , Cyclooxygenase 1/genetics , Animals , Apoptosis/drug effects , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Cyclooxygenase 1/deficiency , Cytokines/biosynthesis , Dinoprostone/biosynthesis , Dinoprostone/pharmacology , Heterozygote , Homozygote , In Situ Nick-End Labeling , Liver Function Tests , Mice, Knockout , Oxidative Stress/drug effects
8.
Sci Rep ; 4: 5587, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24998389

ABSTRACT

This study aimed to investigate the possible therapeutic effects and active components of Lycium barbarum polysaccharides (LBP) on a high fat diet-induced NASH rat model. We induced NASH in a rat model by voluntary oral feeding with a high-fat diet ad libitum for 8 weeks. After 8 weeks, 1 mg/kg LBP was orally administered for another 4 weeks with a high-fat diet. When compared with NASH rats treated for 12 weeks, therapeutic LBP treatment for 4 weeks during 12 weeks of NASH induction showed ameliorative effects on: (1) increased body and wet liver weights; (2) insulin resistance and glucose metabolic dysfunction; (3) elevated level of serum aminotransferases; (4) fat accumulation in the liver and increased serum free fatty acid (FFA) level; (5) hepatic fibrosis; (6) hepatic oxidative stress; (7) hepatic inflammatory response; and (8) hepatic apoptosis. These improvements were partially through the modulation of transcription factor NF-κB, MAPK pathways and the autophagic process. In a palmitate acid-induced rat hepatocyte steatosis cell-based model, we also demonstrated that l-arabinose and ß-carotene partially accounted for the beneficial effects of LBP on the hepatocytes. In conclusion, LBP possesses a variety of hepato-protective properties which make it a potent supplementary therapeutic agent against NASH in future clinical trials.


Subject(s)
Anti-Obesity Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Hepatocytes/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Apoptosis , Arabinose/pharmacology , Autophagy , Cell Survival , Cells, Cultured , Diet, High-Fat/adverse effects , Female , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , MAP Kinase Signaling System , Non-alcoholic Fatty Liver Disease/etiology , Obesity/drug therapy , Oxidative Stress , Rats, Sprague-Dawley , beta Carotene/pharmacology
9.
PLoS One ; 9(4): e95214, 2014.
Article in English | MEDLINE | ID: mdl-24740309

ABSTRACT

In the current study, the therapeutic effects of zeaxanthin dipalmitate (ZD) on a rat alcoholic fatty liver disease (AFLD) model were evaluated. After-treatment with ZD from the 5th week to the 10th week in a 10-week ethanol intragastric administration in rats significantly alleviated the typical AFLD symptoms, including reduction in rat body weight, accumulation of hepatic fat droplets, occurrence of oxidative stress, inflammation, chemoattractive responses and hepatic apoptosis in the liver. The reduction of liver function abnormalities by ZD was partly through lower expression level of cytochrome P450 2E1 (CYP2E1), diminished activity of nuclear factor kappa B (NF-κB) through the restoration of its inhibitor kappa B alpha (IκBα), and the modulation of MAPK pathways including p38 MAPK, JNK and ERK. ZD treatment alone did not pose obvious adverse effect on the healthy rat. In the cellular AFLD model, we also confirmed the inhibition of p38 MAPK and ERK abolished the beneficial effects of ZD. These results provide a scientific rationale for the use of zeaxanthin and its derivatives as new complementary agents for the prevention and treatment of alcoholic liver diseases.


Subject(s)
Antioxidants/pharmacology , Ethanol/administration & dosage , Fatty Liver, Alcoholic/drug therapy , Palmitates/pharmacology , Protective Agents/pharmacology , Xanthophylls/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Female , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction , Weight Loss/drug effects , p38 Mitogen-Activated Protein Kinases/genetics
10.
Exp Physiol ; 99(1): 220-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24036592

ABSTRACT

The carotid body (CB) plays an important role in the alteration of cardiorespiratory activity in chronic intermittent hypoxia (IH) associated with sleep-disordered breathing, which may be mediated by local expression of the renin-angiotensin system (RAS). We hypothesized a pathogenic role for IH-induced RAS expression in the CB. The CB expression of RAS components was examined in rats exposed to IH resembling a severe sleep-apnoeic condition for 7 days. In situ hybridization showed an elevated expression of angiotensinogen in the CB glomus cells in the hypoxic group when compared with the normoxic control group. Immunohistochemical studies and Western blot analysis revealed increases in the protein level of both angiotensinogen and angiotensin II type 1 (AT1) receptors in the hypoxic group, which were localized to the glomic clusters containing tyrosine hydroxylase. RT-PCR studies confirmed that levels of the mRNA expression of angiotensinogen, angiotensin-converting enzyme, AT1a and AT2 receptors were significantly increased in the CBs of the hypoxic rats. Functionally, the [Ca(2+)]i response to exogenous angiotensin II was enhanced in fura-2-loaded glomus cells dissociated from hypoxic rats when compared with those of the normoxic control animals. Pretreatment with losartan, but not PD123319, abolished the angiotensin II-induced [Ca(2+)]i response, suggesting an involvement of AT1 receptors. Moreover, daily treatment of the IH group of rats with losartan attenuated the levels of oxidative stress, gp91(phox) expression and macrophage infiltration in the CB. Collectively, the upregulated local RAS expression could play a pathogenic role in the augmented CB activity and local inflammation via AT1 receptor activation during IH conditions in patients with sleep-disordered breathing.


Subject(s)
Carotid Body/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Renin-Angiotensin System/genetics , Up-Regulation/genetics , Angiotensin II/genetics , Angiotensin II/metabolism , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Calcium/metabolism , Fura-2/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidative Stress/genetics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Sleep Apnea Syndromes/genetics , Sleep Apnea Syndromes/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
11.
Eur J Nutr ; 53(1): 187-99, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23515587

ABSTRACT

PURPOSE: To investigate the protective mechanisms of an 85 % pure extract of (-) epigallocatechin gallate (EGCG) in the development of fibrosis, oxidative stress and inflammation in a recently developed dietary-induced animal model of non-alcoholic fatty liver disease (NAFLD). METHODS: Female Sprague-Dawley rats were fed with either normal rat diet or high-fat diet for 8 weeks to develop NAFLD. For both treatments, rats were treated with or without EGCG (50 mg/kg, i.p. injection, 3 times per week). At the end, blood and liver tissue samples were obtained for histology, molecular, and biochemical analyses. RESULTS: Non-alcoholic fatty liver disease (NAFLD) rats showed significant amount of fatty infiltration, necrosis, fibrosis, and inflammation. This was accompanied by a significant expressional increase in markers for fibrosis, oxidative stress, and inflammation. TGF/SMAD, PI3 K/Akt/FoxO1, and NF-κB pathways were also activated. Treatment with EGCG improved hepatic histology (decreased number of fatty score, necrosis, and inflammatory foci), reduced liver injury (from ~0.5 to ~0.3 of ALT/AST ratio), attenuated hepatic changes including fibrosis (reduction in Sirius Red and synaptophysin-positive stain) with down-regulation in the expressions of key pathological oxidative (e.g. nitrotyrosine formation) and pro-inflammatory markers (e.g. iNOS, COX-2, and TNF-α). EGCG treatment also counteracted the activity of TGF/SMAD, PI3 K/Akt/FoxO1, and NF-κB pathways. Treatment with EGCG did not affect the healthy rats. CONCLUSIONS: Epigallocatechin gallate (EGCG) reduced the severity of liver injury in an experimental model of NAFLD associated with lower concentration of pro-fibrogenic, oxidative stress, and pro-inflammatory mediators partly through modulating the activities of TGF/SMAD, PI3 K/Akt/FoxO1, and NF-κB pathways. Therefore, green tea polyphenols and EGCG are useful supplements in the prevention of NAFLD.


Subject(s)
Antioxidants/pharmacology , Catechin/analogs & derivatives , Fatty Liver/drug therapy , Oxidative Stress/drug effects , Signal Transduction , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Catechin/pharmacology , Cyclooxygenase 2/metabolism , Diet, High-Fat , Down-Regulation , Fatty Liver/pathology , Female , Fibrosis , Forkhead Transcription Factors/metabolism , Inflammation/metabolism , Inflammation/pathology , Liver/drug effects , Liver/metabolism , Matrix Metalloproteinase 2/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Non-alcoholic Fatty Liver Disease , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Smad Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Article in English | MEDLINE | ID: mdl-23861709

ABSTRACT

Our previous study demonstrated that administration of garlic-derived antioxidant S-allylmercaptocysteine (SAMC) ameliorated hepatic injury in a nonalcoholic fatty liver disease (NAFLD) rat model. Our present study aimed to investigate the mechanism of SAMC on NAFLD-induced hepatic apoptosis and autophagy. Adult female rats were fed with a high-fat diet for 8 weeks to develop NAFLD with or without intraperitoneal injection of 200 mg/kg SAMC for three times per week. During NAFLD development, increased apoptotic cells and caspase-3 activation were observed in the liver. Increased apoptosis was modulated through both intrinsic and extrinsic apoptotic pathways. NAFLD treatment also enhanced the expression of key autophagic markers in the liver with reduced activity of LKB1/AMPK and PI3K/Akt pathways. Increased expression of proapoptotic regulator p53 and decreased activity of antiautophagic regulator mTOR were also observed. Administration of SAMC reduced the number of apoptotic cells through downregulation of both intrinsic and extrinsic apoptotic mechanisms. SAMC also counteracted the effects of NAFLD on LKB1/AMPK and PI3K/Akt pathways. Treatment with SAMC further enhanced hepatic autophagy by regulating autophagic markers and mTOR activity. In conclusion, administration of SAMC during NAFLD development in rats protects the liver from chronic injury by reducing apoptosis and enhancing autophagy.

13.
Hepatobiliary Pancreat Dis Int ; 12(2): 125-35, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23558065

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver injury and mortality in Western countries and China. However, as to date, there is no direct and effective therapy for this disease. The aim of this review is to analyze the key progress and challenges of main current therapeutic approaches in NAFLD. DATA SOURCE: We carried out a PubMed search of English-language articles relevant to NAFLD therapy. RESULTS: There are two major therapeutic strategies for NAFLD treatment: (1) lifestyle interventions (including weight reduction, dietary modification and physical exercise) and (2) pharmaceutical therapies. Lifestyle interventions, particularly chronic and moderate intensity exercise, are the most effective and recognized clinical therapies for NAFLD. For pharmaceutical therapies, although their effects and mechanisms have been extensively investigated in laboratory studies, they still need further tests and investigations in clinical human trials. CONCLUSION: Future advancement of NAFLD therapy should focus on the mechanistic studies on cell based and animal models and human clinical trials of exercise, as well as the combination of lifestyle intervention and pharmaceutical therapy specifically targeting main signaling pathways related to lipid metabolism, oxidative stress and inflammation.


Subject(s)
Fatty Liver/therapy , Risk Reduction Behavior , Animals , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Diet, Reducing , Disease Progression , Exercise , Fatty Liver/diagnosis , Fatty Liver/drug therapy , Fatty Liver/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Hypolipidemic Agents/therapeutic use , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Renin-Angiotensin System/drug effects , Treatment Outcome
14.
Histol Histopathol ; 28(4): 463-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23322610

ABSTRACT

Chronic liver diseases are commonly associated with tissue hypoxia that may cause inflammation, oxidative stress, liver cell injury and increased nuclear transcriptional regulation. The hepatic response to chronic hypoxia at the molecular level has not yet been clearly understood until now. The aim of this study is to investigate whether nuclear transcription factors [hypoxia-inducible factor-1 (HIF-1α), activator protein-1 (AP-1), nuclear factor-kappa B (NF-κB)] exhibit activity changes during hepatic response to chronic hypoxia. Blood and liver samples were collected from adult Sprague-Dawley rats living in atmospheric air or 10% oxygen for four weeks. Levels of serum alanine aminotransferase (ALT), 8-isoprostane and nitrotyrosine were measured. The activities of nuclear transcription factors and the expression of downstream genes (iNOS, eNOS, ET-1 and VEGF) were measured using RT-PCR, Western blotting and Gel shift analysis. Results showed that serum ALT level, 8-isoprostane level and formation of nitrotyrosine were within normal range at all time-points. In the hypoxic liver, DNA-binding activities of HIF-1α, NF-κB and AP-1 increased significantly. Expression levels of iNOS, VEGF and ET-1 progressively increased from day 7 to day 28. eNOS was also elevated in the hypoxic liver. In conclusion, our study suggests that increased activity of HIF-1α, AP-1 and NF-κB may partly play a significant role in the hepatic response to oxidative stress and liver injury under chronic hypoxia. The increased expression of VEGF, ET-1, iNOS and eNOS may be partly due to the compensatory mechanism in the vascular beds of the liver in response to chronic hypoxia.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Liver/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Alanine Transaminase/blood , Animals , Biomarkers/blood , Blotting, Western , Chronic Disease , Dinoprost/analogs & derivatives , Dinoprost/blood , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Endothelin-1/genetics , Endothelin-1/metabolism , Gene Expression Regulation , Hypoxia/genetics , Male , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/blood , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Alcohol Clin Exp Res ; 37(1): 31-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22676331

ABSTRACT

BACKGROUND: Thromboxane levels are increased in rats fed ethanol (EtOH), whereas thromboxane inhibitors reduce alcoholic liver injury. The aim of this study is to determine whether thromboxane inhibitors could attenuate the already established alcoholic liver injury. METHODS: Rats were fed EtOH and liquid diet for 6 weeks by intragastric infusion to induce liver injury after which EtOH was continued for 2 more weeks, and the rats were treated with either a thromboxane synthase inhibitor (TXSI) or a thromboxane receptor antagonist (TXRA). Liver pathology, lipid peroxidation, nuclear factor-kappa-B (NF-κB) activity, tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2), and transforming growth factor-beta1 (TGF-ß(1) ) were evaluated. RESULTS: Administration of fish oil and EtOH caused fatty liver, necrosis, inflammation and fibrosis accompanied by increased in lipid peroxidation, NF-κB activity, and expression of TNF-α, COX-2, and TGF-ß(1) . Treatment with the thromboxane inhibitors ameliorated a certain level of the pathological and biochemical abnormalities. In particular, TXSI in addition to reducing necrosis, inflammation and fibrosis also decrease the severity of fatty liver. CONCLUSIONS: Thromboxane inhibitors attenuated the alcoholic liver injury, inflammation and fibrotic changes despite continued EtOH administration. Inhibition of the production of thromboxane by thromboxane inhibitor and receptor antagonists may be a useful treatment strategy in clinical alcoholic liver disease.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Ethanol/adverse effects , Fatty Liver, Alcoholic/drug therapy , Oxazoles/therapeutic use , Pyridines/therapeutic use , Receptors, Thromboxane/antagonists & inhibitors , Thromboxane-A Synthase/antagonists & inhibitors , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/adverse effects , Cyclooxygenase 2/metabolism , Endotoxins/blood , Ethanol/administration & dosage , Fatty Liver, Alcoholic/blood , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/pathology , Fibrosis , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/pathology , Male , NF-kappa B/metabolism , Oxazoles/pharmacology , Pyridines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transforming Growth Factor beta1/blood , Tumor Necrosis Factor-alpha/metabolism
16.
Eur J Nutr ; 52(1): 179-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22278044

ABSTRACT

PURPOSE: To investigate the hepato-protective properties and underlying mechanisms of SAMC in a non-alcoholic fatty liver disease (NAFLD) rat model. METHODS: Female rats were fed with a diet comprising highly unsaturated fat diet (30% fish oil) for 8 weeks to develop NAFLD with or without an intraperitoneal injection of 200 mg/kg SAMC three times per week. After euthanasia, blood and liver samples of rats were collected for histological and biochemical analyses. RESULTS: Co-treatment of SAMC attenuated NAFLD-induced liver injury, fat accumulation, collagen formation and free fatty acids (FFAs). At the molecular level, SAMC decreased the lipogenesis marker and restored the lipolysis marker. SAMC also reduced the expression levels of pro-fibrogenic factors and diminished liver oxidative stress partly through the inhibition in the activity of cytochrome P450 2E1-dependent pathway. NAFLD-induced inflammation was also partially mitigated by SAMC treatment via reduction in the pro-inflammatory mediators, chemokines and suppressor of cytokine signaling. The protective effect of SAMC is also shown partly through the restoration of altered phosphorylation status of FFAs-dependent MAP kinase pathways and diminished in the nuclear transcription factors (NF-κB and AP-1) activity during NAFLD development. CONCLUSIONS: SAMC is a novel hepato-protective agent against NAFLD caused by abnormal liver functions. Garlic or garlic derivatives could be considered as a potent food supplement in the prevention of fatty liver disease.


Subject(s)
Cysteine/analogs & derivatives , Fatty Liver/drug therapy , Garlic/chemistry , Liver/drug effects , Plant Extracts/pharmacology , Animals , Blotting, Western , Cysteine/pharmacology , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP2E1 Inhibitors , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Inflammation/drug therapy , Inflammation/pathology , Lipogenesis/drug effects , Liver/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease , Oxidative Stress/drug effects , Phosphorylation , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
17.
J Tradit Complement Med ; 3(2): 88-94, 2013 Apr.
Article in English | MEDLINE | ID: mdl-24716162

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the leading causes of chronic liver injury across the world. It is also strongly related to other pathological conditions, including obesity, diabetes, cardiovascular diseases, and symptoms of metabolic syndrome. Pathogenesis of NAFLD remains not fully characterized but is generally attributed to the occurrence of insulin resistance, lipid metabolism dysfunction,0 oxidative stress, inflammation, and necro-apoptosis. Every potential therapeutic strategy should target one or some of these pathological events in the liver. Over the past decades, application of herbal treatment for NAFLD has received increasing attention due to its wide availability, low side effects, and proven therapeutic mechanisms and benefits. In recent years, some monomers and certain functional mixtures of herbs have been extensively examined for their potential uses in NAFLD treatment. In the present review, we selected several herbal derivatives under intense basic and/or clinical investigations by carrying out a PubMed search of English language articles relevant to herbal derivatives and NAFLD, such as polysaccharide portion of wolfberry, garlic-derived monomers, red grape-derived resveratrol, and milk thistle-derived substances. They have been shown to target the pathological events during NAFLD initiation and progression both in pre-clinical studies and clinical trials. Although more detailed mechanistic researches and long-term clinical evaluations are needed for their future applications, they offer unanticipated and great health benefits without obvious adverse effects in NAFLD therapy.

18.
Food Chem Toxicol ; 50(3-4): 861-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22107987

ABSTRACT

We investigated the protective effects of two non-steroid anti-inflammatory drugs, indomethacin (COX-1 and COX-2 inhibitors) and nimesulide (specific COX-2 inhibitor) on the hepatic injury induced by lipopolysaccharide in d-galactosamine sensitized (Gal/LPS) mice. ICR male mice were injected with a single dose of Gal/LPS with or without pre-treatment of 3mg/kg indomethacin or 30 mg/kg nimesulide (single i.p. injection). Sixteen hours later, blood and liver tissues of mice were collected for histological, molecular, and biochemical analyses. Our results showed marked reduction of hepatic necrosis, serum ALT, and tissue TBARS levels in both indomethacin- and nimesulide-pre-treated mice when compared with Gal/LPS-treated mice. Western blot and RT-PCR analysis showed decreased levels of iNOS mRNA, iNOS protein, and nitrotyrosine formation in both COX inhibitor pre-treated groups when compared with Gal/LPS-treated group. There was an inverse relationship between COX-1 and COX-2 expressions, as well as between COX-2 and C/EBP-α expressions in COX inhibitors groups, Gal/LPS and control groups. COX inhibitors reduced the expression of TNF-α mRNA and the activity of NF-κB which were elevated by Gal/LPS treatment. We conclude that COX inhibitors protected the liver from Gal/LPS-induced hepatotoxicity. COX inhibitors could be considered as potential agents in the prevention of acute liver failure and sepsis.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Galactosamine/toxicity , Lipopolysaccharides/toxicity , Alanine Transaminase/blood , Animals , Blotting, Western , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/enzymology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred ICR , Nitric Oxide Synthase Type II/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thiobarbituric Acid Reactive Substances/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
Eur J Nutr ; 51(3): 323-33, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21681437

ABSTRACT

PURPOSE: To study the protective effects and underlying molecular mechanisms of SAMC on carbon tetrachloride (CCl4)-induced acute hepatotoxicity in the mouse model. METHODS: Mice were intraperitoneally injected with CCl4 (50 µl/kg; single dose) to induce acute hepatotoxicity with or without a 2-h pre-treatment of SAMC intraperitoneal injection (200 mg/kg; single dose). After 8 h, the blood serum and liver samples of mice were collected and subjected to measurements of histological and molecular parameters of hepatotoxicity. RESULTS: SAMC reduced CCl4-triggered cellular necrosis and inflammation in the liver under histological analysis. Since co-treatment of SAMC and CCl4 enhanced the expressions of antioxidant enzymes, reduced the nitric oxide (NO)-dependent oxidative stress, and inhibited lipid peroxidation induced by CCl4. SAMC played an essential antioxidative role during CCl4-induced hepatotoxicity. Administration of SAMC also ameliorated hepatic inflammation induced by CCl4 via inhibiting the activity of NF-κB subunits p50 and p65, thus reducing the expressions of pro-inflammatory cytokines, mediators, and chemokines, as well as promoting pro-regenerative factors at both transcriptional and translational levels. CONCLUSIONS: Our results indicate that SAMC mitigates cellular damage, oxidative stress, and inflammation in CCl4-induced acute hepatotoxicity mouse model through regulation of NF-κB. Garlic or garlic derivatives may therefore be a potential food supplement in the prevention of liver damage.


Subject(s)
Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/pathology , Cysteine/analogs & derivatives , Inflammation/prevention & control , NF-kappa B/metabolism , Oxidative Stress/drug effects , Alanine Transaminase/blood , Animals , Antioxidants/pharmacology , Cysteine/pharmacology , Female , Inflammation/chemically induced , Inflammation/pathology , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Male , Malondialdehyde/blood , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism
20.
J Ethnopharmacol ; 139(2): 462-70, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22138659

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lycium barbarum has been used as a traditional Chinese medicine to nourish liver, kidneys and the eyes. AIM OF THE STUDY: We investigated the protective mechanisms of Wolfberry, Lycium barbarum polysaccharides (LBP) in carbon tetrachloride (CCl(4))-induced acute liver injury. MATERIALS AND METHODS: Mice were intraperitoneally injected with a 50 µl/kg CCl(4) to induce acute hepatotoxicity (8h) and were orally fed with LBP 2 h before the CCl(4) injection. There were six experimental groups of mice (n=7-8 per group), namely: control mice (vehicle only; 1 mg/kg LBP or 10 mg/kg LBP), CCl(4)-treated mice and CCl(4)+LBP treated mice (1 mg/kg LBP or 10 mg/kg LBP). RESULTS: Pre-treatment with LBP effectively reduced the hepatic necrosis and the serum ALT level induced by CCl(4) intoxication. LBP remarkably inhibited cytochrome P450 2E1 expression and restored the expression levels of antioxidant enzymes. It also decreased the level of nitric oxide metabolism and lipid peroxidation induced by CCl(4). LBP attenuated hepatic inflammation via down-regulation of proinflammatory mediators and chemokines. Furthermore, LBP promoted liver regeneration after CCl(4) treatment. The protective effects of LBP against hepatotoxicity were partly through the down-regulation of nuclear factor kappa-B activity. CONCLUSION: LBP is effective in reducing necroinflammation and oxidative stress induced by a chemical toxin, thus it has a great potential use as a food supplement in the prevention of hepatic diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/prevention & control , Drugs, Chinese Herbal/pharmacology , Liver/drug effects , Lycium , Oxidative Stress/drug effects , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP2E1 Inhibitors , Cytokines/genetics , Cytokines/metabolism , Cytoprotection , Disease Models, Animal , Drugs, Chinese Herbal/isolation & purification , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic/drug effects , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Liver/pathology , Lycium/chemistry , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Necrosis , Nitric Oxide/metabolism , Plants, Medicinal
SELECTION OF CITATIONS
SEARCH DETAIL
...