Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 21, 2016; 22(27): 6192-6200
Published online Jul 21, 2016. doi: 10.3748/wjg.v22.i27.6192
Aberrant post-translational protein modifications in the pathogenesis of alcohol-induced liver injury
Natalia A Osna, Murali Ganesan, Kusum K Kharbanda, Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
Natalia A Osna, Murali Ganesan, Kusum K Kharbanda, Department of Internal Medicine, 982000 Nebraska Medical Center, Omaha, NE 68198, United States
Wayne G Carter, School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby DE22 3DT, United Kingdom
Irina A Kirpich, Craig J McClain, Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Department of Medicine and Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, United States
Dennis R Petersen, Colin T Shearn, University of Colorado Denver, School of Pharmacy, Department of Pharmaceutical Sciences, Aurora, CO 80045, United States
Maria L Tomasi, Division of Gastroenterology, Department of Medicine, Davis research building, suite 2096/2035 Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
Kusum K Kharbanda, Department of Biochemistry and Molecular Biology, 985870 Nebraska Medical Center, Omaha, NE 68198, United States
Author contributions: All authors equally contributed to this paper with conception, literature review, drafting and critical revision, editing, and approval of the final version.
Conflict-of-interest statement: No potential conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Kusum K Kharbanda, PhD, Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE 68105, United States. kkharbanda@unmc.edu
Telephone: +1-402-9953752 Fax: +1-402-4490604
Received: March 26, 2016
Peer-review started: March 26, 2016
First decision: May 12, 2016
Revised: May 28, 2016
Accepted: June 15, 2016
Article in press: June 15, 2016
Published online: July 21, 2016

Abstract

It is likely that the majority of proteins will undergo post-translational modification, be it enzymatic or non-enzymatic. These modified protein(s) regulate activity, localization and interaction with other cellular molecules thereby maintaining cellular hemostasis. Alcohol exposure significantly alters several of these post-translational modifications leading to impairments of many essential physiological processes. Here, we present new insights into novel modifications following ethanol exposure and their role in the initiation and progression of liver injury. This critical review condenses the proceedings of a symposium at the European Society for the Biomedical Research on Alcoholism Meeting held September 12-15, 2015, in Valencia, Spain.

Key Words: Alcohol, Acetylation, Liver, Carbonylation methylation, Dysfunction, Methylation, Glycosylation, Phosphorylation, Ubiquitination, Sumoylation, Betaine, Post-translational protein modification

Core tip: A majority of proteins in our body undergo orchestrated post-translational modifications that influence protein structure and function. Chronic ethanol administration causes aberrant post-translational modification of proteins that play a critical role in the pathogenesis of alcoholic-induced liver damage.



INTRODUCTION

Ethanol consumption leads to many adverse functional and structural molecular changes in multiple organs and accounts for 2.5 million deaths globally each year[1,2]. Ethanol is mainly metabolized in the liver; this organ is therefore most susceptible to its toxic effects[3,4]. Sustained alcohol misuse produces a wide spectrum of hepatic lesions, the most characteristic being steatosis, hepatitis and fibrosis/cirrhosis[5]. Steatosis, characterized by fat accumulation in hepatocytes, develops in 90% of individuals who drink more than 16 g of alcohol per day[6]. In 30%-40% of individuals reporting chronic alcohol abuse, there is development of hepatitis which is characterized by inflammatory changes in the liver and ballooning degeneration of hepatocytes. In later stages of alcoholic liver disease (ALD), collagen deposition and regenerative nodules can result in the development of fibrosis and cirrhosis, respectively[2]. Better understanding of the mechanisms by which alcohol damages the liver may yield new pharmacologic strategies to blunt, halt, or reverse disease progression, potentially even in inveterate alcoholics.

Hepatic dysfunction due to chronic ethanol consumption is multifactorial involving dysregulation of multiple cellular pathways[7-9]. Significant to the aforementioned dysregulation is abnormal post-translational modification of proteins[10]. About 50%-90% of proteins in our body undergo orchestrated post-translational modifications that could influence protein structure and function. These modifications may be enzymatic or non-enzymatic and play an important role in functions of proteins through the regulation of activity, turnover and localization and/or interactions to maintain cellular hemostasis. These modifications include phosphorylation, acetylation, methylation, glycosylation, ubiquitination, sumoylation and ISGylation. Directed or inadvertent exposure to stressful factors, including chronic ethanol exposure, has been shown to cause aberrant post-translational modifications. Additionally, bioactive products of enzymatic or non-enzymatic lipid oxidation may also cause protein modifications with potential functional damage to protein[11] as well as impact the function of various metabolic pathways. This brief overview will focus on recent advances that have been made using global proteomic approaches and bioinformatics to better understand the impact of these altered post-translational modifications by ethanol.

Alcohol consumption significantly alters several post-translational modifications of proteins and this has been reviewed recently[10]. Here, we review the newly described and pathogenically relevant alterations that play important roles in the progression of ALD. First we discuss the undesired post-translational modifications that may occur via electrophilic species including reactive aldehydes (carbonylation), acyl groups (acetylation) and sugar moieties (glycosylation).

CARBONYLATION

A key contributor to the pathogenesis of ALD is enhanced hepatocellular oxidative stress resulting from the production of reactive oxygen species via induction of Cyp2E1 as well as xanthine and NADPH oxidases[12-16]. These reactive species, in turn, induce lipid peroxidation of unsaturated fatty acids including linoleic acid forming α/β unsaturated aldehydes[17,18]. The best characterized of these carbonyl-derivatives include 4-hydroxy-2-nonenal (4-HNE), 4-oxo-2-nonenal, malondialdehyde (MDA) and acrolein. Following their formation, these highly reactive lipid electrophiles modify DNA as well as lysine, cysteine and histidine residues on proteins, thereby impairing their structural or catalytic capabilities. Early proteomic approaches to identify carbonylated proteins in ALD used 2-dimensional electrophoresis followed by protein identification. These techniques were not very sensitive and only a handful of proteins were identified[19,20]. A commonality of all these proteins was the fact that all were very highly expressed, which permitted easier identification. Of interest, the majority of identified proteins were involved in either protein folding (heat shock proteins) or hepatocellular oxidative stress responses.

Recent advances in biotin hydrazide chemistry and in the sensitivity of mass spectrometry have allowed for a more in depth proteomic approach to identify less abundant proteins modified by reactive aldehydes in ALD. To date, using global proteomic approaches, over 2000 proteins that undergo carbonylation have been identified in either murine models or in human hepatic tissue isolated from patients with end-stage ALD[21-23]. Using enriched cellular fractions, chronic ethanol consumption led to an increase in carbonylation of microsomal and cytosolic proteins. Comprehensive pathway analysis of identified proteins revealed that ethanol consumption impacted many different cellular pathways foremost of which are the fatty acid metabolic, tricarboxylic acid cycle and amino acid metabolism. By increasing carbonylation of proteins involved in these pathways, mechanistic links have been proposed for ethanol’s impact on lipid accumulation as well as how acetyl CoA contributes to nutritional imbalances evident in alcoholics. These findings are further supported by an additional study that examined the effects of deletion of glutathione S-transferase A4-4 (GSTA4-4) which functions to remove 4-HNE reducing the effects of reactive aldehydes[24]. Using GSTA4-4 knockout mice and employing proteomics approaches, it was determined that carbonylation was increased in mitochondrial fractions especially in pathways regulating oxidative stress, fatty acid metabolism and amino acid metabolism supporting the contribution of GSTA4-4 in protecting mitochondria from reactive aldehydes (Supplementary Table 1). Concurrently, we have reported that carbonylation is increased in tissue obtained from end-stage alcoholics[23]. Not surprisingly, following mass spectral analysis, increased carbonylation of proteins regulating oxidative stress, metabolic and cytoskeletal processes were increased[24].

GLYCOSYLATION

In cells, glycosylation of proteins contributes to numerous cellular functions including assisting in proper protein folding as well as cell to cell adhesion. Global proteomic approaches and 2-dimensional electrophoresis were performed on microsomal fractions consisting primarily of smooth and rough endoplasmic reticulum, isolated from chronically ethanol fed mice. These studies revealed a significantly decrease in microsomal glycosylation following 8 wk of alcohol consumption. Subsequent bioinformatic pathway analysis revealed significant decreases in glycosylation of proteins regulating protein folding, redox homeostasis and the unfolded protein response among others. These results suggest that decreased glycosylation may contribute to the observed increased in ubiquitinated proteins in murine models of ALD[25].

ACETYLATION

In hepatocytes, acetylation of lysine residues results in regulation of many cellular functions including gene expression and metabolism. As it is metabolized, ethanol is converted to acetaldehyde by alcohol dehydrogenases followed by removal of the aldehyde group by mitochondrial aldehyde dehydrogenases (ALDH2) to produce acetate, which is converted to acetyl CoA[26]. By way of protein acyl transferases, acetyl CoA is then utilized in part as a substrate for protein acetylation. Therefore, it is not surprising that over the last decade, ethanol abuse has been determined to directly affect protein acetylation[27-30]. In murine models of ALD, ethanol decreases expression of the class III nicotinamide adenine dinucleotide (NAD+/NADH) dependent protein deacetylases, Sirtuin 1 (SIRT1) and Sirtuin 5 and decreases enzymatic activity of Sirtuin 3[31-35]. All these changes combined decrease the overall cellular deacetylase activity that ultimately results in an increase in protein acetylation. Using traditional Western blotting and immunoprecipitation techniques chronic ethanol induces hyperacetylation of several key metabolic regulators, including PPARγ co-activator 1α, sterol regulatory binding element protein 1 (SREBP-1c) and forkhead transcription factor 1.

In recent experiments, mass spectrometry and proteomic approaches have been applied to identify proteins and pathways that are acetylated following chronic ethanol consumption. Using whole cell extracts and matrix-assisted laser desorption mass spectrometry, Shepard et al[30] identified 40 proteins that are acetylated following chronic ethanol administration. Pathway analysis revealed that of these 40, the majority was predominantly mitochondrial proteins and there was a significant preference for proteins regulating lipid metabolism as well as oxidative stress[29,30]. More recently using mitochondrial enriched fractions isolated from ethanol-fed wild-type and SIRT3 knockout mice, we determined that chronic ethanol impacted acetylation of proteins regulating lipid metabolism, oxidative stress, as well as mitochondrial pathways including amino acid biosynthesis and the electron transport chain[32].

SUMOYLATION

A member of the ubiquitin family, SUMO, comprised of four distinct proteins in humans (SUMO-1, -2, -3 and -4), is receiving growing interest since its discovery less than a decade ago[36]. SUMO-4 shows similarity to -2/3 but it is as yet unclear whether it is a pseudogene or merely restricted in its expression pattern[37]. The sumoylation cycle is a multistep process, involving maturation, activation, conjugation and deconjugation, and regulates the function and fate of a large number of proteins involved in many cellular pathways including transcription, intracellular transport, DNA repair, replication, and cell signaling[38,39]. Sumoylation, as an enzymatic cascade, resembles that of ubiquitination, including an ATP dependent step, the E1-activating enzyme Aos1/Uba2 (SAE1/SAE2) forms a thioester bond between its catalytic cysteine (Uba2 C173) and the C-terminal carboxy group of mature SUMO. From there, SUMO is transferred to the catalytic cysteine (C93) of the E2-conjugating enzyme (Ubc9). In the last step of this cascade, an isopeptide bond is formed between SUMO and the 3-amino group of a lysine side chain. Specific isopeptidases, members of the SENP family, ensure reversibility of this modification[40,41]. Sumoylation is often increased under oxidative stress[42]. Recent reports demonstrate that ubiquitin conjugating enzyme 9 (Ubc9), the sole E2 enzyme of sumoylation, is induced in ethanol treated mice[43]. However, the functional significance of this finding remains unknown. However, a number of sumoylated proteins have been identified in the liver after ethanol administration and other injury models. A notable example is the enzyme methionine adenosyltransferase II α (MATα2) which has been shown to increase upon ethanol exposure[44] is sumoylated. This modification likely plays a critical role in its stability[45].

Nrf2, a well-characterized transcription factor is known for its role in activating anti-oxidant response element (ARE), forms heterodimers with small Maf (MafG, MafK and MafF) proteins. We recently reported that Nrf2 and MafG are sumoylated and this facilitates their heterodimerization and trans-activation of the ARE in activated hepatic stellate cells[46].

Increased levels of lipopolysaccharide (LPS), a major component of the cell wall of gram-negative bacteria, is frequently found in cirrhotic patients[47] and is observed to lower glutathione (GSH), a potent anti-oxidant. GSH is highly concentrated in the liver and synthesized in the cytosol in a tightly regulated manner. Key determinants of GSH synthesis are the availability of the sulfur amino acid precursor, cysteine, and the activity of the rate-limiting enzyme, glutamate cysteine ligase, which is composed of a catalytic and a modifier (GCLM) subunit. LPS inhibits the sumoylation machinery suppressing the expression of the sole E2 enzyme Ubc9. This results in reduced Nrf2 and MafG sumoylation affecting their heterodimerization and trans-activation of the ARE present in GCLC and GCLM in macrophages and hepatocytes[48].

Although considerable progress has been made in the identification of sumoylated proteins and the characterization of the effects of the modification of these particular substrates, little is known in regards to the global regulation of SUMO conjugation in ALD.

ISOASPARTYL DAMAGE

Ethanol consumption specifically triggers a unique protein post-translational damage as isoaspartate peptide linkages in proteins[49]. This protein isoaspartate damage is due to an inhibition of the protein repair enzyme, protein isoaspartyl methyltransferase (PIMT).

PIMT normally acts to resist the accumulation of isoaspartate damage that arises through protein aging, and as a consequence of oxidative damage to proteins[50]. PIMT is a methyltransferase that utilises S-adenosylmethionine (SAM) as a methyl donor. PIMT methylates isoaspartate residues in peptides and proteins, a process that triggers isoaspartate elimination and restoration of protein function. One of the detrimental actions of ethanol consumption is impaired methionine synthase-catalysed remethylation of homocysteine to generate methionine, the metabolic precursor of SAM. A subsequent depletion of SAM availability limits the activity of SAM-dependent methyltransferases, such as PIMT. This inhibition of methylation reactions is further exacerbated by the ethanol-induced increase in the level of S-adenosylhomocysteine (SAH), a potent inhibitor of numerous SAM-dependent methyltransferases including PIMT[49,51-54].

Animal studies have demonstrated the benefits of dietary supplementation with betaine, a pro-methylating agent, to counter some of the ethanol-induced changes in the metabolite levels of the methionine metabolic pathway[49,51-54]. Rats fed a control or ethanol liquid diet (36% of calories) for a period of 4 wk with or without dietary supplementation with 1% betaine revealed that the ethanol-induced reduction of the methylation potential (i.e,. the lowering of the hepatocellular SAM:SAH ratio to approximately 43% of control level) was rectified in rats fed an ethanol diet supplemented with betaine[49,51-54]. Concomitant with the changes in the SAM:SAH ratio, the ethanol-induced increase of damage to cellular proteins as isoaspartate was also alleviated by betaine supplementation[49,51-54].

A proteomic approach was adopted to investigate the mechanism by which betaine was able to alleviate the ethanol-induced increase of isoaspartate damage. One dimensional and two dimensional protein separations and differential protein staining techniques revealed that betaine supplementation increased the expression of betaine homocysteine methyltransferase-1, methionine adenosyl transferase-1 and glycine N-methyltransferase, and these enzymes act to collectively increase SAM levels and normalise the SAM:SAH ratio[55].

To further investigate the influence of the SAM:SAH ratio on PIMT activity and cellular isoaspartate damage, primary hepatocytes taken from control or ethanol-fed rats were cultured. Cells were incubated in vitro with tubercidin or adenosine, agents that elevate cellular SAH levels[56]. These agents produced an additive increase of isoaspartate damage to that detected from ethanol consumption, indicative of an additional lowering of the SAM:SAH ratio and further inhibition of PIMT activity.

To identify liver protein target(s) of PIMT that accrue isoaspartate damage after ethanol consumption, proteins from control and ethanol fed rats were exogenously methylated using PIMT and 3H-SAM methyl donor. Novel, sensitive autoradiographic imaging[57] was used to reveal increased isoaspartate methylation at liver protein bands of 75-80 kDa, 95-100 kDa, and 155-160 kDa. Column chromatography used to enrich isoaspartate-damaged liver proteins indicated that damaged proteins from ethanol-fed rats mirrored those that accumulate in the livers of PIMT knockout mice. The about 160 kDa protein target of PIMT was further purified and fractionated, and identified as carbamoyl phosphate synthase-1 (CPS-1)[58]. This is a mitochondrial enzyme that catalyses the synthesis of carbamoyl phosphate from ammonia and bicarbonate, and is the first and rate-limiting step of the urea cycle. Resolution of liver proteins by one dimensional polyacrylamide gel electrophoresis and Coomassie blue staining also showed that cytosolic CPS-1 protein levels increased by approximately 20% in rats administered ethanol for 4 wk. A subsequent study of ethanol administration for 8 wk showed that the levels of cytosolic CPS-1 now increased approximately 2-fold over those of control animals; indicating that cytosolic CPS-1 levels correlated with the duration of alcohol consumption. Increased cytosolic CPS-1 was also detected in PIMT knockout mice compared to their control littermates. This release of liver CPS-1 into the cytosol as a response to ethanol consumption or in PIMT knockout mice is presumed to reflect mitochondrial damage and redox stress.

These studies highlight the accumulation of atypical isoaspartate-containing abnormal proteins following chronic ethanol exposure. The animal studies employed, however, are of relatively acute alcohol administration, and it is hypothesised that in alcoholic patients sustained and cumulative isoaspartate protein damage across a broad number of target proteins would ensue and contribute to liver cell damage and pathology.

METHYLATION

Here, we will discuss the role of this post-translational modification in the regulation of innate immunity in hepatitis C virus (HCV) infection combined with ethanol exposure. About 3% of world population is infected with HCV, the most common blood-borne infection in the United States. By 2010, 2.7-3.9 million people were diagnosed with chronic HCV-infection, and there are about 17000 new cases of acute infection per year. Hepatitis C and alcohol are the most widespread causes of liver disease worldwide, and approximately 80% of patients with a history of Hepatitis C and alcohol abuse develop chronic liver injury[59]. Almost one-third of alcoholics with clinical symptoms of liver disease have been infected with HCV, which is four times the rate of HCV infection found in alcoholics who do not have liver disease. Alcohol consumption in HCV-infected patients exacerbates liver disease leading to rapid progression to fibrosis, cirrhosis and even hepatocellular carcinoma[60]. Alcohol-consumption reduces responsiveness of HCV patients to anti-viral treatment; only 7% of heavy drinking HCV patients are responders to interferon therapy[61]. However, despite the direct acting anti-viral agents (DAA) changing the treatment backbone of HCV infection from IFN-ribavirin, the effectiveness of DAA would also depend on the endogenous IFNα-mediated activation of antiviral genes in HCV-infected hepatocytes.

The mechanism by which alcohol consumption exacerbates the course of HCV progression is not clear. Since HCV and alcohol alter innate immunity in hepatocytes, there is a strong possibility that their synergistic effect on innate immunity contributes to HCV spread and progressive liver injury. Activation of an anti-viral innate immune response is based on IFN signaling, which requires activation of IFN-sensitive genes (ISG) via the Janus kinase/signal transducers and activators of transcription (JAK-STAT) pathway. Transduction of IFNα signal requires phosphorylation of STAT1 and STAT2. The attachment of STAT1 to DNA becomes possible if STAT1 is methylated on arginine residue(s). It has been shown that HCV subverts the IFNα-mediated JAK-STAT signaling through the reduction of intrahepatic STAT-1 and -2 phosphorylation[62] and reduces STAT-1 methylation leading to suppression of ISGs[63,64].

Ethanol also is known to suppress methylation reactions leading to impaired methylation of multiple proteins and enzymes[49,51-54]. We hypothesize that ethanol potentiates HCV-mediated impairment of methylation-regulated IFN signaling in liver cells, thereby decreasing antiviral protection in liver cells. When HCV-infected Huh7.5-CYP2E1 cells were exposed to an extracellular system that continuously generates the ethanol metabolite, acetaldehyde (Ach) in physiological quantities, STAT1 methylation was suppressed on both arginine and lysine residues[65]. Suppression of STAT1 methylation is regulated by protein arginine N-methyltransferase 1 and lysine methyltransferases, and can be induced by specific methyltransferase inhibitors. The effects of Ach on STAT1 methylation are protein phosphatase 2 dependent. The impaired methylation of STAT1 increases the complex formation between STAT1 and the pathway inhibitor, protein inhibitor of activated STAT-1 (PIAS1), preventing the attachment of IFN-activated STAT1 to DNA followed by antiviral gene activation. This mechanism is schematically presented as Figure 1. Methylation-dependent dysregulations of IFN signaling in hepatocytes were attenuated by supplementation with the pro-methylating agent, betaine[65].

Figure 1
Figure 1 Acetaldehyde suppresses interferon-α signaling in hepatitis C virus -infected liver cells by impairing signal transducers and activators of transcription 1 methylation. The most downstream event in interferon (IFN)α signaling is the attachment of methylated STAT1 to DNA, interferon stimulated response element (ISRE) and gamma-interferon activated site (GAS), for activation of anti-viral interferon-stimulated genes (ISGs). Acetaldehyde suppresses STAT1 methylation, which facilitates increased STAT1 interaction with protein inhibitor of activated STAT 1 (PIAS1, a negative regulator of IFN signaling) preventing STAT1 binding to DNA. This ultimately results in reduced ISG activation and decreased induction of anti-viral proteins.

Thus, Ach potentiates the ability of HCV to down-regulate activation of ISGs by interferon and plays a pivotal role in methylation-dependent suppression of innate immunity in hepatocytes, which are primary sites of both HCV replication and ethanol metabolism.

OXIDIZED METABOLITES OF FATTY ACIDS

Alcohol administration results in increased production of enzymatic or non-enzymatic lipid oxidation products, which may also cause protein modifications and potential functional damage to proteins[11] as well as impact on the function of various metabolic pathways exacerbating alcohol-induced hepatic injury. Many lines of evidence, from animals to humans, have shown that dietary factors, including dietary fat, along with heavy alcohol consumption, play critical roles in the ALD pathogenesis. Indeed, the relative beneficial effects of dietary saturated fat (SF) and damaging effects of dietary unsaturated fat [USF, primarily corn oil/linoleic acid (LA) enriched] on alcohol-induced liver injury have been well documented in experimental animal models of ALD[66-72]. A number of mechanisms have been proposed for the opposing effects of dietary USF vs SF in ALD, including (1) induction of lipid peroxidation and oxidative stress[70,73,74]; (2) altered gut microbiota, impaired intestinal barrier integrity, endotoxemia, and associated increase in liver pro-inflammatory cytokine production[66,67,72]; (3) modulation of hepatic lipid metabolism via SIRT1-SREBP-1-histone H3 axis[75]; and (4) modulation of hepatocyte nuclear factor-4α expression, a master transcription factor in the regulation of lipid metabolism[76]. A new concept has recently emerged that the bioactive oxidized LA metabolites (OXLAMs), which are formed enzymatically from LA primarily via the actions of 12/15-lipoxygenase (12/15-LOX), or non-enzymatically via free radical-mediated oxidation in response to oxidative stress, might contribute to ALD pathogenesis. It has been demonstrated that plasma OXLAMs, specifically 9- and 13-hydroxy-octadecadienoic acids (9- and 13-HODEs), were elevated in patients with alcoholic cirrhosis in parallel with the increase in lipoxygenases (15-LOX-1 and 15-LOX-2 mRNA) in the liver samples. The plasma levels of HODEs in patients with ALD were significantly higher than in healthy subjects as well as in NAFLD patients[77]. Further, increased levels of 9- and 13-HODEs were observed in experimental animal models of ALD[78,79] in parallel with the hepatic steatosis, oxidative stress, and inflammation and hepatocyte damage. It has been reported that 9- and 13-HODEs are natural endogenous ligands for the Transient Receptor Potential Vanilloid 1 (TRPV1)[80,81]. Our recent study demonstrated that chronic-binge ethanol-mediated increases in circulating OXLAMs and TRPV1 levels in mice were associated with hepatic steatosis, inflammation and injury[78]. Genetic depletion of TRPV1 did not blunt hepatic steatosis caused by ethanol, but prevented hepatic injury. TRPV1 deficiency protected from hepatocyte death and prevented the increase in pro-inflammatory cytokine and chemokine expression, including TNF-α, interleukin-6, macrophage inflammatory protein-2 and monocyte chemotactic protein-1. Moreover, TRPV1 depletion markedly blunted ethanol-mediated induction of plasminogen activator inhibitor-1, an important mediator of alcohol-induced hepatic inflammation, via fibrin accumulation[78]. Exposure of HepG2 cells to 9- and 13-HODEs resulted in activation of TRPV1 signal transduction with the increased intracellular Ca2+ levels, suggesting that OXLAM/TRPV1/Ca2+ signaling may be a relevant pathway contributing to ALD pathogenesis.

Alcohol consumption increases hepatic oxidative stress with the production of reactive oxygen species. One of the major sources of in vivo protein modification during oxidative stress is thought to be oxidative products of polyunsaturated fatty acids (PUFAs). LA is the most abundant PUFA in mammalian tissue. Non-enzymatic oxidative degradation of PUFAs, including LA, generate a variety of lipid peroxidation products (LPOs, e.g., MDA, HNE, acrolein, various epoxyketooctadecenoic acid isomers)[11]. Enhanced amounts of peroxidized phospholipids and their truncation products in the circulation have previously been observed in rats with alcohol-induced liver disease[79]. Some LPOs can modify DNA, peptides and proteins leading to formation of advanced lipoxidation end-products (ALEs). These modifications can potentially cause functional damage to proteins. Numerous LPOs and ALEs exert diverse biologic activities (e.g., damaging and pro-inflammatory effects in some cases) through different as yet not well-defined mechanisms. The role and the significance of oxidized lipids, both dietary and in vivo-produced, as well as possible mechanisms underlying their beneficial or deleterious effects in liver pathology remain to be determined.

CONCLUSION

In summary, chronic ethanol consumption dysregulates post-translational modifications of numerous important proteins that regulate many cellular processes. Not surprisingly, there is considerable overlap in the pathways that are targeted. Understanding how each individual modification affects specific protein function and thereby, alters metabolic pathways will be of critical importance to deciphering the impact of the aforementioned modifications to alcohol- induced steatosis and hepatocellular damage. It could also provide an insight into disease pathogenesis and progression, and may help to identify additional useful targets of drug action. In addition, the activation of enzymatic and/or non-enzymatic degradation of polyunsaturated fatty acids resulting in the formation of numerous bioactive lipid compounds underlies the deleterious effects of certain dietary fat intake in promoting indices of alcoholic liver damage.

Footnotes

Manuscript source: Invited manuscript

P- Reviewer: Balaban YH, Kadayifci A S- Editor: Ma YJ L- Editor: A E- Editor: Ma S

References
1.  Global status report on alcohol and health. Geneva: World Health Organization 2011; .  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Massey VL, Arteel GE. Acute alcohol-induced liver injury. Front Physiol. 2012;3:193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 87]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
3.  Zakhari S. Overview: how is alcohol metabolized by the body? Alcohol Res Health. 2006;29:245-254.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Cederbaum AI. Alcohol metabolism. Clin Liver Dis. 2012;16:667-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 767]  [Cited by in F6Publishing: 667]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
5.  Ishak KG, Zimmerman HJ, Ray MB. Alcoholic liver disease: pathologic, pathogenetic and clinical aspects. Alcohol Clin Exp Res. 1991;15:45-66.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Crabb DW. Pathogenesis of alcoholic liver disease: newer mechanisms of injury. Keio J Med. 1999;48:184-188.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Altamirano J, Bataller R. Alcoholic liver disease: pathogenesis and new targets for therapy. Nat Rev Gastroenterol Hepatol. 2011;8:491-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 197]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
8.  Gao B, Bataller R. Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology. 2011;141:1572-1585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1244]  [Cited by in F6Publishing: 1342]  [Article Influence: 103.2]  [Reference Citation Analysis (0)]
9.  Orman ES, Odena G, Bataller R. Alcoholic liver disease: pathogenesis, management, and novel targets for therapy. J Gastroenterol Hepatol. 2013;28 Suppl 1:77-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 172]  [Article Influence: 15.6]  [Reference Citation Analysis (2)]
10.  Ji C. Advances and New Concepts in Alcohol-Induced Organelle Stress, Unfolded Protein Responses and Organ Damage. Biomolecules. 2015;5:1099-1121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
11.  Zhu X, Tang X, Anderson VE, Sayre LM. Mass spectrometric characterization of protein modification by the products of nonenzymatic oxidation of linoleic acid. Chem Res Toxicol. 2009;22:1386-1397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
12.  Aubert J, Begriche K, Knockaert L, Robin MA, Fromenty B. Increased expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: mechanisms and pathophysiological role. Clin Res Hepatol Gastroenterol. 2011;35:630-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 197]  [Cited by in F6Publishing: 203]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
13.  Leung TM, Nieto N. CYP2E1 and oxidant stress in alcoholic and non-alcoholic fatty liver disease. J Hepatol. 2013;58:395-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 345]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
14.  Enomoto N, Takei Y, Yamashina S, Ikejima K, Kitamura T, Sato N. Anti-inflammatory strategies in alcoholic steatohepatitis. J Gastroenterol Hepatol. 2007;22 Suppl 1:S59-S61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
15.  Albano E. Alcohol, oxidative stress and free radical damage. Proc Nutr Soc. 2006;65:278-290.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Wu KC, Liu J, Klaassen CD. Role of Nrf2 in preventing ethanol-induced oxidative stress and lipid accumulation. Toxicol Appl Pharmacol. 2012;262:321-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 105]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
17.  Bondy SC. Ethanol toxicity and oxidative stress. Toxicol Lett. 1992;63:231-241.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Cederbaum AI. Introduction-serial review: alcohol, oxidative stress and cell injury. Free Radic Biol Med. 2001;31:1524-1526.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Carbone DL, Doorn JA, Kiebler Z, Petersen DR. Cysteine modification by lipid peroxidation products inhibits protein disulfide isomerase. Chem Res Toxicol. 2005;18:1324-1331.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Newton BW, Russell WK, Russell DH, Ramaiah SK, Jayaraman A. Liver proteome analysis in a rodent model of alcoholic steatosis. J Proteome Res. 2009;8:1663-1671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 48]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
21.  Galligan JJ, Smathers RL, Fritz KS, Epperson LE, Hunter LE, Petersen DR. Protein carbonylation in a murine model for early alcoholic liver disease. Chem Res Toxicol. 2012;25:1012-1021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 242]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
22.  Shearn CT, Fritz KS, Shearn AH, Saba LM, Mercer KE, Engi B, Galligan JJ, Zimniak P, Orlicky DJ, Ronis MJ. Deletion of GSTA4-4 results in increased mitochondrial post-translational modification of proteins by reactive aldehydes following chronic ethanol consumption in mice. Redox Biol. 2016;7:68-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
23.  Shearn CT, Orlicky DJ, Saba LM, Shearn AH, Petersen DR. Increased hepatocellular protein carbonylation in human end-stage alcoholic cirrhosis. Free Radic Biol Med. 2015;89:1144-1153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
24.  Ronis MJ, Mercer KE, Gannon B, Engi B, Zimniak P, Shearn CT, Orlicky DJ, Albano E, Badger TM, Petersen DR. Increased 4-hydroxynonenal protein adducts in male GSTA4-4/PPAR-α double knockout mice enhance injury during early stages of alcoholic liver disease. Am J Physiol Gastrointest Liver Physiol. 2015;308:G403-G415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
25.  Galligan JJ, Smathers RL, Shearn CT, Fritz KS, Backos DS, Jiang H, Franklin CC, Orlicky DJ, Maclean KN, Petersen DR. Oxidative Stress and the ER Stress Response in a Murine Model for Early-Stage Alcoholic Liver Disease. J Toxicol. 2012;2012:207594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
26.  Zakhari S, Li TK. Determinants of alcohol use and abuse: Impact of quantity and frequency patterns on liver disease. Hepatology. 2007;46:2032-2039.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 240]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
27.  Harris PS, Roy SR, Coughlan C, Orlicky DJ, Liang Y, Shearn CT, Roede JR, Fritz KS. Chronic ethanol consumption induces mitochondrial protein acetylation and oxidative stress in the kidney. Redox Biol. 2015;6:33-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 38]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
28.  Picklo MJ. Ethanol intoxication increases hepatic N-lysyl protein acetylation. Biochem Biophys Res Commun. 2008;376:615-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 51]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
29.  Shepard BD, Tuma DJ, Tuma PL. Chronic ethanol consumption induces global hepatic protein hyperacetylation. Alcohol Clin Exp Res. 2010;34:280-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
30.  Shepard BD, Tuma PL. Alcohol-induced protein hyperacetylation: mechanisms and consequences. World J Gastroenterol. 2009;15:1219-1230.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Ajmo JM, Liang X, Rogers CQ, Pennock B, You M. Resveratrol alleviates alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol. 2008;295:G833-G842.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Fritz KS, Galligan JJ, Hirschey MD, Verdin E, Petersen DR. Mitochondrial acetylome analysis in a mouse model of alcohol-induced liver injury utilizing SIRT3 knockout mice. J Proteome Res. 2012;11:1633-1643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 98]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
33.  Lieber CS, Leo MA, Wang X, Decarli LM. Alcohol alters hepatic FoxO1, p53, and mitochondrial SIRT5 deacetylation function. Biochem Biophys Res Commun. 2008;373:246-252.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Shen Z, Liang X, Rogers CQ, Rideout D, You M. Involvement of adiponectin-SIRT1-AMPK signaling in the protective action of rosiglitazone against alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol. 2010;298:G364-G374.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Yin H, Hu M, Zhang R, Shen Z, Flatow L, You M. MicroRNA-217 promotes ethanol-induced fat accumulation in hepatocytes by down-regulating SIRT1. J Biol Chem. 2012;287:9817-9826.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Marx J. Cell biology. SUMO wrestles its way to prominence in the cell. Science. 2005;307:836-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
37.  Bohren KM, Nadkarni V, Song JH, Gabbay KH, Owerbach D. A M55V polymorphism in a novel SUMO gene (SUMO-4) differentially activates heat shock transcription factors and is associated with susceptibility to type I diabetes mellitus. J Biol Chem. 2004;279:27233-27238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 247]  [Cited by in F6Publishing: 270]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
38.  Gill G. SUMO and ubiquitin in the nucleus: different functions, similar mechanisms? Genes Dev. 2004;18:2046-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 556]  [Cited by in F6Publishing: 559]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
39.  Hay RT. SUMO: a history of modification. Mol Cell. 2005;18:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1249]  [Cited by in F6Publishing: 1288]  [Article Influence: 67.8]  [Reference Citation Analysis (0)]
40.  Johnson ES. Protein modification by SUMO. Annu Rev Biochem. 2004;73:355-382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1332]  [Cited by in F6Publishing: 1303]  [Article Influence: 65.2]  [Reference Citation Analysis (0)]
41.  Melchior F, Schergaut M, Pichler A. SUMO: ligases, isopeptidases and nuclear pores. Trends Biochem Sci. 2003;28:612-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 320]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
42.  Le NT, Corsetti JP, Dehoff-Sparks JL, Sparks CE, Fujiwara K, Abe J. Reactive Oxygen Species, SUMOylation, and Endothelial Inflammation. Int J Inflam. 2012;2012:678190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
43.  Tomasi ML, Tomasi I, Ramani K, Pascale RM, Xu J, Giordano P, Mato JM, Lu SC. S-adenosyl methionine regulates ubiquitin-conjugating enzyme 9 protein expression and sumoylation in murine liver and human cancers. Hepatology. 2012;56:982-993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
44.  Tsukamoto H, Lu SC. Current concepts in the pathogenesis of alcoholic liver injury. FASEB J. 2001;15:1335-1349.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Tomasi ML, Ryoo M, Ramani K, Tomasi I, Giordano P, Mato JM, Lu SC. Methionine adenosyltransferase α2 sumoylation positively regulate Bcl-2 expression in human colon and liver cancer cells. Oncotarget. 2015;6:37706-37723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
46.  Ramani K, Tomasi ML, Yang H, Ko K, Lu SC. Mechanism and significance of changes in glutamate-cysteine ligase expression during hepatic fibrogenesis. J Biol Chem. 2012;287:36341-36355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
47.  Bryant CE, Spring DR, Gangloff M, Gay NJ. The molecular basis of the host response to lipopolysaccharide. Nat Rev Microbiol. 2010;8:8-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 261]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
48.  Tomasi ML, Ryoo M, Yang H, Iglesias Ara A, Ko KS, Lu SC. Molecular mechanisms of lipopolysaccharide-mediated inhibition of glutathione synthesis in mice. Free Radic Biol Med. 2014;68:148-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
49.  Kharbanda KK, Mailliard ME, Baldwin CR, Sorrell MF, Tuma DJ. Accumulation of proteins bearing atypical isoaspartyl residues in livers of alcohol-fed rats is prevented by betaine administration: effects on protein-L-isoaspartyl methyltransferase activity. J Hepatol. 2007;46:1119-1125.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Carter WG, Aswad DW. Formation, localization, and repair of L-isoaspartyl sites in histones H2A and H2B in nucleosomes from rat liver and chicken erythrocytes. Biochemistry. 2008;47:10757-10764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
51.  Kharbanda KK, Mailliard ME, Baldwin CR, Beckenhauer HC, Sorrell MF, Tuma DJ. Betaine attenuates alcoholic steatosis by restoring phosphatidylcholine generation via the phosphatidylethanolamine methyltransferase pathway. J Hepatol. 2007;46:314-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 151]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
52.  Kharbanda KK, Rogers DD, Mailliard ME, Siford GL, Barak AJ, Beckenhauer HC, Sorrell MF, Tuma DJ. Role of elevated S-adenosylhomocysteine in rat hepatocyte apoptosis: protection by betaine. Biochem Pharmacol. 2005;70:1883-1890.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Kharbanda KK. Alcoholic liver disease and methionine metabolism. Semin Liver Dis. 2009;29:155-165.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Kharbanda KK. Methionine metabolic pathway in alcoholic liver injury. Curr Opin Clin Nutr Metab Care. 2013;16:89-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
55.  Kharbanda KK, Vigneswara V, McVicker BL, Newlaczyl AU, Bailey K, Tuma D, Ray DE, Carter WG. Proteomics reveal a concerted upregulation of methionine metabolic pathway enzymes, and downregulation of carbonic anhydrase-III, in betaine supplemented ethanol-fed rats. Biochem Biophys Res Commun. 2009;381:523-527.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Kharbanda KK, Todero SL, Moats JC, Harris RM, Osna NA, Thomes PG, Tuma DJ. Alcohol consumption decreases rat hepatic creatine biosynthesis via altered guanidinoacetate methyltransferase activity. Alcohol Clin Exp Res. 2014;38:641-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
57.  Tarhoni MH, Vigneswara V, Smith M, Anderson S, Wigmore P, Lees JE, Ray DE, Carter WG. Detection, quantification, and microlocalisation of targets of pesticides using microchannel plate autoradiographic imagers. Molecules. 2011;16:8535-8551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
58.  Carter WG, Vigneswara V, Newlaczyl A, Wayne D, Ahmed B, Saddington S, Brewer C, Raut N, Gerdes HK, Erdozain AM. Isoaspartate, carbamoyl phosphate synthase-1, and carbonic anhydrase-III as biomarkers of liver injury. Biochem Biophys Res Commun. 2015;458:626-631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
59.  Jamal MM, Morgan TR. Liver disease in alcohol and hepatitis C. Best Pract Res Clin Gastroenterol. 2003;17:649-662.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Poynard T, Bedossa P, Opolon P. Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet. 1997;349:825-832.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Feeney ER, Chung RT. Antiviral treatment of hepatitis C. BMJ. 2014;348:g3308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 153]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
62.  Gunduz F, Mallikarjun C, Balart LA, Dash S. Interferon alpha induced intrahepatic pSTAT1 inversely correlate with serum HCV RNA levels in chronic HCV infection. Exp Mol Pathol. 2014;96:36-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
63.  Duong FH, Christen V, Berke JM, Penna SH, Moradpour D, Heim MH. Upregulation of protein phosphatase 2Ac by hepatitis C virus modulates NS3 helicase activity through inhibition of protein arginine methyltransferase 1. J Virol. 2005;79:15342-15350.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Duong FH, Filipowicz M, Tripodi M, La Monica N, Heim MH. Hepatitis C virus inhibits interferon signaling through up-regulation of protein phosphatase 2A. Gastroenterology. 2004;126:263-277.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Ganesan M, Zhang J, Bronich T, Poluektova LI, Donohue TM, Tuma DJ, Kharbanda KK, Osna NA. Acetaldehyde accelerates HCV-induced impairment of innate immunity by suppressing methylation reactions in liver cells. Am J Physiol Gastrointest Liver Physiol. 2015;309:G566-G577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
66.  Chen P, Torralba M, Tan J, Embree M, Zengler K, Stärkel P, van Pijkeren JP, DePew J, Loomba R, Ho SB. Supplementation of saturated long-chain fatty acids maintains intestinal eubiosis and reduces ethanol-induced liver injury in mice. Gastroenterology. 2015;148:203-214.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 214]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
67.  Kirpich IA, Feng W, Wang Y, Liu Y, Barker DF, Barve SS, McClain CJ. The type of dietary fat modulates intestinal tight junction integrity, gut permeability, and hepatic toll-like receptor expression in a mouse model of alcoholic liver disease. Alcohol Clin Exp Res. 2012;36:835-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 96]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
68.  Mezey E. Dietary fat and alcoholic liver disease. Hepatology. 1998;28:901-905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 69]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
69.  Nanji AA. Role of different dietary fatty acids in the pathogenesis of experimental alcoholic liver disease. Alcohol. 2004;34:21-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 58]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
70.  Ronis MJ, Korourian S, Zipperman M, Hakkak R, Badger TM. Dietary saturated fat reduces alcoholic hepatotoxicity in rats by altering fatty acid metabolism and membrane composition. J Nutr. 2004;134:904-912.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  You M, Considine RV, Leone TC, Kelly DP, Crabb DW. Role of adiponectin in the protective action of dietary saturated fat against alcoholic fatty liver in mice. Hepatology. 2005;42:568-577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 209]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
72.  Zhong W, Li Q, Xie G, Sun X, Tan X, Sun X, Jia W, Zhou Z. Dietary fat sources differentially modulate intestinal barrier and hepatic inflammation in alcohol-induced liver injury in rats. Am J Physiol Gastrointest Liver Physiol. 2013;305:G919-G932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
73.  Nanji AA, Zhao S, Lamb RG, Dannenberg AJ, Sadrzadeh SM, Waxman DJ. Changes in cytochromes P-450, 2E1, 2B1, and 4A, and phospholipases A and C in the intragastric feeding rat model for alcoholic liver disease: relationship to dietary fats and pathologic liver injury. Alcohol Clin Exp Res. 1994;18:902-908.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Kono H, Enomoto N, Connor HD, Wheeler MD, Bradford BU, Rivera CA, Kadiiska MB, Mason RP, Thurman RG. Medium-chain triglycerides inhibit free radical formation and TNF-alpha production in rats given enteral ethanol. Am J Physiol Gastrointest Liver Physiol. 2000;278:G467-G476.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  You M, Cao Q, Liang X, Ajmo JM, Ness GC. Mammalian sirtuin 1 is involved in the protective action of dietary saturated fat against alcoholic fatty liver in mice. J Nutr. 2008;138:497-501.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Li Q, Zhong W, Qiu Y, Kang X, Sun X, Tan X, Zhao Y, Sun X, Jia W, Zhou Z. Preservation of hepatocyte nuclear factor-4α contributes to the beneficial effect of dietary medium chain triglyceride on alcohol-induced hepatic lipid dyshomeostasis in rats. Alcohol Clin Exp Res. 2013;37:587-598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
77.  Raszeja-Wyszomirska J, Safranow K, Milkiewicz M, Milkiewicz P, Szynkowska A, Stachowska E. Lipidic last breath of life in patients with alcoholic liver disease. Prostaglandins Other Lipid Mediat. 2012;99:51-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
78.  Liu H, Beier JI, Arteel GE, Ramsden CE, Feldstein AE, McClain CJ, Kirpich IA. Transient receptor potential vanilloid 1 gene deficiency ameliorates hepatic injury in a mouse model of chronic binge alcohol-induced alcoholic liver disease. Am J Pathol. 2015;185:43-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
79.  Yang L, Latchoumycandane C, McMullen MR, Pratt BT, Zhang R, Papouchado BG, Nagy LE, Feldstein AE, McIntyre TM. Chronic alcohol exposure increases circulating bioactive oxidized phospholipids. J Biol Chem. 2010;285:22211-22220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
80.  Patwardhan AM, Akopian AN, Ruparel NB, Diogenes A, Weintraub ST, Uhlson C, Murphy RC, Hargreaves KM. Heat generates oxidized linoleic acid metabolites that activate TRPV1 and produce pain in rodents. J Clin Invest. 2010;120:1617-1626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 188]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
81.  Patwardhan AM, Scotland PE, Akopian AN, Hargreaves KM. Activation of TRPV1 in the spinal cord by oxidized linoleic acid metabolites contributes to inflammatory hyperalgesia. Proc Natl Acad Sci USA. 2009;106:18820-18824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 176]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]