Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 418
Filter
1.
Biomed Pharmacother ; 175: 116590, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653109

ABSTRACT

Alcohol-associated liver disease (ALD) is a leading factor of liver-related death worldwide. ALD has various manifestations that include steatosis, hepatitis, and cirrhosis and is currently without approved pharmacotherapies. The Src homology phosphatase 2 (Shp2) is a drug target in some cancers due to its positive regulation of Ras-mitogen-activated protein kinase signaling and cell proliferation. Shp2 pharmacological inhibition yields beneficial outcomes in animal disease models, but its impact on ALD remains unexplored. This study aims to investigate the effects of Shp2 inhibition and its validity using a preclinical mouse model of ALD. We report that the administration of SHP099, a potent and selective allosteric inhibitor of Shp2, partially ameliorated ethanol-induced hepatic injury, inflammation, and steatosis in mice. Additionally, Shp2 inhibition was associated with reduced ethanol-evoked activation of extracellular signal-regulated kinase (ERK), oxidative, and endoplasmic reticulum (ER) stress in the liver. Besides the liver, excessive alcohol consumption induces multi-organ injury and dysfunction, including the intestine. Notably, Shp2 inhibition diminished ethanol-induced intestinal inflammation and permeability, abrogated the reduction in tight junction protein expression, and the activation of ERK and stress signaling in the ileum. Collectively, Shp2 pharmacological inhibition mitigates the deleterious effects of ethanol in the liver and intestine in a mouse model of ALD. Given the multifactorial aspects underlying ALD pathogenesis, additional studies are needed to decipher the utility of Shp2 inhibition alone or as a component in a multitherapeutic regimen to combat this deadly malady.


Subject(s)
Disease Models, Animal , Ethanol , Liver Diseases, Alcoholic , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Animals , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/prevention & control , Liver Diseases, Alcoholic/enzymology , Liver Diseases, Alcoholic/drug therapy , Mice , Male , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Ethanol/toxicity , Liver/drug effects , Liver/pathology , Liver/enzymology , Liver/metabolism , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects
2.
Nat Commun ; 13(1): 557, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35091576

ABSTRACT

MATα1 catalyzes the synthesis of S-adenosylmethionine, the principal biological methyl donor. Lower MATα1 activity and mitochondrial dysfunction occur in alcohol-associated liver disease. Besides cytosol and nucleus, MATα1 also targets the mitochondria of hepatocytes to regulate their function. Here, we show that mitochondrial MATα1 is selectively depleted in alcohol-associated liver disease through a mechanism that involves the isomerase PIN1 and the kinase CK2. Alcohol activates CK2, which phosphorylates MATα1 at Ser114 facilitating interaction with PIN1, thereby inhibiting its mitochondrial localization. Blocking PIN1-MATα1 interaction increased mitochondrial MATα1 levels and protected against alcohol-induced mitochondrial dysfunction and fat accumulation. Normally, MATα1 interacts with mitochondrial proteins involved in TCA cycle, oxidative phosphorylation, and fatty acid ß-oxidation. Preserving mitochondrial MATα1 content correlates with higher methylation and expression of mitochondrial proteins. Our study demonstrates a role of CK2 and PIN1 in reducing mitochondrial MATα1 content leading to mitochondrial dysfunction in alcohol-associated liver disease.


Subject(s)
Liver Diseases, Alcoholic/metabolism , Methionine Adenosyltransferase/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Blotting, Western , Casein Kinase II/metabolism , Cell Line , Ethanol/pharmacology , Female , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Liver/cytology , Liver/drug effects , Liver/metabolism , Liver Diseases, Alcoholic/enzymology , Methionine Adenosyltransferase/genetics , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mutation , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Protein Binding
3.
Acta Pharmacol Sin ; 42(7): 1101-1110, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33028983

ABSTRACT

Alcoholic liver disease (ALD) is one of the pathogenic factors of chronic liver disease with the highest clinical morbidity worldwide. Ursolic acid (UA), a pentacyclic terpenoid carboxylic acid, has shown many health benefits including antioxidative, anti-inflammatory, anticancer, and hepatoprotective activities. We previously found that UA was metabolized in vivo into epoxy-modified UA containing an epoxy electrophilic group and had the potential to react with nucleophilic groups. In this study we prepared an alkynyl-modified UA (AM-UA) probe for tracing and capturing the target protein of UA from liver in mice, then investigated the mode by which UA bound to its target in vivo. By conducting proteome identification and bioinformatics analysis, we identified caspase-3 (CASP3) as the primary target protein of UA associated with liver protection. Molecule docking analysis showed that the epoxy group of the UA metabolite reacted with Cys-163 of CASP3, forming a covalent bond with CASP3. The binding mode of the UA metabolites (UA, CM-UA, and EM-UA) was verified by biochemical evaluation, demonstrating that the epoxy group produced by metabolism played an important role in the inhibition of CASP3. In alcohol-treated HepG2 cells, pretreatment with the UA metabolite (10 µM) irreversibly inhibited CASP3 activities, and subsequently decreased the cleavage of PARP and cell apoptosis. Finally, pre-administration of UA (20-80 mg· kg-1 per day, ig, for 1 week) dose-dependently alleviated alcohol-induced liver injury in mice mainly via the inhibition of CASP3. In conclusion, this study demonstrates that UA is a valuable lead compound for the treatment of ALD.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Caspase Inhibitors/therapeutic use , Liver Diseases, Alcoholic/drug therapy , Liver/drug effects , Triterpenes/therapeutic use , Amino Acid Sequence , Animals , Caspase 3/chemistry , Caspase Inhibitors/metabolism , Cysteine/chemistry , Epoxy Compounds/chemistry , Epoxy Compounds/therapeutic use , Hep G2 Cells , Hepatocytes/drug effects , Humans , Liver/enzymology , Liver/pathology , Liver Diseases, Alcoholic/enzymology , Liver Diseases, Alcoholic/pathology , Male , Mice , Molecular Docking Simulation , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Sequence Alignment , Triterpenes/metabolism , Ursolic Acid
4.
Chem Commun (Camb) ; 56(75): 11102-11105, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32812955

ABSTRACT

A probe has been developed for imaging alcoholic liver injury through detecting the overexpressed cytochrome P450 reductase in hypoxia in the hepatic region. Upon response to the enzyme, the activated probe displays turn-on fluorescence and near-infrared absorption and generates prominent optoacoustic signals.


Subject(s)
Fluorescent Dyes/chemistry , Liver Diseases, Alcoholic/diagnostic imaging , NADPH-Ferrihemoprotein Reductase/analysis , Optical Imaging , Photoacoustic Techniques , Animals , Cell Hypoxia , Cell Line , Disease Models, Animal , Humans , Liver Diseases, Alcoholic/enzymology , Mice , Molecular Structure , NADPH-Ferrihemoprotein Reductase/metabolism
5.
Drug Chem Toxicol ; 43(5): 546-551, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31094237

ABSTRACT

Alcohol is the most abused psychoactive substance and known hepatotoxicant. Present study elucidates possible therapeutic effect of oral alpha-ketoglutarate (AKG) supplementation against alcohol induced hepatic dysfunction, using biochemical, histopathological and most importantly, in vivo functional imaging approaches. Animals were divided into three groups of 6 animals each. Group-I (control): Normal saline; Group-II: 20% (v/v) solution of ethanol (5 ml/day) intragastrically using oral gavage for 2 months. Group-III: ethanol treatment as in group-II along with AKG supplementation (2g/kg/bw; intragastrically using oral gavage for 2 months). In vivo hepatobiliary scintigraphy was performed in all animals using 99mTc-mebrofenin (99mTc-MEB) as radiotracer to determine changes in (a) Hepatic extraction fraction (HEF), for quantification of radiotracer uptake, (b) Time to reach maximum hepatic uptake (Tpeak), and (c) Time for hepatic uptake to reduce by 50% (T1/2peak). Biochemical (alanine aminotransferase, aspartate aminotransferase, reduced glutathione, superoxide dismutase, catalase, and lipid peroxidation) and histological parameters were also studied. Hepatic uptake and excretion kinetics using 99mTc-MEB scintigraphy showed prompt 99mTc-MEB clearance from liver in control group (HEF: 91.26 ± 2.32; Tpeak: 143 ± 23 sec; T1/2peak: 434 ± 41 sec), while it was significantly abnormal in ethanol group and showed less efficient radiotracer accumulation (HEF: 62.72 ± 5.6; Tpeak: 201 ± 33 sec; T1/2peak: 542 ± 52 sec). Supplementation of AKG along with ethanol significantly improved liver function (HEF: 76.42 ± 5.3; Tpeak: 155 ± 34 sec; T1/2peak: 455 ± 22 sec). Biochemical and histopathology parameters were correlative to findings of functional imaging study. Results strongly indicate hepatoprotective potential of AKG against alcohol-induced hepatic injury. Study further proposes the use of in vivo hepatobiliary scintigraphy for high throughput screening of other hepatoprotectants.


Subject(s)
Ethanol/toxicity , Ketoglutaric Acids/therapeutic use , Liver Diseases, Alcoholic/drug therapy , Liver Diseases/prevention & control , Liver/drug effects , Alanine Transaminase/analysis , Animals , Aspartate Aminotransferases/analysis , Disease Models, Animal , Ketoglutaric Acids/pharmacology , Liver/enzymology , Liver Diseases, Alcoholic/enzymology , Male , Radionuclide Imaging , Rats , Rats, Sprague-Dawley
7.
Dig Liver Dis ; 51(8): 1154-1163, 2019 08.
Article in English | MEDLINE | ID: mdl-31003959

ABSTRACT

Alcoholic liver disease (ALD) is one of the most common liver diseases worldwide. However, the exact mechanisms underlying ALD remain unclear. Previous studies reported that sphingosine kinase 2 (SphK2) plays an essential role in regulating hepatic lipid metabolism. In the current study, we demonstrate that compared to wild-type (WT) mice, SphK2 deficient (SphK2-/-) mice exhibited a greater degree of liver injury and hepatic lipid accumulation after feeding with an alcohol diet for 60 days. This is accompanied by a down-regulation of steroid 7-alpha-hydroxylase (Cyp7b1) and an up-regulation of pro-inflammatory mediators (Tnfα, F4/80, Il-1ß). In vitro experiments showed that alcohol induced SphK2 expression in mouse primary hepatocytes and cultured mouse macrophages. Furthermore, alcohol feeding induced a more severe intestinal barrier dysfunction in SphK2-/- mice than WT mice. Deficiency of SphK2 impaired the growth of intestinal organoids. Finally, SphK2 expression levels were down-regulated in the livers of human patients with alcoholic cirrhosis and hepatocellular carcinoma compared to healthy controls. In summary, these findings suggest that SphK2 is a crucial regulator of hepatic lipid metabolism and that modulating the SphK2-mediated signaling pathway may represent a novel therapeutic strategy for the treatment of ALD and other metabolic liver diseases.


Subject(s)
Liver Diseases, Alcoholic/enzymology , Liver/enzymology , Liver/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Cells, Cultured , Female , Hepatocytes/enzymology , Humans , Intestines/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Steroid Hydroxylases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
8.
Alcohol ; 79: 71-79, 2019 09.
Article in English | MEDLINE | ID: mdl-30611703

ABSTRACT

BACKGROUND: We previously reported that nanoformulated copper/zinc superoxide dismutase (Nano) attenuates non-alcoholic fatty liver disease and adipose tissue (AT) inflammation in obese animals. Here, we sought to determine whether Nano treatment attenuates alcohol-associated liver disease (AALD) and AT inflammation in alcohol-fed mice. METHODS: We pre-treated E-47 cells (HepG2 cells that over-express CYP2E1) with native- or nano-superoxide dismutase (SOD) for 6 h, followed by treatment with ethanol and/or linoleic acid (LA), a free fatty acid. For in vivo studies, male C57BL/6 mice were fed the Lieber-DeCarli control or ethanol liquid diet for 4 weeks. The mice received Nano once every 2 days during the last 2 weeks of ethanol feeding. RESULTS: Our in vitro studies revealed that Nano pretreatment reduced LA + ethanol-induced oxidative stress in E-47 cells. Our in vivo experiments showed that ethanol-fed Nano-treated mice had 22% lower hepatic triglyceride levels than mice fed ethanol alone. Nano-treated ethanol-fed mice also had 2-fold lower levels of Cd68 and similarly reduced levels of Ccl2 and Mmp12 mRNAs, than in untreated ethanol-fed mice. We also noted that ethanol feeding caused a remarkable increase in hepatic and/or plasma MCP-1 and CCR2 protein, which was blunted in ethanol + Nano-treated animals. The hepatic content of SREBP-1c, a transcription factor that promotes lipogenesis, was higher in ethanol-fed mice than controls but was attenuated in ethanol + Nano-treated animals. Further, livers of ethanol + Nano-treated mice had significantly higher levels of phosphorylated adenosine monophosphate-activated protein kinase (AMPK) than both control and ethanol-fed mice. In AT, the levels of Il6 mRNA, a hepatoprotective cytokine, and that of Arg1, a marker of anti-inflammatory macrophages, were significantly increased in ethanol + Nano-treated mice compared with control mice. CONCLUSION: Our data indicate that Nano treatment attenuates ethanol-induced steatohepatitis and that this effect is associated with an apparent activation of AMPK signaling. Our data also suggest that Nano induces Arg1 and Il6 expression in AT, suggesting anti-inflammatory effects in this tissue.


Subject(s)
Adipose Tissue/drug effects , Ethanol/adverse effects , Free Radical Scavengers/pharmacology , Inflammation/enzymology , Liver Diseases, Alcoholic/enzymology , Liver/drug effects , Superoxide Dismutase/pharmacology , AMP-Activated Protein Kinase Kinases , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Chemokine CCL2/metabolism , Cytochrome P-450 CYP2E1/genetics , Drug Compounding , Gene Expression , Hep G2 Cells , Humans , Lipid Metabolism , Male , Matrix Metalloproteinase 12/metabolism , Mice , Mice, Inbred C57BL , Nanostructures , Oxidative Stress , Protein Kinases/metabolism , Receptors, CCR2/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
9.
Med Sci Monit ; 24: 8372-8382, 2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30457983

ABSTRACT

BACKGROUND Excessive alcohol consumption can cause hepatocellular injury. ATPase II (ATP8A1) can display an ATP-dependent phospholipid translocase activity. However, the function of ATP8A1 in hepatocyte injury is still unclear. In the present study we explored the effect of ATP8A1 on ethanol-induced hepatocyte injury. MATERIAL AND METHODS A human hepatocyte strain, HL-7702, was pretreated by ethanol with gradient concentration for 2, 4, 8, and 12 h, and were then divided into 6 groups after the cells were transfected. We detected cell viability by use of the Cell Counting Kit-8 (CCK-8) assay. Reactive oxygen species (ROS), apoptosis rate, and mitochondrial membrane potential (MMP) were measured using flow cytometry. We used quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot to measure the mRNA and protein expression, respectively. RESULTS Ethanol inhibited the viability of HL-7702 cells and suppressed the expression of ATP8A1 in dose- and time-dependent manners. Furthermore, over-expression of ATP8A1 reduced the level of ROS and the apoptosis rate and recovered the MMP. Additionally, over-expressed ATP8A1 regulated the protein and mRNA levels of apoptosis-related molecules. Moreover, over-expression of ATP8A1 enhanced the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt). CONCLUSIONS Over-expression of ATP8A1 alleviated ethanol-induced hepatocyte injury. Moreover, the PI3K/Akt signaling pathway appears to participate in inhibition of ethanol-induced hepatocyte apoptosis and may provide a candidate target for the treatment of alcoholic liver diseases (ALD).


Subject(s)
Adenosine Triphosphatases/biosynthesis , Ethanol/toxicity , Liver/drug effects , Liver/enzymology , Phospholipid Transfer Proteins/biosynthesis , Adenosine Triphosphatases/genetics , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Diseases, Alcoholic/enzymology , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/pathology , Membrane Potential, Mitochondrial/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phospholipid Transfer Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
10.
Cell Mol Life Sci ; 75(17): 3143-3157, 2018 09.
Article in English | MEDLINE | ID: mdl-29947925

ABSTRACT

Alcoholic liver disease (ALD) remains to be a worldwide health problem. It is generally accepted that oxidative stress plays critical roles in the pathogenesis of ALD, and antioxidant therapy represents a logical strategy for the prevention and treatment of ALD. Nuclear factor erythroid-derived 2-like 2 (NFE2L2 or Nrf-2) is essential for the antioxidant responsive element (ARE)-mediated induction of endogenous antioxidant enzymes such as heme oxygenase 1 (HO-1) and glutamate-cysteine ligase [GCL, the rate-limiting enzyme in the synthesis of glutathione (GSH)]. Activation of Nrf-2 pathway by genetic manipulation or pharmacological agents has been demonstrated to provide protection against ALD, which suggests that targeting Nrf-2 may be a promising approach for the prevention and treatment of ALD. Herein, we review the relevant literature about the potential hepatoprotective roles of Nrf-2 activation against ALD.


Subject(s)
Antioxidants/metabolism , Glutathione/biosynthesis , Liver Diseases, Alcoholic/prevention & control , NF-E2-Related Factor 2/metabolism , Animals , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/metabolism , Humans , Liver Diseases, Alcoholic/enzymology , Liver Diseases, Alcoholic/metabolism , Models, Biological
11.
FASEB J ; 32(6): 3278-3288, 2018 06.
Article in English | MEDLINE | ID: mdl-29401608

ABSTRACT

Alcohol acts through numerous pathways leading to alcoholic liver disease (ALD). Cytochrome P450 (CYP2E1), an ethanol-inducible enzyme, metabolizes ethanol-producing toxic reactive oxygen species (ROS) and is regulated at the posttranslational level. Small ubiquitin-like modifier (SUMO)ylation is a posttranslational modification that involves the addition of SUMOs, which modulate protein stability, activity, and localization. We demonstrated that ubiquitin-conjugation enzyme 9, the SUMO-conjugating enzyme, is induced in the livers of an intragastric ethanol mouse model. Our aim is to examine whether SUMOylation could regulate ethanol-induced CYP2E1 expression in ALD and to elucidate the molecular mechanism(s). CYP2E1 and UBC9 expression in vitro and in vivo was detected by real-time PCR and immunoblotting/immunostaining. SUMOylation was assayed by mass spectrometry and coimmunoprecipitation. Ubc9 expression was induced in ethanol-fed mouse livers, and silencing inhibited ethanol-mediated CYP2E1 microsomal retention and enzymatic activity. CYP2E1 SUMOylation was found to be induced by ethanol in vitro and in vivo. Ubc9 silencing prevents ethanol-induced lipid accumulation and ROS production. UBC9 was highly expressed in human ALD livers. Finally, we found that lysine 410 is a key SUMOylated residue contributing to CYP2E1 protein stability and activity preventing CYP2E1 SUMOylation. Ethanol-mediated up-regulation of CYP2E1 via SUMOylation enhancing its protein stability and activity and may have important implications in ALD.-Tomasi, M. L., Ramani, K., Ryoo, M., Cossu, C., Floris, A., Murray, B. J., Iglesias-Ara, A., Spissu, Y., Mavila, N. SUMOylation regulates cytochrome P450 2E1 expression and activity in alcoholic liver disease.


Subject(s)
Cytochrome P-450 CYP2E1/biosynthesis , Ethanol/adverse effects , Gene Expression Regulation, Enzymologic/drug effects , Liver Diseases, Alcoholic/enzymology , Sumoylation/drug effects , Animals , Enzyme Stability/drug effects , Ethanol/pharmacology , Liver Diseases, Alcoholic/pathology , Mice , Microsomes, Liver/enzymology , Microsomes, Liver/pathology , Reactive Oxygen Species/metabolism , Ubiquitin-Conjugating Enzymes/biosynthesis
12.
Cell Death Dis ; 8(10): e3152, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29072704

ABSTRACT

Hepatic apoptosis is involved in the progression of alcoholic liver disease (ALD). Caspase-8, the apical initiator in death receptor-mediated apoptosis, has been implicated in acute liver injury and in non-alcoholic steatohepatitis. However, the relevance of Caspase-8 in the pathogenesis of ALD remains unclear. In the present study, we investigated the impact of Caspase-8 in human and murine alcohol-induced apoptosis and in ALD. We investigated human samples from ALD patients, primary mouse hepatocytes, and hepatocyte-specific Caspase-8 knockout (Casp8Δhepa) mice in acute and chronic models of ethanol (EtOH) administration. Caspase-8 activation was detected in liver biopsies from ALD patients, as well as in livers of wild-type (WT) mice after chronic ethanol feeding for 8 weeks using the Lieber-DeCarli model. Lack of Caspase-8 expression in Casp8Δhepa animals failed to prevent alcohol-induced liver damage and apoptosis. Instead, inhibition of Caspase-8 shifted the ethanol-induced death signals towards pronounced activation of the intrinsic, mitochondria-dependent apoptosis pathway in Casp8Δhepa livers involving enhanced release of cytochrome c, stronger Caspase-9 activation and specific morphological changes of mitochondria. In vitro and in vivo intervention using a pan-caspase inhibitor markedly attenuated alcohol-induced hepatocyte damage in a Caspase-8-independent manner. Surprisingly, EtOH-fed Casp8Δhepa mice displayed significantly attenuated steatosis and reduced hepatic triglyceride and free fatty acids content. Caspase-8 is dispensable for alcohol-induced apoptosis, but plays an unexpected role for alcohol-dependent fat metabolism. We provide evidence that simultaneous inhibition of extrinsic and intrinsic apoptosis signaling using pan-caspase inhibitors in vivo might be an optimal approach to treat alcohol-induced liver injury.


Subject(s)
Caspase 8/metabolism , Liver Diseases, Alcoholic/enzymology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Enzyme Activation/drug effects , Ethanol/pharmacology , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Lipid Metabolism , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Knockout
13.
Exp Mol Pathol ; 102(1): 162-180, 2017 02.
Article in English | MEDLINE | ID: mdl-28077318

ABSTRACT

This paper is based upon the "8th Charles Lieber's Satellite Symposium" organized by Manuela G. Neuman at the Research Society on Alcoholism Annual Meeting, on June 25, 2016 at New Orleans, Louisiana, USA. The integrative symposium investigated different aspects of alcohol-induced liver disease (ALD) as well as non-alcohol-induced liver disease (NAFLD) and possible repair. We revealed the basic aspects of alcohol metabolism that may be responsible for the development of liver disease as well as the factors that determine the amount, frequency and which type of alcohol misuse leads to liver and gastrointestinal diseases. We aimed to (1) describe the immuno-pathology of ALD, (2) examine the role of genetics in the development of alcoholic hepatitis (ASH) and NAFLD, (3) propose diagnostic markers of ASH and non-alcoholic steatohepatitis (NASH), (4) examine age and ethnic differences as well as analyze the validity of some models, (5) develop common research tools and biomarkers to study alcohol-induced effects, 6) examine the role of alcohol in oral health and colon and gastrointestinal cancer and (7) focus on factors that aggravate the severity of organ-damage. The present review includes pre-clinical, translational and clinical research that characterizes ALD and NAFLD. Strong clinical and experimental evidence lead to recognition of the key toxic role of alcohol in the pathogenesis of ALD with simple fatty infiltrations and chronic alcoholic hepatitis with hepatic fibrosis or cirrhosis. These latter stages may also be associated with a number of cellular and histological changes, including the presence of Mallory's hyaline, megamitochondria, or perivenular and perisinusoidal fibrosis. Genetic polymorphisms of ethanol metabolizing enzymes and cytochrome p450 (CYP) 2E1 activation may change the severity of ASH and NASH. Other risk factors such as its co-morbidities with chronic viral hepatitis in the presence or absence of human deficiency virus were discussed. Dysregulation of metabolism, as a result of ethanol exposure, in the intestine leads to colon carcinogenesis. The hepatotoxic effects of ethanol undermine the contribution of malnutrition to the liver injury. Dietary interventions such as micro and macronutrients, as well as changes to the microbiota have been suggested. The clinical aspects of NASH, as part of the metabolic syndrome in the aging population, have been presented. The symposium addressed mechanisms and biomarkers of alcohol induced damage to different organs, as well as the role of the microbiome in this dialog. The microbiota regulates and acts as a key element in harmonizing immune responses at intestinal mucosal surfaces. It is known that microbiota is an inducer of proinflammatory T helper 17 cells and regulatory T cells in the intestine. The signals at the sites of inflammation mediate recruitment and differentiation in order to remove inflammatory inducers and promote tissue homeostasis restoration. The change in the intestinal microbiota also influences the change in obesity and regresses the liver steatosis. Evidence on the positive role of moderate alcohol consumption on heart and metabolic diseases as well on reducing steatosis have been looked up. Moreover nutrition as a therapeutic intervention in alcoholic liver disease has been discussed. In addition to the original data, we searched the literature (2008-2016) for the latest publication on the described subjects. In order to obtain the updated data we used the usual engines (Pub Med and Google Scholar). The intention of the eighth symposia was to advance the international profile of the biological research on alcoholism. We also wish to further our mission of leading the forum to progress the science and practice of translational research in alcoholism.


Subject(s)
Alcoholism/complications , Life Style , Liver Diseases, Alcoholic/complications , Microbiota , Non-alcoholic Fatty Liver Disease/complications , Congresses as Topic , Cytochrome P-450 CYP2E1/genetics , Cytochrome P-450 CYP2E1/metabolism , Hepatitis, Alcoholic/complications , Hepatitis, Alcoholic/enzymology , Hepatitis, Alcoholic/genetics , Humans , Liver Diseases, Alcoholic/enzymology , Liver Diseases, Alcoholic/genetics , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Polymorphism, Genetic
14.
Curr Mol Pharmacol ; 10(3): 172-178, 2017.
Article in English | MEDLINE | ID: mdl-26278389

ABSTRACT

This article reviews recent studies on CYP2E1-mediated alcoholic liver injury, the induction of CYP2A5 by alcohol and the mechanism for this upregulation, especially the permissive role of CYP2E1 in the induction of CYP2A5 by alcohol and the CYP2E1-ROS-Nrf2 pathway, and protective effects of CYP2A5 against ethanol-induced oxidative liver injury. Ethanol can induce CYP2E1, an active generator of reactive oxygen species (ROS), and CYP2E1 is a contributing factor for alcoholinduced oxidative liver injury. CYP2A5, another isoform of cytochrome P450, can also be induced by ethanol. Chronic feeding of ethanol to wild type mice increased CYP2A5 catalytic activity, protein and mRNA levels as compared to pair-fed controls. This induction was blunted in CYP2E1 knockout (cyp2e1-/-) mice but was restored when human CYP2E1 was reintroduced and expressed in cyp2e1-/- mice. Ethanol-induced CYP2E1 co-localized with CYP2A5 and preceded the elevation of CYP2A5. The antioxidants N-acetyl cysteine and vitamin C lowered the alcohol elevation of ROS and blunted the alcohol induction of CYP2A5, but not CYP2E1, suggesting ROS play a novel role in the crosstalk between CYP2E1 and CYP2A5. The antioxidants blocked the activation of Nrf2, a transcription factor known to upregulate expression of CYP2A5. When alcohol-induced liver injury was enhanced in Nrf2 knockout (Nrf2-/-) mice, alcohol elevation of CYP2A5 but not CYP2E1 was also lower in Nrf2-/- mice. CYP2A5 knockout (cyp2a5-/-) mice exhibited an enhanced alcoholic liver injury compared with WT mice as indicated by serum ALT, steatosis and necroinflammation. Alcohol-induced hyperglycemia were observed in cyp2a5-/- mice but not in WT mice.


Subject(s)
Cytochrome P450 Family 2/metabolism , Liver Diseases, Alcoholic/enzymology , Animals , Antioxidants/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 CYP2E1/metabolism , Humans , Hyperglycemia/chemically induced , Hyperglycemia/enzymology , Hyperglycemia/pathology , Liver Diseases, Alcoholic/pathology , Signal Transduction , Up-Regulation
15.
Toxicol Lett ; 267: 1-10, 2017 Feb 05.
Article in English | MEDLINE | ID: mdl-27989594

ABSTRACT

Salvianolic acid B (SalB), a water-soluble polyphenol extracted from Radix Salvia miltiorrhiza, has been reported to possess many pharmacological activities. This study investigated the hepatoprotective effects of SalB in chronic alcoholic liver disease (ALD) and explored the related signaling mechanisms. In vivo, SalB treatment significantly attenuated ethanol-induced liver injury by blocking the elevation of serum aminotransferase activities and markedly decreased hepatic lipid accumulation by reducing serum and liver triglyceride (TG) and total cholesterol (TC) levels. Moreover, SalB treatment ameliorated ethanol-induced hepatic inflammation by decreasing the levels of hepatotoxic cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Importantly, SalB pretreatment significantly increased the expression of SIRT1 and downregulated the expression of inflammatory mediator C-reactive protein (CRP) and lipoprotein carbohydrate response element-binding protein (ChREBP). In vitro, SalB significantly reversed ethanol-induced down-regulation of SIRT1 and increased CRP and ChREBP expression. Interestingly, the effects of SalB on SIRT1, CRP and ChREBP were mostly abolished by treatment with either SIRT1 siRNA or EX527, a specific inhibitor of SIRT1, indicating that SalB decreased CRP and ChREBP expression by activating SIRT1. SalB exerted anti-steatotic and anti-inflammatory effects against alcoholic liver injury by inducing SIRT1-mediated inhibition of CRP and ChREBP expression.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Benzofurans/pharmacology , Carrier Proteins/metabolism , Liver Diseases, Alcoholic/prevention & control , Liver/drug effects , Sirtuin 1/metabolism , Animals , Biomarkers/blood , Carbazoles/pharmacology , Chronic Disease , Cytokines/metabolism , Cytoprotection , Disease Models, Animal , Dose-Response Relationship, Drug , Hep G2 Cells , Hepatocyte Nuclear Factor 1-alpha/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Inflammation Mediators/metabolism , Liver/enzymology , Liver/pathology , Liver Diseases, Alcoholic/enzymology , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/pathology , Male , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Transfection
16.
Alcohol Clin Exp Res ; 40(10): 2076-2084, 2016 10.
Article in English | MEDLINE | ID: mdl-27575873

ABSTRACT

BACKGROUND: Overproduction of reactive oxygen species is associated with the development of alcoholic liver disease (ALD). Plant polyphenols have been used as dietary interventions for multiple diseases including ALD. The objective of this study was to determine whether dietary supplementation with fisetin, a novel flavonoid, exerts beneficial effect on alcohol-induced liver injury. METHODS: C57BL/6J mice were pair-fed with the Lieber-DeCarli control or ethanol (EtOH) diet for 4 weeks with or without fisetin supplementation at 10 mg/kg/d. RESULTS: Alcohol feeding induced lipid accumulation in the liver and increased plasma alanine aminotransferase and aspartate aminotransferase activities, which were attenuated by fisetin supplementation. The EtOH concentrations in the plasma and liver were significantly elevated by alcohol exposure but were reduced by fisetin supplementation. Although fisetin did not affect the protein expression of alcohol metabolism enzymes, the aldehyde dehydrogenase activities were significantly increased by fisetin compared to the alcohol alone group. In addition, fisetin supplementation remarkably reduced hepatic NADPH oxidase 4 levels along with decreased plasma hydrogen peroxide and hepatic superoxide and 4-hydroxynonenal levels after alcohol exposure. Alcohol-induced apoptosis and up-regulation of Fas and cleaved caspase-3 in the liver were prevented by fisetin. Moreover, fisetin supplementation attenuated alcohol-induced hepatic steatosis through increasing plasma adiponectin levels and hepatic protein levels of p-AMPK, ACOX1, CYP4A, and MTTP. CONCLUSIONS: This study demonstrated that the protective effect of fisetin on ALD is achieved by accelerating EtOH clearance and inhibition of oxidative stress. The data suggest that fisetin has a therapeutical potential for treating ALD.


Subject(s)
Dietary Supplements , Ethanol/adverse effects , Flavonoids/therapeutic use , Liver Diseases, Alcoholic/diet therapy , AMP-Activated Protein Kinases/metabolism , Acyl-CoA Oxidase/metabolism , Adiponectin/blood , Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Animals , Apoptosis/drug effects , Carrier Proteins/metabolism , Cytochrome P-450 CYP4A/metabolism , Ethanol/blood , Ethanol/pharmacokinetics , Fatty Liver/complications , Fatty Liver/diet therapy , Flavonols , Hydrogen Peroxide/blood , Liver/enzymology , Liver/metabolism , Liver Diseases, Alcoholic/blood , Liver Diseases, Alcoholic/complications , Liver Diseases, Alcoholic/enzymology , Male , Mice , NADPH Oxidase 4/metabolism , Protective Agents/therapeutic use , Superoxides/metabolism , Up-Regulation/drug effects
17.
Hepatology ; 64(4): 1057-71, 2016 10.
Article in English | MEDLINE | ID: mdl-27302565

ABSTRACT

UNLABELLED: The spectrum of alcoholic liver disease (ALD) is a major cause of mortality with limited therapies available. Because alcohol targets numerous signaling pathways in hepatocytes and in immune cells, the identification of a master regulatory target that modulates multiple signaling processes is attractive. In this report, we assessed the role of spleen tyrosine kinase (SYK), a nonreceptor tyrosine kinase, which has a central modulatory role in multiple proinflammatory signaling pathways involved in the pathomechanism of ALD. Using mouse disease models that represent various phases in the progression of human ALD, we found that alcohol, in all of these models, induced SYK activation in the liver, both in hepatocytes and liver mononuclear cells. Furthermore, significant SYK activation also occurred in liver samples and peripheral blood mononuclear cells of patients with ALD/alcoholic hepatitis compared to controls. Functional inhibition of SYK activation in vivo abrogated alcohol-induced hepatic neutrophil infiltration, resident immune cell activation, as well as inflammasome and extracellular signal-regulated kinase 1 and 2-mediated nuclear factor kappa B activation in mice. Strikingly, inhibition of SYK activation diminished alcohol-induced hepatic steatosis and interferon regulatory factor 3-mediated apoptosis. CONCLUSION: Our data demonstrate a novel, functional, and multicellular role for SYK phosphorylation in modulating immune cell-driven liver inflammation, hepatocyte cell death, and steatosis at different stages of ALD. These novel findings highlight SYK as a potential multifunctional target in the treatment of alcoholic steatohepatitis. (Hepatology 2016;64:1057-1071).


Subject(s)
Cell Death , Fatty Liver/prevention & control , Hepatocytes/pathology , Inflammation/prevention & control , Liver Diseases, Alcoholic/enzymology , Oxazines/pharmacology , Oxazines/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Syk Kinase/antagonists & inhibitors , Animals , Fatty Liver/etiology , Female , Humans , Inflammation/etiology , Liver Diseases, Alcoholic/complications , Male , Mice , Mice, Inbred C57BL , Middle Aged
18.
Alcohol Clin Exp Res ; 40(5): 988-99, 2016 05.
Article in English | MEDLINE | ID: mdl-27062444

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor gamma (PPARγ) signaling has been shown to regulate lipogenesis and lipid accumulation. Previous studies have shown that hepatic PPARγ is up-regulated in steatotic liver of both animal and human. However, the effects of hepatic PPARγ signaling on alcoholic liver disease (ALD) remain elusive. METHODS: To determine the role of hepatic PPARγ signaling on ALD, wild-type (WT) and hepatocyte-specific PPARγ knockdown (PPARγ∆Hep) mice were fed a modified Lieber-DeCarli alcohol or isocaloric maltose dextrin control liquid diet for 8 weeks to induce ALD. Blood parameters, hepatic steatosis, and inflammation were measured after 8-week alcohol feeding. RESULTS: Alcohol feeding to WT mice resulted in liver damage (alanine aminotransferase [ALT], 94.68 ± 17.05 U/L; aspartate aminotransferase [AST], 55.87 ± 11.29 U/L), which was significantly alleviated by hepatic PPARγ knockdown (ALT, 57.36 ± 14.98 U/L; AST, 38.06 ± 3.35 U/L). Alcohol feeding led to marked lipid accumulation and up-regulation of lipogenic genes including fatty acid transport protein 1 (FATP1), acetyl-CoA carboxylase (ACC), fatty acid synthase (FASN), lipin1 (LIPIN1), diacylglycerol acyltransferase 1 (DGAT1), and diacylglycerol acyltransferase 2 (DGAT2) in the livers of WT mice. Knockdown of hepatic PPARγ significantly alleviated alcohol-induced lipid accumulation and abolished the up-regulation of FASN, DGAT1, and DGAT2. Silencing of PPARγ in FL83B cells significantly decreased ethanol (EtOH)-, linoleic acid-, and EtOH plus linoleic acid-induced lipid accumulation. Knockdown of hepatic PPARγ also significantly reduced alcohol-induced inflammatory chemokine (monocyte chemotactic protein 1 [MCP1], keratinocyte-derived chemokine [KC], interferon gamma-induced protein 10 [IP-10]) and inflammatory infiltration (lymphocyte antigen 6 complex, locus G [Ly6G], and F4/80). CONCLUSIONS: The results suggest that hepatic PPARγ signaling contributes to alcohol-induced liver injury by promoting hepatic steatosis and inflammation.


Subject(s)
Ethanol/toxicity , Fatty Liver, Alcoholic/metabolism , Inflammation/metabolism , Liver Diseases, Alcoholic/metabolism , Liver/metabolism , PPAR gamma/metabolism , Signal Transduction/drug effects , Acetyl-CoA Carboxylase/biosynthesis , Animals , Cells, Cultured , Chemokines/metabolism , Diacylglycerol O-Acyltransferase/biosynthesis , Fatty Acid Synthases/biosynthesis , Fatty Acid Transport Proteins/biosynthesis , Fatty Liver, Alcoholic/enzymology , Gene Knockdown Techniques , Inflammation/enzymology , Liver Diseases, Alcoholic/enzymology , Male , Mice , Nuclear Proteins/biosynthesis , PPAR gamma/deficiency , PPAR gamma/genetics , Phosphatidate Phosphatase/biosynthesis , Up-Regulation
19.
Oncotarget ; 7(14): 17681-98, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26769846

ABSTRACT

Chronic alcohol exposure increased hepatic receptor-interacting protein kinase (RIP) 3 expression and necroptosis in the liver but its mechanisms are unclear. In the present study, we demonstrated that chronic alcohol feeding plus binge (Gao-binge) increased RIP3 but not RIP1 protein levels in mouse livers. RIP3 knockout mice had decreased serum alanine amino transferase activity and hepatic steatosis but had no effect on hepatic neutrophil infiltration compared with wild type mice after Gao-binge alcohol treatment. The hepatic mRNA levels of RIP3 did not change between Gao-binge and control mice, suggesting that alcohol-induced hepatic RIP3 proteins are regulated at the posttranslational level. We found that Gao-binge treatment decreased the levels of proteasome subunit alpha type-2 (PSMA2) and proteasome 26S subunit, ATPase 1 (PSMC1) and impaired hepatic proteasome function. Pharmacological or genetic inhibition of proteasome resulted in the accumulation of RIP3 in mouse livers. More importantly, human alcoholics had decreased expression of PSMA2 and PSMC1 but increased protein levels of RIP3 compared with healthy human livers. Moreover, pharmacological inhibition of RIP1 decreased Gao-binge-induced hepatic inflammation, neutrophil infiltration and NF-κB subunit (p65) nuclear translocation but failed to protect against steatosis and liver injury induced by Gao-binge alcohol. In conclusion, results from this study suggest that impaired hepatic proteasome function by alcohol exposure may contribute to hepatic accumulation of RIP3 resulting in necroptosis and steatosis while RIP1 kinase activity is important for alcohol-induced inflammation.


Subject(s)
Fatty Liver/enzymology , Liver Diseases, Alcoholic/enzymology , Receptor-Interacting Protein Serine-Threonine Kinases/biosynthesis , Animals , Binge Drinking/enzymology , Binge Drinking/pathology , Ethanol/administration & dosage , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/metabolism , Humans , Liver/drug effects , Liver/enzymology , Liver/pathology , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Pore Complex Proteins/biosynthesis , Nuclear Pore Complex Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
20.
Sci Rep ; 6: 18685, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26725521

ABSTRACT

Activation of Kupffer cells (KCs) plays a central role in the pathogenesis of alcoholic liver disease (ALD). C57BL/6 mice fed EtOH-containing diet showed a mixed induction of hepatic classical (M1) and alternative (M2) macrophage markers. Since telomerase activation occurs at critical stages of myeloid and lymphoid cell activation, we herein investigated the role of telomerase reverse transcriptase (TERT), the determining factor of telomerase, in macrophage activation during ALD. In our study, TERT expression and telomerase activity (TA) were remarkably increased in liver tissue of EtOH-fed mice. Moreover, EtOH significantly up-regulated TERT in isolated KCs and RAW 264.7 cells and LPS induced TERT production in vitro. These data indicate that up-regulation of TERT may play a critical role in macrophages during ALD. Furthermore, loss- and gain-of-function studies suggested that TERT switched macrophages towards M1 phenotype by regulating NF-κB signaling, but had limited effect on M2 macrophages polarization in vitro. Additionally, PDTC, a chemical inhibitor of NF-κB, could dramatically down-regulate TERT expression and the hallmarks of M1 macrophages. Therefore, our study unveils the role of TERT in macrophage polarization and the cross-talk between TERT and p65, which may provide a possible explanation for the ethanol-mediated hepatic proinflammatory response and M1 macrophage polarization.


Subject(s)
Kupffer Cells/physiology , Liver Diseases, Alcoholic/enzymology , NF-kappa B/metabolism , Telomerase/physiology , Animals , Cell Polarity , Feedback, Physiological , Gene Expression , Lipopolysaccharides/pharmacology , Liver/enzymology , Liver/immunology , Liver/pathology , Liver Diseases, Alcoholic/immunology , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...