Mitochondrial dysfunction and oxidative stress in the pathogenesis of alcohol- and obesity-induced fatty liver diseases.
Journal: 2008/June - Free Radical Biology and Medicine
ISSN: 0891-5849
Abstract:
Fatty liver disease associated with chronic alcohol consumption or obesity/type 2 diabetes has emerged as a serious public health problem. Steatosis, accumulation of triglyceride in hepatocytes, is now recognized as a critical "first-hit" in the pathogenesis of liver disease. It is proposed that steatosis "primes" the liver to progress to more severe liver pathologies when individuals are exposed to subsequent metabolic and/or environmental stressors or "second-hits." Genetic risk factors can also influence the susceptibility to and severity of fatty liver disease. Furthermore, oxidative stress, disrupted nitric oxide (NO) signaling, and mitochondrial dysfunction are proposed to be key molecular events that accelerate or worsen steatosis and initiate progression to steatohepatitis and fibrosis. This review article will discuss the following topics regarding the pathobiology and molecular mechanisms responsible for fatty liver disease: (1) the "two-hit" or "multi-hit" hypothesis, (2) the role of mitochondrial bioenergetic defects and oxidant stress, (3) the interplay between NO and mitochondria in fatty liver disease, (4) genetic risk factors and oxidative stress-responsive genes, and (5) the feasibility of antioxidants for treatment.
Relations:
Content
Citations
(124)
References
(191)
Pathways
(1)
Diseases
(1)
Conditions
(3)
Chemicals
(4)
Organisms
(2)
Processes
(1)
Anatomy
(2)
Affiliates
(2)
Similar articles
Articles by the same authors
Discussion board
Free Radic Biol Med 44(7): 1259-1272

Mitochondrial Dysfunction and Oxidative Stress in the Pathogenesis of Alcohol and Obesity Induced Fatty Liver Diseases

INTRODUCTION

Heavy, prolonged alcohol consumption is estimated to be the third leading cause of all preventable deaths in the US with up to 12,000 deaths each year specifically due to alcohol induced liver disease [1]. Moreover, with the increasing prevalence of obesity and type 2 diabetes, non-alcoholic fatty liver disease (NAFLD) has become a serious medical problem in many developed countries. In general, the spectrum of liver pathology in these circumstances runs the gamut from simple steatosis (triglyceride accumulation in hepatocytes) to the more serious conditions of steatohepatitis, fibrosis, and cirrhosis. Moreover, it should be pointed out that steatosis, while reversible, is not benign, and may actually “prime” the liver to progress to more serious pathologies including hepatocellular carcinoma. In the case of alcohol induced liver injury there is a longstanding belief that the severity of disease is dependent on the cumulative dose and duration of alcohol consumption, with decades of heavy drinking needed to induce pathology. However, studies show that only about 25% of heavy drinkers develop steatohepatitis with less than 10% developing alcoholic cirrhosis [2,3]. Based on this, it has been suggested that exposure to environmental toxicants like cigarette smoke or the presence of pre-existing conditions like type 2 diabetes, obesity, hypercholesterolemia, and/or hyperglycemia; conditions linked to the cardiometabolic syndrome, could worsen liver disease in the chronic alcohol consumer. Importantly, epidemiologic evidence demonstrates that alcohol, tobacco smoke, and obesity are synergistic risk factors for hepatocellular carcinoma [4]. Similarly, short-term binge alcohol exposure increases apoptosis and liver injury in obese rats compared to lean controls via increased oxidative stress [5]. Thus, there is clearly support for the concept that conditions of the cardiometabolic syndrome may exacerbate the hepatotoxic potential of alcohol in the liver and vice-versa.

Almost 30 years ago, Ludwig et al. [6] first coined the term non-alcoholic steatohepatitis (NASH) as a type of fatty liver disease that was histologically identical to alcoholic steatohepatitis (ASH), but occurred in overweight and/or type 2 diabetic patients who drank less than 20–40 g of ethanol/day (i.e. 1.5–3.5 drinks per day). Affected individuals typically present with moderate to severe steatosis and lobular inflammation with fibrosis in 70% and cirrhosis in 15% of patients. Since these early studies, it is currently estimated that the prevalence of NAFLD in the general US population is reaching 20% with the more severe form, NASH present in 3% [7,8]. Even more important, may be the fact that NAFLD is present in the pediatric population with prevalence estimated at 2–10% among children and adolescents in the US and Asia [9]. NASH is characterized histologically by macrosteatosis, lobular inflammation, hepatocyte ballooning, and pericellular fibrosis in the centri-lobular (zone 3) region of the liver lobule. Multiple factors have been associated with NAFLD including obesity, central adiposity, type 2 diabetes, and hyperlipidemia with insulin resistance postulated as an essential factor. Interestingly, the role of insulin resistance in alcohol-dependent liver has been invoked; however, the data in support of this mechanism is equivocal [1012].

“Two-Hit” or “Multi-Hit” Hypothesis for Alcoholic and Non-Alcoholic Fatty Liver Disease

Mechanisms of alcoholic liver disease have been extensively studied; however, the events that cause progression to hepatitis and fibrosis/cirrhosis are still undefined. The same can also be said for the pathogenesis of NAFLD. Currently, the leading hypothesis for both alcohol and non-alcohol induced liver disease is the “two-hit” or “multi-hit” hypothesis with the “first-hit” being the accumulation of triglyceride in hepatocytes (steatosis) followed by “second-hits” which trigger the progression to more serious liver pathologies like steatohepatitis and fibrosis/cirrhosis (Figure 1). Examples of “second-hits” could include environmental stressors like dietary components, cigarette smoke, or pollutants and/or metabolic stressors such as hyperglycemia, hypertriglyceridemia, and hypercholesterolemia, which are components linked to the cardiometabolic syndrome. Interestingly, what these factors all have in common is the ability to induce oxidative stress arising largely in part via mitochondrial defects. It has been proposed that the elusive “second-hit” arises from the enhanced production of reactive oxygen and/or nitrogen species (ROS/RNS) in liver [13]. Once steatosis is present, the liver becomes more susceptible to the “second-hit”; oxidative/nitrosative/nitrative stress, which is thought to be one of several stimuli for the progression from simple fatty liver to ASH or NASH. The molecular mechanism underlying how steatosis predisposes liver to transition from simple fatty liver to steatohepatitis is not clear; however, several studies point to the possibility that when hepatocytes accumulate fat the fatty acids themselves are toxic and initiate a pathological response called “lipotoxicity” [14]. Studies show that free fatty acids are toxic to hepatocytes via deregulation of lysosomal metabolism [15,16] and induction of endoplasmic reticulum stress [17,18] resulting in apoptosis. Similarly, palmitate induced lipid accumulation in hepatocytes stimulated production of the neutrophil chemoattractant interleukin-8, which could have the effect of initiating hepatic inflammation and injury [19]. The oxidation and release of reactive lipid species (i.e. lipid peroxidation products) present in fat deposits may also contribute to liver injury through the abilities of these molecules to adduct to cellular macromolecules thereby disrupting function. For example, reactive lipid species have been shown to bind to mitochondrial proteins, induce mitochondrial ROS production, and promote cytochrome c release [20,21]. Therefore, increased reactive lipid species may be a critical factor predisposing liver to more severe injury when exposed to other insults or “hits”. Finally, as both a site for fat metabolism and the main source of ROS/RNS in hepatocytes, the mitochondrion is postulated to play a central role in the development of alcoholic and non-alcoholic liver disease and this will be discussed in more detail later in the article.

An external file that holds a picture, illustration, etc.
Object name is nihms46170f1.jpg
Pathophysiology of alcoholic steatohepatitis (ASH) and non-alcoholic steatohepatitis (NASH)

To understand the molecular mechanisms by which chronic alcohol consumption and the cardiometabolic syndrome cause liver disease, it is important to define the pathways leading to fat accumulation in liver. Excess fat accumulation in liver can result from one or a combination of the following metabolic alterations:

  1. decreased β-oxidation of fatty acids;

  2. increased fatty acid synthesis due to up-regulation of lipogenic pathways;

  3. increased delivery of fatty acids from adipose and other organs due to lipolysis; and

  4. inhibition of VLDL-triglyceride export.

In general, it is the metabolic consequence of ethanol oxidation; increase in the NADH/NAD ratio, which disrupts fat metabolism leading to triglyceride accumulation in hepatocytes. Increased NADH leads to increased esterification of fatty acids into triglyceride and increased fatty acid synthesis, while inhibiting fatty acid β-oxidation and tricarboxylic (TCA) cycle activity. In the case of NAFLD, insulin resistance is thought to be responsible for the “first-hit”, steatosis, through the mobilization of peripheral fat to the liver through the development of hyperinsulinemia. Peripheral lipolysis and hyperinsulinemia lead to fatty liver through increased hepatic uptake of fatty acids and accumulation of fat via increased fatty acid synthesis, which again can be accompanied by decreased β-oxidation activity [22]. Recent data also suggests that peripheral lipolysis may contribute to alcohol-induced fatty liver in response to an alcohol-dependent suppression of insulin’s anti-lipolytic action in adipocytes [23]. While emerging data indicates that alcohol dependent insulin resistance may contribute to the accumulation of fat in hepatocytes, the evidence in support of this is less clear; primarily due to observations that alcohol has “dose-dependent” effects on insulin signaling [24]. Studies by Badger and colleagues have demonstrated that while high dose chronic alcohol consumption impairs hepatic insulin signaling, low dose ethanol exposure activates insulin signaling in hepatocytes [24,25]. Importantly, for the first time, these findings provide biochemical support for epidemiologic evidence suggesting that while low dose alcohol consumption may be cardio-protective and decrease the risk of developing type 2 diabetes; heavy, prolonged alcohol consumption increases the risk for type 2 diabetes.

It should also be mentioned that steatosis induced by chronic alcohol consumption and/or type 2 diabetes can be directly linked to two critical signaling pathways that increase lipogenesis in the liver; AMP kinase inhibition and SREBP-1 (sterol regulatory element binding protein-1) activation. AMP kinase, a protein kinase that inhibits lipogenesis and activates fatty acid oxidation, is inhibited by chronic alcohol feeding, which results in increased SREBP-1 activation [26,27]. As a consequence of the alcohol-dependent activation of SREBP-1, a number of lipogenic genes are up-regulated in liver contributing to enhanced lipid synthesis. Similarly, the ethanol-dependent inhibition of AMP kinase leads to activation of acetyl CoA carboxylase; enhancing malonyl CoA levels, which inhibits fatty acid uptake and β-oxidation in mitochondria. Thus, alcohol-associated inhibition of AMP kinase contributes to fat accumulation via stimulation of lipogenesis and inhibition of fat oxidation. Additionally, it has been demonstrated that activation of AMP kinase by the adipocytokine adiponectin and other agents improves both alcohol and obesity-induced fatty liver disease in rodent models [2830] demonstrating the key role of AMP kinase in the development of steatosis. Clearly, the question that still remains to be answered is, what is the mechanism through which ethanol inhibits AMP kinase activity? AMP kinase is activated by phosphorylation thus possible mechanisms of inactivation could include allosteric modification by AMP and ATP, activation of protein phosphatases, and modulation by other signaling pathways [31,32]. More recently, studies have suggested that the finely tuned mitochondrial ROS/RNS balance may also be a critical controlling factor in the activation of AMP kinase [3335]. Therefore, it is possible that chronic alcohol and obesity mediated effects on mitochondrial functioning and redox signaling inhibit AMP kinase activity thereby contributing to the development and progression of fatty liver disease. Future studies are warranted to address this very interesting and provocative new question.

Before considering the role of mitochondrial dysfunction in the pathogenesis of alcohol and non-alcohol mediated fatty liver disease, it is important to highlight emerging evidence, which indicates that steatosis may not be an unvarying feature of ASH and NASH. Recent work by the Nagy [36] and Lindros [37] laboratories have revealed that activation of the complement pathway is necessary for the development of chronic alcohol induced steatosis. Indeed, both laboratories have independently shown that, unlike wild-type mice, complement component C3 mice do not develop fatty liver when fed an ethanol-containing diet for 6 weeks [36,37]. More interesting, however, is the observation by Pritchard et al. showing that even though the C3 mice were protective against ethanol-mediated steatosis, mice still had elevated serum ALT and hepatic TNFα levels following alcohol exposure indicating hepatic inflammatory injury in the absence of steatosis [36]. In support of this, Diehl and colleagues demonstrated that inhibition of hepatocyte triglyceride synthesis and steatosis increased liver injury and fibrosis in obese mice with NASH [38]. Moreover, oleate- or palmitate-loaded HepG2 spheroids (hepatocyte-derived cells) were less susceptible to cytokine or peroxide induced cell death [39]. Taken together, these results are intriguing because they indicate that steatosis may not be the “first-hit” or a pre-requisite in the pathogenesis of ASH or NASH. In fact, steatosis may even be protective against hepatotoxicity [38]. Clearly, additional research is required to determine the role of steatosis in inflammatory and/or fibrotic liver diseases.

Mitochondria Dysfunction in Fatty Liver Diseases – Bioenergetic and Oxidative Stress

While the detrimental effects of chronic alcohol consumption on liver mitochondria have been known for many years [40], it has only been recently recognized that mitochondrial dysfunction may play a critical role in the development of NAFLD and progression to NASH [22]. Even though the molecular defects responsible for alcohol and non-alcohol dependent mitochondrial dysfunction remain to be defined, it is clear that under conditions of fatty liver disease one key functional change to mitochondria is the inability to maintain sufficient levels of ATP.

Interestingly though, this event may occur by different types of biochemical and molecular changes to mitochondria. In general, respiration in the presence of ADP (state 3 respiration) with NADH-linked substrates, fatty acids, or succinate is decreased as a consequence of chronic alcohol consumption, whereas respiration in the absence of ADP (state 4 respiration) is largely unaffected [41]. Moreover, this decrease in ADP-stimulated respiration is linked to decreased electron transport in all segments of the respiratory chain as chronic alcohol consumption decreases the activities of all the respiratory complexes, except complex II [41,42]. Several laboratories have presented strong evidence that inhibition of mitochondrial protein synthesis [43] linked to mtDNA damage [4446] and ribosomal defects [47,48] contribute, in part, to decreased functioning of the oxidative phosphorylation system following chronic alcohol consumption. These alterations translate into profound modifications to the mitochondrial proteome that encompass not only losses in the 13 mitochondrial encoded polypeptides, but also decreases in numerous nuclear encoded proteins that make up the oxidative phosphorylation complexes [46]. Proteomic analyses have also shown alcohol-dependent changes in mitochondrial matrix enzymes both at the level of abundance and post-translational modifications [46,49,50].

Like chronic alcohol induced liver injury, emerging evidence strongly implicates mitochondrial dysfunction contributing to obesity induced fatty liver disease (i.e. NAFLD). Early studies by Diehl and colleagues reported that obesity induced steatosis was associated with increased expression of uncoupling protein-2 (UCP-2) in liver, which promoted ATP depletion [51]. This results from increased H leak across the inner mitochondrial membrane, which dissipates the membrane potential and decreases ATP synthesis. Decreased activity of all five oxidative phosphorylation complexes was found in liver biopsies from human patients with NASH as compared to normal liver [52]. Similarly, decreased respiratory complex activities were also measured in the livers of ob/ob mice [53]. While these studies clearly point to a defect in mitochondrial bioenergetics, additional studies are required to identify and better understand the molecular mechanisms and targets responsible for mitochondrial dysfunction in obesity induced fatty liver disease. This is particularly important because studies have shown that state 3 respiration is not decreased in liver mitochondria isolated from ob/ob mice and that ob/ob mitochondria maintain a similar level of respiratory control compared to lean control mitochondria [51]. It is only when substrate is limited (i.e. decreased supply of electrons) to the respiratory chain that the membrane potential declines and ATP production is inhibited in mitochondria isolated from the severely steatotic liver of ob/ob mice. Thus, it has been proposed that up-regulation of UCPs negatively impacts cellular energy conservation only when the availability of oxidizable substrates becomes limited, e.g. under conditions of acute stress like ischemia, partial hepatectomy, and lipopolysaccharide exposure [51,54,55]. Based on this information, further studies are required to elucidate the impact of obesity on hepatic mitochondrial physiology in vivo particularly in the context of whether mitochondria are coupled or un-coupled as a consequence of steatosis.

Undeniably, one question that continues to come up in the field is whether mitochondrial dysfunction, i.e. the inability to maintain hepatic ATP levels, contributes to, or is simply a consequence of, fatty liver disease. While a difficult question to “prove” a few studies have shed light on this. As mentioned above, up-regulation of UCPs as an adaptive response to obesity results in hepatocyte ATP depletion when energy needs are acutely increased. Importantly, these studies demonstrated that hepatic necrosis occurred only when mitochondrial electron transport was inhibited [51,54]. Similarly, Diehl and colleagues demonstrated using P NMR spectroscopy that recovery from hepatic ATP depletion is severely impaired in NASH patients [56] suggesting a NASH dependent bioenergetic defect. However, what was more revealing from this small pilot study was the observation that body mass index was inversely correlated with ATP recovery even in the healthy lean control subjects. This finding indicates that defects in energy conservation (i.e. mitochondrial dysfunction) may occur before fatty liver disease is present [56]. Studies have also shown that alcohol mediated liver damage accompanies defects in mitochondrial function and the inability to maintain hepatic ATP concentrations [57,58]. While these findings are not unequivocal, they do support the hypothesis that defects in hepatic energy metabolism may indeed increase the susceptibility and risk for disease when the liver experiences other insults or “hits” like increased ROS/RNS.

Alcohol dependent disturbances in structure and function of the electron transport chain have also been proposed to be associated with increases in mitochondrial ROS production and oxidative injury in steatotic liver. Mitochondrial ROS production occurs as a by-product of electron transport; electrons “leak” from Complexes I and III to oxygen to form superoxide anion radicals (O2 ), which can be metabolized to hydrogen peroxide, interact with other free radicals like nitric oxide to form more toxic RNS, and initiate reactions leading to the generation of reactive lipid species (i.e. RLS). While mitochondria are known to be a significant source of oxidants in response to alcohol consumption [40,59], fewer studies have linked mitochondrial oxidant production to the development of NAFLD and NASH. More importantly, the molecular mechanisms responsible for oxidant production in obesity induced fatty liver are not known, especially because mitochondrial un-coupling has been shown in experimental models of NAFLD [51]. Studies using liver mitochondria from ob/ob mice have however demonstrated increased O2 compared to mitochondria from lean mice [60,61]. Possible mechanisms for enhanced mitochondrial ROS production include molecular defects within Complexes I and III, as well as increases in RLS, and TNFα mediated ceramide [22,59,62,63]. See review [64] for a detailed description of the mechanisms responsible for alcohol induced increases in mitochondrial ROS.

The mechanisms discussed in the previous paragraph are proposed to function by blocking or inhibiting the transfer of electrons along the respiratory chain, which results in increased reduction of the redox active centers of the respiratory complexes. These “overly” reduced complexes (I and III) then transfer electrons one at a time to oxygen to increase O2 generation, which can initiate a cycle of more lipid peroxidation, mitochondrial damage, and further ROS/RNS production. RLS (i.e. lipid peroxidation products), ROS, and RNS can also damage mtDNA. Studies have suggested that Complex II-linked respiration may lead to increased production of O2 within Complex I as a consequence of reverse electron transfer [65,66]. It is proposed that reverse electron transfer may contribute to increased mitochondrial production of O2 due to chronic alcohol-mediated defects within Complex I [43,46]. This would have the effect to compromise the functioning of the oxidative phosphorylation system as a result of the loss of mitochondrial gene products that make up segments of the respiratory chain; subsequently leading to more ROS production. This concept is supported by observations of significant mtDNA mutations and abnormalities in NASH patients [67,68] and in an animal model of fatty liver disease [60]. Whether these mtDNA defects translate into decreased activity and content of the respiratory complexes in obesity induced fatty liver disease is not known. Thus, future studies should be directed at determining whether alterations to the mitochondrial genome and proteome accompany oxidant damage and pathology of NAFLD.

In addition to the respiratory chain being a significant source of mitochondrial ROS production, work by Fromenty and colleagues demonstrated an alcohol dependent increase in both microsomal and mitochondrial cytochrome P450 2E1 (CYP450 2E1) in cultured hepatocytes and liver of wild-type mice [69] suggesting an additional source of oxidants in alcohol exposed mitochondria. Indeed, CYP450 2E1 is known to be a significant source of ROS as it generates high amounts of hydrogen peroxide in the absence or presence of oxidizable co-substrates due to the “un-coupled” nature of this P450 isoform [70]. In contrast, short-term alcohol exposure did not enhance mitochondrial CYP450 2E1 levels in liver of ob/ob mice. It is important to note that there was no difference in the levels and activity of CYP450 2E1 in liver from lean and ethanol-naïve ob/ob mice [69]. Mitochondrial ROS generation has also been ascribed to several matrix enzymes including α-ketoglutarate dehydrogenase [71,72] and α-glycerophosphate dehydrogenase [73]. Whether these enzymes contribute to increased ROS production during the conditions of alcohol and obesity induced fatty liver disease is not known, however it is predicted that higher rates of ROS in mitochondria during these pathologies will negatively affect mitochondrial and cellular function by oxidative modification and alteration in redox-sensitive signaling pathways.

Depletion of mitochondrial glutathione (GSH) has also been implicated as a contributor to the development of alcoholic liver disease as GSH functions in pathways responsible for ROS detoxification [74]. Studies by Fernandez-Checa and colleagues demonstrated that chronic alcohol consumption depletes mitochondrial GSH [75] and treatments that prevent the loss in inner membrane fluidity maintain GSH uptake and preserve mitochondrial GSH levels in liver of alcohol-fed animals [76,77]. Importantly, it is proposed that loss of mitochondrial GSH sensitizes hepatocytes from alcohol-fed animals to TNFα induced cell death [78,79], specifically when mitochondria are “loaded” with free cholesterol [80]. In contrast, several studies have shown that mitochondrial GSH depletion may not be a consistent outcome following chronic alcohol consumption as studies have shown increased mitochondrial GSH in response to alcohol consumption [8184]. Similarly, Cederbaum and colleagues have shown increased GSH in HepG2 cells over-expressing CYP450 2E1 due to upregulation of glutamate cysteine ligase [85]. Based on this, increases in mitochondrial GSH in response to mild-moderate alcohol consumption may represent an adaptive response to counteract oxidative stress induced by chronic alcohol exposure. Indeed, adaptive increases in GSH have been demonstrated in multiple models of mild oxidative stress [86,87]. Conflicting effects of obesity and NAFLD on mitochondrial GSH levels have also been reported. For example, decreased [53] and increased [61] levels of mitochondrial GSH have been reported in liver of ob/ob mice. In addition, two glutathione-replenishing agents, S-adenosylmethionine and 2(RS)-n-propylthiazolidine-4(R)-carboxylic acid (i.e. an L-cysteine pro-drug), were shown to protect against steatohepatitis induced by the methionine-choline deficient diet; however, these agents did not prevent alterations in GSH and GSSG levels in response to steatohepatitis [88]. Nonetheless, increased GSSG concentrations were found in the blood of NASH patients suggesting defects in glutathione metabolism [89]. Taken together, these findings demonstrate that while a potential player in the pathogenesis of fatty liver disease, additional studies are necessary to delineate the precise role of mitochondrial GSH depletion in the pathogenesis of alcohol and non-alcohol mediated mitochondrial dysfunction and liver injury.

Interplay Between Nitric Oxide and Mitochondria in Fatty Liver Disease

Nitric oxide (NO) and other RNS are increased in response to chronic alcohol consumption through induction of inducible nitric oxide synthase (iNOS) [82,90]. In normal healthy liver iNOS levels are very low or undetectable [91], whereas during inflammation and other disease states iNOS levels are increased in liver due to infiltrating inflammatory cells and via induction in Kupffer cells (resident liver macrophage), hepatocytes, and biliary epithelial cells [9294]. Studies also report increased iNOS expression in liver of ob/ob mice [53], CCl4-induced steatotic liver following normothermic ischemia [95], and in the obese fa/fa rat exposed to binge alcohol [5]. This is important for hepatotoxicity because NO and peroxynitrite (ONOO ) are known to mediate mitochondrial dysfunction [96,97] and increases in iNOS expression in fatty liver are correlated with nitration of mitochondrial proteins [53,98]. Moreover, NO regulates mitochondrial respiration through reversible binding at the redox-active heme site in cytochrome c oxidase [99101] and affects mitochondrial biogenesis through interactions with soluble guanylate cyclase [102]. Studies have shown that mitochondria from chronic alcohol-fed animals are more sensitive to the inhibitory effect of NO on respiration [90,103]. While it is known that chronic alcohol consumption results in cytochrome c oxidase defects [82,90,104], it is likely that this alteration in NO responsiveness may be due to alcohol-dependent changes in interactions of NO with sites other than Complex IV. It is postulated that this disruption in NO signaling contributes to alcohol hepatotoxicity through inhibition of ATP synthesis, increased ROS, and inability to adapt to hypoxic stress [105]. This concept is supported because increased sensitivity to NO is absent in iNOS mice exposed to chronic alcohol [90]. Similarly, alcohol-associated inflammation and steatosis is abrogated in iNOS mice [90,106]. Taken together, these studies demonstrate that iNOS induction is linked to alterations in NO-dependent control of mitochondrial respiration, which potentially contributes to the development of alcoholic steatohepatitis. A scheme summarizing the effects of alcohol and obesity to disrupt mitochondrial function is provided in Figure 2.

An external file that holds a picture, illustration, etc.
Object name is nihms46170f2.jpg
Alcohol and obesity induced mitochondrial dysfunction and oxidative stress

Alcohol and obesity disrupt the functioning of the oxidative phosphorylation (OxPhos) system which results in the generation of superoxide (O2 ) within Complexes I and III. The O2 is dismutated to hydrogen peroxide (H2O2) by manganese superoxide dismutase (MnSOD) that can then diffuse from the mitochondrion into the cytoplasm to affect cellular redox signaling pathways. Alcohol and obesity also cause an increase in iNOS expression that leads to diffusion of nitric oxide (NO) into mitochondria where it inhibits cytochrome c oxidase; reacts with O2 to form highly reactive peroxynitrite (ONOO ). Hydrogen peroxide, O2 , and ONOO can initiate reactions that lead to increased generation of reactive lipid species (RLS), induce post-translational modification of proteins, and damage mitochondrial DNA (mtDNA).

It is also important to highlight the beneficial effects of NO in the context of liver diseases. Studies implicate that decreased production of NO from endothelial NOS (eNOS) contributes to liver pathology via dysregulation of blood flow and oxygen delivery [107]. In support of this, NO-donor administration and eNOS overexpression have been shown to prevent liver injury in animal models of hepatotoxicity [108,109]. For example, the oral administration of S-nitroso-N-acetylcysteine prevented the onset of NAFLD in response to a methionine-choline deficient diet [110,111]; however, it is unclear whether the benefit was due to NO or N-acetylcysteine exposure. Recently, inhaled NO administration was shown to accelerate the restoration of liver function in transplantation patients presumably through increased circulating levels of nitrite [112], a newly recognized vascular endocrine transporter of NO [113] that protects against heart and liver ischemia-reperfusion injury [114]. In models of chronic alcohol consumption eNOS activity is decreased in liver through increased expression of the inhibitory protein caveolin-1 and/or decreased eNOS phosphorylation [115,116]. Similarly, Nanji and colleagues have demonstrated L-arginine supplementation prevents and reverses chronic alcohol mediated liver injury [117,118]. Based on this, they propose that decreased NO from non-parenchymal cells, i.e. endothelial cells and eNOS, and increased NO from hepatocyte iNOS work in concert to exacerbate chronic alcohol mediated liver injury [117,118]. While these data support the hypothesis that the source, site, and concentration of NO produced are critical for determining the functional consequence of NO, i.e. good or bad NO, the complex interplay between NO and mitochondria remains to be defined in the context of fatty liver disease from alcohol and obesity. Moreover, the critical importance of understanding the regulation of the mitochondrial respiratory chain by NO in hepatic physiology and pathophysiology is supported by several studies demonstrating that localized NO production can occur within the organelle through a specific mitochondrial NOS isoform or via metabolism of nitrite to NO [119121].

Genetic Factors Determining the Pathobiology of Fatty Liver Diseases – “Oxidative Stress Responsive Genes”

Emerging evidence indicates that genetic factors contribute to increase the risk and severity of alcohol and non-alcohol induced liver disease [3,122]. Indeed, several genes have been identified as influencing the impact of oxidative stress in the pathogenesis of fatty liver disease. Studies have reported genetic polymorphisms in the genes encoding enzymes responsible for alcohol metabolism; alcohol dehydrogenase, CYP450 2E1, and the mitochondrial low Km aldehyde dehydrogenase [123]. Unfortunately, only weak associations between these polymorphisms and increased alcohol hepatotoxicity have been shown [124].

In contrast to these findings, a polymorphism responsible for a valine to alanine substitution in the mitochondrial targeting sequence for manganese superoxide dismutase (SOD2) was found to be a major risk factor for severe alcoholic liver disease in two separate studies [125,126]. The alanine-SOD2 variant, whose pre-sequence is an α-helix, is readily imported and achieves high activity in mitochondria, whereas the valine-SOD2 variant, whose pre-sequence is a partial β-sheet, is poorly imported resulting in low SOD2 activity [127,128]. It is proposed that the presence of the high-activity, alanine-SOD2 variant will increase hydrogen peroxide production in mitochondria, which might have the effect to disrupt cellular redox signaling pathways and initiate oxidative damage [129]. This is supported by studies demonstrating an interaction of the high-activity, alanine-SOD2 variant with a low activity glutathione peroxidase-1 variant (leucine-GPx1) to increase hepatic iron accumulation and risk for hepatocellular carcinoma in alcoholic cirrhotics [129]. Indeed, cirrhotics possessing two copies of the low-activity, valine-SOD2 variant and high-activity, proline-GPx1 variant are protected from cancer. Furthermore, chronic alcohol consumption is known to increase SOD2 activity [130,131] and decrease mitochondria GPx1 activity [81]. Thus, an imbalance resulting in increased hydrogen peroxide formation and decreased detoxification may be a critical event for the initiation and progression of alcohol induced liver injury. With this said, however, studies performed in the UK with a larger cohort of alcoholic patients showed no association among the valine-alanine SOD2 polymorphism, alcohol-dependent markers of oxidative stress, and liver fibrosis [132]. Preliminary studies indicate that the SOD2 polymorphism may be correlated to fibrosis in NAFLD patients [133].

Other studies have reported associations between genetic polymorphisms in endotoxin receptors, cytokines, and fibrosis genes with the pathogenesis of fatty liver diseases. The role of endotoxin mediated cytokine release in the etiology of alcohol hepatotoxicity was established by Thurman and colleagues in the 1990’s through an elegant series of experimental animal studies [134,135]. Likewise, some genetic studies have shown an association between alcoholic liver disease [136] and NASH [137] with a polymorphism present in the CD14 promoter that results in increased CD14 levels [138]. The CD14 receptor binds endotoxin and enhances signaling through the toll-like receptor-4 (TLR4). However, other investigations have reported no association between the CD14 and TLR4 polymorphisms and established alcoholic liver disease [139,140]. Similar conflicting results have been reported for the cytokines TNF-α and interleukin-10 [140]. Moreover, while fibrogenesis/fibrinolysis genes are predicted to be strong candidates to influence liver fibrosis, only one report has shown a link between transforming growth factor-β 1 (TGF-β 1) and angiotensinogen polymorphisms and obesity induced liver fibrosis [141]. Lastly, it should be mentioned that an association between naturally occurring variations in the mitochondrial genome and conditions of the cardiometabolic syndrome has been proposed [142,143]. Recently, Pravenec et al. [142] using conplastic rat strains with the same nuclear genome but dissimilar mitochondrial genomes encoding oxidative phosphorylation proteins of differing amino acid composition showed differences in glucose and glycogen metabolism; two risk factors for type 2 diabetes. Whether mtDNA variants are linked to risk factors associated with steatosis, hepatitis, and fibrosis/cirrhosis is a stimulating concept and one that clearly requires further study. Taken together, these findings indicate that genetic factors may contribute to the pathobiology of alcohol and non-alcohol induced steatohepatitis and/or fibrosis, however additional investigations with larger populations are needed to solidify these linkages.

Are Antioxidants Feasible Treatments for Fatty Liver Disease?

While abstinence and weight loss are critically important for ensuring successful outcomes from alcohol and obesity induced liver diseases, respectively, numerous therapeutic agents have been investigated for treatment of these liver diseases (Figure 3). Unfortunately, no proven drug therapies have emerged as being highly effective. This section will provide a brief overview of some of the newer agents that show promise in treatment of fatty liver diseases.

An external file that holds a picture, illustration, etc.
Object name is nihms46170f3.jpg
Proposed therapeutic agents for the treatment of fatty liver diseases

Corticosteriods, pentoxifylline, propylthiouracil, and anti-TNFα have all been used to treat patients with alcoholic hepatitis with some success; however, issues regarding timing of administration in relation to alcohol exposure and efficacy need to be addressed through more randomized clinical trials [144]. Similarly, insulin sensitizing drugs such as metformin and the thiazolidinediones show promise in treating fatty liver disease. Metformin was shown to prevent alcohol induced liver injury in mice [145]. Interestingly, the protective effect of metformin in this study did not correlate to AMP kinase activation, but via prevention of plasminogen activator inhibitor (PAI)-I upregulation by alcohol [145]. Rosiglitazone and pioglitazone, thiazolidinedione peroxisomal proliferator-activated receptor (PPAR)-γ agonists, decrease insulin resistance and have been shown to decrease TNF-α levels and inflammation in rodent models of NASH [146150]. Similarly, the PPAR-γ agonist pioglitazone prevents alcohol induced liver injury through interactions with the c-Met signaling pathway resulting in decreased triglyceride synthesis [151]. It should also be mentioned that treatment with the PPAR-α agonist WY14,643 prevented alcohol induced steatosis via up-regulation of β-oxidation activity [152]. However caution should be exercised when using these medications because recent reports have demonstrated mitochondrial abnormalities associated with rosiglitazone therapy in both experimental [153] and human patients [154] with NAFLD.

Both traditional and newly recognized antioxidants have been proposed to slow the progression, decrease the severity, and/or reverse pathology from fatty liver diseases presumably through their ability to decrease oxidative stress in tissues. And, like the therapeutic agents described above, results using antioxidants have been varied with some antioxidants showing positive effects and others not. In both adults and children with NASH, treatment with α-tocopherol (vitamin E) has been shown to improve serum liver enzymes [155157]. Similarly, NASH patients treated with vitamin E (400 IU/day) and pioglitazone for 6 months showed significant improvement in liver histology over vitamin E treatment alone [158]. While results with α-tocopherol are promising, recent reports have suggested that γ-tocopherol may be a more effective therapeutic agent in treating diseases involving oxidant stress and inflammation due to its potent anti-inflammatory activity [159,160]. Furthermore, Murphy and colleagues have developed novel strategies to “target” traditional antioxidants, like vitamin E, ubiquinone, and lipoic acid to the mitochondrion by attaching them to a lipophilic triphenylphosphonium moiety [161163]. This modification allows the antioxidant to accumulate several-hundred fold in the mitochondrion in response to the mitochondrial membrane potential where it can exert maximum benefit. Indeed, mitochondria-targeted ubiquinone (a.k.a. MitoQ) was shown to decrease cardiac ischemia-reperfusion injury [164]. In contrast, mitochondria-targeted vitamin E (a.k.a. MitoE) caused neurotoxicity in a hypoxic-ischemia model of striatal injury [165]. Thus, additional studies are required to demonstrate the safety and efficacy of these novel mitochondrially targeted antioxidants in diseases where mitochondrial damage and oxidant stress are proposed to play a role in pathology.

Numerous other antioxidants show efficacy in treating fatty liver diseases and include N-acetylcysteine [166], zinc [167,168], silymarin [169], and ursodeoxycholic acid [170], just to name a few. Please refer to the following excellent review articles for additional information on antioxidant therapy for treatment of fatty liver diseases from multiple causes [144,171174].

Defects in methionine metabolism occur in response to both alcohol and non-alcohol induced liver disease and are postulated to contribute to the disease process. One key metabolite of the methionine metabolic pathway is S-adenosylmethionine (SAM), which is the main methyl donor for biological reactions and serves as a precursor in glutathione and polyamine synthesis [175].

In mammalian cells SAM is synthesized by the enzyme methionine adenosylmethytransferase (MAT). Alcohol and other hepatotoxicants decrease hepatic levels of SAM due to decreased activity of the hepatocyte specific form of MAT [176] and methionine synthase [177]. Studies have shown that NO inactivates rat liver MAT in vivo by S-nitrosation of cysteine 121, which leads to decreased synthesis and depletion of hepatic SAM [178,179]. SAM depletion correlates with DNA hypomethylation and strand breaks [176] and several parameters of oxidative stress including decreased GSH and increased lipid peroxidation [180]. Accordingly, exogenous administration of SAM in animal models of alcohol toxicity has been shown to be protective [82,181,182]. SAM supplementation was also shown to significantly improve the outcome of alcoholic cirrhotic patients [183]. Emerging evidence suggests that the mitochondrion may be an important target for the benefits afforded by SAM in the treatment of fatty liver disease.

Studies by Mato and colleagues showed that MAT1A mice, which are deficient in SAM, spontaneously develop steatohepatitis and hepatocellular carcinoma demonstrating that SAM depletion primes the liver for progression to more severe injury [184,185]. Mitochondria isolated from liver of MAT1A− /− mice also have decreased levels of the mitochondrial encoded subunits for cytochrome c oxidase and reduced membrane potential [186]. This finding is consistent with the concept that SAM plays an essential role in the maintenance and proper function of mitochondria and the oxidative phosphorylation system. Early studies showed that co-administration of SAM during alcohol feeding maintained mitochondrial GSH transport and GSH levels, which preserved ATP levels and membrane potential [76,77]. Recently, we have extended these earlier studies and shown that SAM supplementation prevents alcohol-dependent decreases in mitochondrial respiration, increases in superoxide production, mtDNA damage, and iNOS induction [82]. Similarly, SAM prevented alcohol-associated losses in both nuclear and mitochondrial encoded subunits of cytochrome c oxidase, which preserved oxidase activity [82]. These results are supported by observations that depletion of mitochondrial SAM levels sensitizes HepG2 cells to TNF-α cytotoxicity [187].

In addition to SAM, the methionine pathway metabolite betaine has been shown to prevent hepatotoxicity in animal models of alcohol [188,189] and diet [190] induced steatosis, as well as improve serum liver enzymes and liver histology in NASH patients [191]. Betaine, a natural antioxidant, functions as an osmolyte and a methyl donor for the remethylation of homocysteine, “the bad thiol” [192], back to methionine [193]. Hyperhomocysteinemia is associated with steatosis presumably through induction of endoplasmic reticulum stress and activation of SREBPs leading to increased hepatic synthesis and uptake of triglycerides and cholesterol [194]. Indeed, hyperhomocysteinemia may be a suitable predictor of NASH [195]. Preliminary data demonstrates that betaine supplementation prevented chronic alcohol-induced steatosis and losses in the respiratory chain complexes, particularly subunits of Complex I and IV [196]. Taken together, these data reveal a novel molecular mechanism through which SAM and betaine may work to prevent one key component of alcohol and obesity-induced liver disease; mitochondria dysfunction.

Summary

Strong evidence indicates that the pathogenesis of chronic alcohol and obesity/type 2 diabetes induced fatty liver disease is linked to mitochondrial dysfunction. The initial formation of an inflammatory response and increased production of ROS/RNS associated with the metabolic alterations induced by these conditions leads to modifications of the mitochondrial genome and proteome. These alterations result in loss of mitochondrial respiration, the inability to maintain sufficient ATP concentrations, and a further enhancement of ROS/RNS production. Indeed, the role of oxidative stress in these disease processes has been demonstrated in numerous experimental models. Thus, the goal of current and future investigations will be to provide a thorough characterization of the molecular mechanisms involved in the pathogenesis of fatty liver diseases with particular emphasis on defining the role of mitochondrial oxidative stress in the disease process. A more complete understanding of the ROS/RNS mediated effects on the spectrum of alcohol and obesity induced liver disease will facilitate the development of new mitochondrially targeted molecular medicines for treatment.

“Two-Hit” or “Multi-Hit” Hypothesis for Alcoholic and Non-Alcoholic Fatty Liver Disease

Mechanisms of alcoholic liver disease have been extensively studied; however, the events that cause progression to hepatitis and fibrosis/cirrhosis are still undefined. The same can also be said for the pathogenesis of NAFLD. Currently, the leading hypothesis for both alcohol and non-alcohol induced liver disease is the “two-hit” or “multi-hit” hypothesis with the “first-hit” being the accumulation of triglyceride in hepatocytes (steatosis) followed by “second-hits” which trigger the progression to more serious liver pathologies like steatohepatitis and fibrosis/cirrhosis (Figure 1). Examples of “second-hits” could include environmental stressors like dietary components, cigarette smoke, or pollutants and/or metabolic stressors such as hyperglycemia, hypertriglyceridemia, and hypercholesterolemia, which are components linked to the cardiometabolic syndrome. Interestingly, what these factors all have in common is the ability to induce oxidative stress arising largely in part via mitochondrial defects. It has been proposed that the elusive “second-hit” arises from the enhanced production of reactive oxygen and/or nitrogen species (ROS/RNS) in liver [13]. Once steatosis is present, the liver becomes more susceptible to the “second-hit”; oxidative/nitrosative/nitrative stress, which is thought to be one of several stimuli for the progression from simple fatty liver to ASH or NASH. The molecular mechanism underlying how steatosis predisposes liver to transition from simple fatty liver to steatohepatitis is not clear; however, several studies point to the possibility that when hepatocytes accumulate fat the fatty acids themselves are toxic and initiate a pathological response called “lipotoxicity” [14]. Studies show that free fatty acids are toxic to hepatocytes via deregulation of lysosomal metabolism [15,16] and induction of endoplasmic reticulum stress [17,18] resulting in apoptosis. Similarly, palmitate induced lipid accumulation in hepatocytes stimulated production of the neutrophil chemoattractant interleukin-8, which could have the effect of initiating hepatic inflammation and injury [19]. The oxidation and release of reactive lipid species (i.e. lipid peroxidation products) present in fat deposits may also contribute to liver injury through the abilities of these molecules to adduct to cellular macromolecules thereby disrupting function. For example, reactive lipid species have been shown to bind to mitochondrial proteins, induce mitochondrial ROS production, and promote cytochrome c release [20,21]. Therefore, increased reactive lipid species may be a critical factor predisposing liver to more severe injury when exposed to other insults or “hits”. Finally, as both a site for fat metabolism and the main source of ROS/RNS in hepatocytes, the mitochondrion is postulated to play a central role in the development of alcoholic and non-alcoholic liver disease and this will be discussed in more detail later in the article.

An external file that holds a picture, illustration, etc.
Object name is nihms46170f1.jpg
Pathophysiology of alcoholic steatohepatitis (ASH) and non-alcoholic steatohepatitis (NASH)

To understand the molecular mechanisms by which chronic alcohol consumption and the cardiometabolic syndrome cause liver disease, it is important to define the pathways leading to fat accumulation in liver. Excess fat accumulation in liver can result from one or a combination of the following metabolic alterations:

  1. decreased β-oxidation of fatty acids;

  2. increased fatty acid synthesis due to up-regulation of lipogenic pathways;

  3. increased delivery of fatty acids from adipose and other organs due to lipolysis; and

  4. inhibition of VLDL-triglyceride export.

In general, it is the metabolic consequence of ethanol oxidation; increase in the NADH/NAD ratio, which disrupts fat metabolism leading to triglyceride accumulation in hepatocytes. Increased NADH leads to increased esterification of fatty acids into triglyceride and increased fatty acid synthesis, while inhibiting fatty acid β-oxidation and tricarboxylic (TCA) cycle activity. In the case of NAFLD, insulin resistance is thought to be responsible for the “first-hit”, steatosis, through the mobilization of peripheral fat to the liver through the development of hyperinsulinemia. Peripheral lipolysis and hyperinsulinemia lead to fatty liver through increased hepatic uptake of fatty acids and accumulation of fat via increased fatty acid synthesis, which again can be accompanied by decreased β-oxidation activity [22]. Recent data also suggests that peripheral lipolysis may contribute to alcohol-induced fatty liver in response to an alcohol-dependent suppression of insulin’s anti-lipolytic action in adipocytes [23]. While emerging data indicates that alcohol dependent insulin resistance may contribute to the accumulation of fat in hepatocytes, the evidence in support of this is less clear; primarily due to observations that alcohol has “dose-dependent” effects on insulin signaling [24]. Studies by Badger and colleagues have demonstrated that while high dose chronic alcohol consumption impairs hepatic insulin signaling, low dose ethanol exposure activates insulin signaling in hepatocytes [24,25]. Importantly, for the first time, these findings provide biochemical support for epidemiologic evidence suggesting that while low dose alcohol consumption may be cardio-protective and decrease the risk of developing type 2 diabetes; heavy, prolonged alcohol consumption increases the risk for type 2 diabetes.

It should also be mentioned that steatosis induced by chronic alcohol consumption and/or type 2 diabetes can be directly linked to two critical signaling pathways that increase lipogenesis in the liver; AMP kinase inhibition and SREBP-1 (sterol regulatory element binding protein-1) activation. AMP kinase, a protein kinase that inhibits lipogenesis and activates fatty acid oxidation, is inhibited by chronic alcohol feeding, which results in increased SREBP-1 activation [26,27]. As a consequence of the alcohol-dependent activation of SREBP-1, a number of lipogenic genes are up-regulated in liver contributing to enhanced lipid synthesis. Similarly, the ethanol-dependent inhibition of AMP kinase leads to activation of acetyl CoA carboxylase; enhancing malonyl CoA levels, which inhibits fatty acid uptake and β-oxidation in mitochondria. Thus, alcohol-associated inhibition of AMP kinase contributes to fat accumulation via stimulation of lipogenesis and inhibition of fat oxidation. Additionally, it has been demonstrated that activation of AMP kinase by the adipocytokine adiponectin and other agents improves both alcohol and obesity-induced fatty liver disease in rodent models [2830] demonstrating the key role of AMP kinase in the development of steatosis. Clearly, the question that still remains to be answered is, what is the mechanism through which ethanol inhibits AMP kinase activity? AMP kinase is activated by phosphorylation thus possible mechanisms of inactivation could include allosteric modification by AMP and ATP, activation of protein phosphatases, and modulation by other signaling pathways [31,32]. More recently, studies have suggested that the finely tuned mitochondrial ROS/RNS balance may also be a critical controlling factor in the activation of AMP kinase [3335]. Therefore, it is possible that chronic alcohol and obesity mediated effects on mitochondrial functioning and redox signaling inhibit AMP kinase activity thereby contributing to the development and progression of fatty liver disease. Future studies are warranted to address this very interesting and provocative new question.

Before considering the role of mitochondrial dysfunction in the pathogenesis of alcohol and non-alcohol mediated fatty liver disease, it is important to highlight emerging evidence, which indicates that steatosis may not be an unvarying feature of ASH and NASH. Recent work by the Nagy [36] and Lindros [37] laboratories have revealed that activation of the complement pathway is necessary for the development of chronic alcohol induced steatosis. Indeed, both laboratories have independently shown that, unlike wild-type mice, complement component C3 mice do not develop fatty liver when fed an ethanol-containing diet for 6 weeks [36,37]. More interesting, however, is the observation by Pritchard et al. showing that even though the C3 mice were protective against ethanol-mediated steatosis, mice still had elevated serum ALT and hepatic TNFα levels following alcohol exposure indicating hepatic inflammatory injury in the absence of steatosis [36]. In support of this, Diehl and colleagues demonstrated that inhibition of hepatocyte triglyceride synthesis and steatosis increased liver injury and fibrosis in obese mice with NASH [38]. Moreover, oleate- or palmitate-loaded HepG2 spheroids (hepatocyte-derived cells) were less susceptible to cytokine or peroxide induced cell death [39]. Taken together, these results are intriguing because they indicate that steatosis may not be the “first-hit” or a pre-requisite in the pathogenesis of ASH or NASH. In fact, steatosis may even be protective against hepatotoxicity [38]. Clearly, additional research is required to determine the role of steatosis in inflammatory and/or fibrotic liver diseases.

Mitochondria Dysfunction in Fatty Liver Diseases – Bioenergetic and Oxidative Stress

While the detrimental effects of chronic alcohol consumption on liver mitochondria have been known for many years [40], it has only been recently recognized that mitochondrial dysfunction may play a critical role in the development of NAFLD and progression to NASH [22]. Even though the molecular defects responsible for alcohol and non-alcohol dependent mitochondrial dysfunction remain to be defined, it is clear that under conditions of fatty liver disease one key functional change to mitochondria is the inability to maintain sufficient levels of ATP.

Interestingly though, this event may occur by different types of biochemical and molecular changes to mitochondria. In general, respiration in the presence of ADP (state 3 respiration) with NADH-linked substrates, fatty acids, or succinate is decreased as a consequence of chronic alcohol consumption, whereas respiration in the absence of ADP (state 4 respiration) is largely unaffected [41]. Moreover, this decrease in ADP-stimulated respiration is linked to decreased electron transport in all segments of the respiratory chain as chronic alcohol consumption decreases the activities of all the respiratory complexes, except complex II [41,42]. Several laboratories have presented strong evidence that inhibition of mitochondrial protein synthesis [43] linked to mtDNA damage [4446] and ribosomal defects [47,48] contribute, in part, to decreased functioning of the oxidative phosphorylation system following chronic alcohol consumption. These alterations translate into profound modifications to the mitochondrial proteome that encompass not only losses in the 13 mitochondrial encoded polypeptides, but also decreases in numerous nuclear encoded proteins that make up the oxidative phosphorylation complexes [46]. Proteomic analyses have also shown alcohol-dependent changes in mitochondrial matrix enzymes both at the level of abundance and post-translational modifications [46,49,50].

Like chronic alcohol induced liver injury, emerging evidence strongly implicates mitochondrial dysfunction contributing to obesity induced fatty liver disease (i.e. NAFLD). Early studies by Diehl and colleagues reported that obesity induced steatosis was associated with increased expression of uncoupling protein-2 (UCP-2) in liver, which promoted ATP depletion [51]. This results from increased H leak across the inner mitochondrial membrane, which dissipates the membrane potential and decreases ATP synthesis. Decreased activity of all five oxidative phosphorylation complexes was found in liver biopsies from human patients with NASH as compared to normal liver [52]. Similarly, decreased respiratory complex activities were also measured in the livers of ob/ob mice [53]. While these studies clearly point to a defect in mitochondrial bioenergetics, additional studies are required to identify and better understand the molecular mechanisms and targets responsible for mitochondrial dysfunction in obesity induced fatty liver disease. This is particularly important because studies have shown that state 3 respiration is not decreased in liver mitochondria isolated from ob/ob mice and that ob/ob mitochondria maintain a similar level of respiratory control compared to lean control mitochondria [51]. It is only when substrate is limited (i.e. decreased supply of electrons) to the respiratory chain that the membrane potential declines and ATP production is inhibited in mitochondria isolated from the severely steatotic liver of ob/ob mice. Thus, it has been proposed that up-regulation of UCPs negatively impacts cellular energy conservation only when the availability of oxidizable substrates becomes limited, e.g. under conditions of acute stress like ischemia, partial hepatectomy, and lipopolysaccharide exposure [51,54,55]. Based on this information, further studies are required to elucidate the impact of obesity on hepatic mitochondrial physiology in vivo particularly in the context of whether mitochondria are coupled or un-coupled as a consequence of steatosis.

Undeniably, one question that continues to come up in the field is whether mitochondrial dysfunction, i.e. the inability to maintain hepatic ATP levels, contributes to, or is simply a consequence of, fatty liver disease. While a difficult question to “prove” a few studies have shed light on this. As mentioned above, up-regulation of UCPs as an adaptive response to obesity results in hepatocyte ATP depletion when energy needs are acutely increased. Importantly, these studies demonstrated that hepatic necrosis occurred only when mitochondrial electron transport was inhibited [51,54]. Similarly, Diehl and colleagues demonstrated using P NMR spectroscopy that recovery from hepatic ATP depletion is severely impaired in NASH patients [56] suggesting a NASH dependent bioenergetic defect. However, what was more revealing from this small pilot study was the observation that body mass index was inversely correlated with ATP recovery even in the healthy lean control subjects. This finding indicates that defects in energy conservation (i.e. mitochondrial dysfunction) may occur before fatty liver disease is present [56]. Studies have also shown that alcohol mediated liver damage accompanies defects in mitochondrial function and the inability to maintain hepatic ATP concentrations [57,58]. While these findings are not unequivocal, they do support the hypothesis that defects in hepatic energy metabolism may indeed increase the susceptibility and risk for disease when the liver experiences other insults or “hits” like increased ROS/RNS.

Alcohol dependent disturbances in structure and function of the electron transport chain have also been proposed to be associated with increases in mitochondrial ROS production and oxidative injury in steatotic liver. Mitochondrial ROS production occurs as a by-product of electron transport; electrons “leak” from Complexes I and III to oxygen to form superoxide anion radicals (O2 ), which can be metabolized to hydrogen peroxide, interact with other free radicals like nitric oxide to form more toxic RNS, and initiate reactions leading to the generation of reactive lipid species (i.e. RLS). While mitochondria are known to be a significant source of oxidants in response to alcohol consumption [40,59], fewer studies have linked mitochondrial oxidant production to the development of NAFLD and NASH. More importantly, the molecular mechanisms responsible for oxidant production in obesity induced fatty liver are not known, especially because mitochondrial un-coupling has been shown in experimental models of NAFLD [51]. Studies using liver mitochondria from ob/ob mice have however demonstrated increased O2 compared to mitochondria from lean mice [60,61]. Possible mechanisms for enhanced mitochondrial ROS production include molecular defects within Complexes I and III, as well as increases in RLS, and TNFα mediated ceramide [22,59,62,63]. See review [64] for a detailed description of the mechanisms responsible for alcohol induced increases in mitochondrial ROS.

The mechanisms discussed in the previous paragraph are proposed to function by blocking or inhibiting the transfer of electrons along the respiratory chain, which results in increased reduction of the redox active centers of the respiratory complexes. These “overly” reduced complexes (I and III) then transfer electrons one at a time to oxygen to increase O2 generation, which can initiate a cycle of more lipid peroxidation, mitochondrial damage, and further ROS/RNS production. RLS (i.e. lipid peroxidation products), ROS, and RNS can also damage mtDNA. Studies have suggested that Complex II-linked respiration may lead to increased production of O2 within Complex I as a consequence of reverse electron transfer [65,66]. It is proposed that reverse electron transfer may contribute to increased mitochondrial production of O2 due to chronic alcohol-mediated defects within Complex I [43,46]. This would have the effect to compromise the functioning of the oxidative phosphorylation system as a result of the loss of mitochondrial gene products that make up segments of the respiratory chain; subsequently leading to more ROS production. This concept is supported by observations of significant mtDNA mutations and abnormalities in NASH patients [67,68] and in an animal model of fatty liver disease [60]. Whether these mtDNA defects translate into decreased activity and content of the respiratory complexes in obesity induced fatty liver disease is not known. Thus, future studies should be directed at determining whether alterations to the mitochondrial genome and proteome accompany oxidant damage and pathology of NAFLD.

In addition to the respiratory chain being a significant source of mitochondrial ROS production, work by Fromenty and colleagues demonstrated an alcohol dependent increase in both microsomal and mitochondrial cytochrome P450 2E1 (CYP450 2E1) in cultured hepatocytes and liver of wild-type mice [69] suggesting an additional source of oxidants in alcohol exposed mitochondria. Indeed, CYP450 2E1 is known to be a significant source of ROS as it generates high amounts of hydrogen peroxide in the absence or presence of oxidizable co-substrates due to the “un-coupled” nature of this P450 isoform [70]. In contrast, short-term alcohol exposure did not enhance mitochondrial CYP450 2E1 levels in liver of ob/ob mice. It is important to note that there was no difference in the levels and activity of CYP450 2E1 in liver from lean and ethanol-naïve ob/ob mice [69]. Mitochondrial ROS generation has also been ascribed to several matrix enzymes including α-ketoglutarate dehydrogenase [71,72] and α-glycerophosphate dehydrogenase [73]. Whether these enzymes contribute to increased ROS production during the conditions of alcohol and obesity induced fatty liver disease is not known, however it is predicted that higher rates of ROS in mitochondria during these pathologies will negatively affect mitochondrial and cellular function by oxidative modification and alteration in redox-sensitive signaling pathways.

Depletion of mitochondrial glutathione (GSH) has also been implicated as a contributor to the development of alcoholic liver disease as GSH functions in pathways responsible for ROS detoxification [74]. Studies by Fernandez-Checa and colleagues demonstrated that chronic alcohol consumption depletes mitochondrial GSH [75] and treatments that prevent the loss in inner membrane fluidity maintain GSH uptake and preserve mitochondrial GSH levels in liver of alcohol-fed animals [76,77]. Importantly, it is proposed that loss of mitochondrial GSH sensitizes hepatocytes from alcohol-fed animals to TNFα induced cell death [78,79], specifically when mitochondria are “loaded” with free cholesterol [80]. In contrast, several studies have shown that mitochondrial GSH depletion may not be a consistent outcome following chronic alcohol consumption as studies have shown increased mitochondrial GSH in response to alcohol consumption [8184]. Similarly, Cederbaum and colleagues have shown increased GSH in HepG2 cells over-expressing CYP450 2E1 due to upregulation of glutamate cysteine ligase [85]. Based on this, increases in mitochondrial GSH in response to mild-moderate alcohol consumption may represent an adaptive response to counteract oxidative stress induced by chronic alcohol exposure. Indeed, adaptive increases in GSH have been demonstrated in multiple models of mild oxidative stress [86,87]. Conflicting effects of obesity and NAFLD on mitochondrial GSH levels have also been reported. For example, decreased [53] and increased [61] levels of mitochondrial GSH have been reported in liver of ob/ob mice. In addition, two glutathione-replenishing agents, S-adenosylmethionine and 2(RS)-n-propylthiazolidine-4(R)-carboxylic acid (i.e. an L-cysteine pro-drug), were shown to protect against steatohepatitis induced by the methionine-choline deficient diet; however, these agents did not prevent alterations in GSH and GSSG levels in response to steatohepatitis [88]. Nonetheless, increased GSSG concentrations were found in the blood of NASH patients suggesting defects in glutathione metabolism [89]. Taken together, these findings demonstrate that while a potential player in the pathogenesis of fatty liver disease, additional studies are necessary to delineate the precise role of mitochondrial GSH depletion in the pathogenesis of alcohol and non-alcohol mediated mitochondrial dysfunction and liver injury.

Interplay Between Nitric Oxide and Mitochondria in Fatty Liver Disease

Nitric oxide (NO) and other RNS are increased in response to chronic alcohol consumption through induction of inducible nitric oxide synthase (iNOS) [82,90]. In normal healthy liver iNOS levels are very low or undetectable [91], whereas during inflammation and other disease states iNOS levels are increased in liver due to infiltrating inflammatory cells and via induction in Kupffer cells (resident liver macrophage), hepatocytes, and biliary epithelial cells [9294]. Studies also report increased iNOS expression in liver of ob/ob mice [53], CCl4-induced steatotic liver following normothermic ischemia [95], and in the obese fa/fa rat exposed to binge alcohol [5]. This is important for hepatotoxicity because NO and peroxynitrite (ONOO ) are known to mediate mitochondrial dysfunction [96,97] and increases in iNOS expression in fatty liver are correlated with nitration of mitochondrial proteins [53,98]. Moreover, NO regulates mitochondrial respiration through reversible binding at the redox-active heme site in cytochrome c oxidase [99101] and affects mitochondrial biogenesis through interactions with soluble guanylate cyclase [102]. Studies have shown that mitochondria from chronic alcohol-fed animals are more sensitive to the inhibitory effect of NO on respiration [90,103]. While it is known that chronic alcohol consumption results in cytochrome c oxidase defects [82,90,104], it is likely that this alteration in NO responsiveness may be due to alcohol-dependent changes in interactions of NO with sites other than Complex IV. It is postulated that this disruption in NO signaling contributes to alcohol hepatotoxicity through inhibition of ATP synthesis, increased ROS, and inability to adapt to hypoxic stress [105]. This concept is supported because increased sensitivity to NO is absent in iNOS mice exposed to chronic alcohol [90]. Similarly, alcohol-associated inflammation and steatosis is abrogated in iNOS mice [90,106]. Taken together, these studies demonstrate that iNOS induction is linked to alterations in NO-dependent control of mitochondrial respiration, which potentially contributes to the development of alcoholic steatohepatitis. A scheme summarizing the effects of alcohol and obesity to disrupt mitochondrial function is provided in Figure 2.

An external file that holds a picture, illustration, etc.
Object name is nihms46170f2.jpg
Alcohol and obesity induced mitochondrial dysfunction and oxidative stress

Alcohol and obesity disrupt the functioning of the oxidative phosphorylation (OxPhos) system which results in the generation of superoxide (O2 ) within Complexes I and III. The O2 is dismutated to hydrogen peroxide (H2O2) by manganese superoxide dismutase (MnSOD) that can then diffuse from the mitochondrion into the cytoplasm to affect cellular redox signaling pathways. Alcohol and obesity also cause an increase in iNOS expression that leads to diffusion of nitric oxide (NO) into mitochondria where it inhibits cytochrome c oxidase; reacts with O2 to form highly reactive peroxynitrite (ONOO ). Hydrogen peroxide, O2 , and ONOO can initiate reactions that lead to increased generation of reactive lipid species (RLS), induce post-translational modification of proteins, and damage mitochondrial DNA (mtDNA).

It is also important to highlight the beneficial effects of NO in the context of liver diseases. Studies implicate that decreased production of NO from endothelial NOS (eNOS) contributes to liver pathology via dysregulation of blood flow and oxygen delivery [107]. In support of this, NO-donor administration and eNOS overexpression have been shown to prevent liver injury in animal models of hepatotoxicity [108,109]. For example, the oral administration of S-nitroso-N-acetylcysteine prevented the onset of NAFLD in response to a methionine-choline deficient diet [110,111]; however, it is unclear whether the benefit was due to NO or N-acetylcysteine exposure. Recently, inhaled NO administration was shown to accelerate the restoration of liver function in transplantation patients presumably through increased circulating levels of nitrite [112], a newly recognized vascular endocrine transporter of NO [113] that protects against heart and liver ischemia-reperfusion injury [114]. In models of chronic alcohol consumption eNOS activity is decreased in liver through increased expression of the inhibitory protein caveolin-1 and/or decreased eNOS phosphorylation [115,116]. Similarly, Nanji and colleagues have demonstrated L-arginine supplementation prevents and reverses chronic alcohol mediated liver injury [117,118]. Based on this, they propose that decreased NO from non-parenchymal cells, i.e. endothelial cells and eNOS, and increased NO from hepatocyte iNOS work in concert to exacerbate chronic alcohol mediated liver injury [117,118]. While these data support the hypothesis that the source, site, and concentration of NO produced are critical for determining the functional consequence of NO, i.e. good or bad NO, the complex interplay between NO and mitochondria remains to be defined in the context of fatty liver disease from alcohol and obesity. Moreover, the critical importance of understanding the regulation of the mitochondrial respiratory chain by NO in hepatic physiology and pathophysiology is supported by several studies demonstrating that localized NO production can occur within the organelle through a specific mitochondrial NOS isoform or via metabolism of nitrite to NO [119121].

Genetic Factors Determining the Pathobiology of Fatty Liver Diseases – “Oxidative Stress Responsive Genes”

Emerging evidence indicates that genetic factors contribute to increase the risk and severity of alcohol and non-alcohol induced liver disease [3,122]. Indeed, several genes have been identified as influencing the impact of oxidative stress in the pathogenesis of fatty liver disease. Studies have reported genetic polymorphisms in the genes encoding enzymes responsible for alcohol metabolism; alcohol dehydrogenase, CYP450 2E1, and the mitochondrial low Km aldehyde dehydrogenase [123]. Unfortunately, only weak associations between these polymorphisms and increased alcohol hepatotoxicity have been shown [124].

In contrast to these findings, a polymorphism responsible for a valine to alanine substitution in the mitochondrial targeting sequence for manganese superoxide dismutase (SOD2) was found to be a major risk factor for severe alcoholic liver disease in two separate studies [125,126]. The alanine-SOD2 variant, whose pre-sequence is an α-helix, is readily imported and achieves high activity in mitochondria, whereas the valine-SOD2 variant, whose pre-sequence is a partial β-sheet, is poorly imported resulting in low SOD2 activity [127,128]. It is proposed that the presence of the high-activity, alanine-SOD2 variant will increase hydrogen peroxide production in mitochondria, which might have the effect to disrupt cellular redox signaling pathways and initiate oxidative damage [129]. This is supported by studies demonstrating an interaction of the high-activity, alanine-SOD2 variant with a low activity glutathione peroxidase-1 variant (leucine-GPx1) to increase hepatic iron accumulation and risk for hepatocellular carcinoma in alcoholic cirrhotics [129]. Indeed, cirrhotics possessing two copies of the low-activity, valine-SOD2 variant and high-activity, proline-GPx1 variant are protected from cancer. Furthermore, chronic alcohol consumption is known to increase SOD2 activity [130,131] and decrease mitochondria GPx1 activity [81]. Thus, an imbalance resulting in increased hydrogen peroxide formation and decreased detoxification may be a critical event for the initiation and progression of alcohol induced liver injury. With this said, however, studies performed in the UK with a larger cohort of alcoholic patients showed no association among the valine-alanine SOD2 polymorphism, alcohol-dependent markers of oxidative stress, and liver fibrosis [132]. Preliminary studies indicate that the SOD2 polymorphism may be correlated to fibrosis in NAFLD patients [133].

Other studies have reported associations between genetic polymorphisms in endotoxin receptors, cytokines, and fibrosis genes with the pathogenesis of fatty liver diseases. The role of endotoxin mediated cytokine release in the etiology of alcohol hepatotoxicity was established by Thurman and colleagues in the 1990’s through an elegant series of experimental animal studies [134,135]. Likewise, some genetic studies have shown an association between alcoholic liver disease [136] and NASH [137] with a polymorphism present in the CD14 promoter that results in increased CD14 levels [138]. The CD14 receptor binds endotoxin and enhances signaling through the toll-like receptor-4 (TLR4). However, other investigations have reported no association between the CD14 and TLR4 polymorphisms and established alcoholic liver disease [139,140]. Similar conflicting results have been reported for the cytokines TNF-α and interleukin-10 [140]. Moreover, while fibrogenesis/fibrinolysis genes are predicted to be strong candidates to influence liver fibrosis, only one report has shown a link between transforming growth factor-β 1 (TGF-β 1) and angiotensinogen polymorphisms and obesity induced liver fibrosis [141]. Lastly, it should be mentioned that an association between naturally occurring variations in the mitochondrial genome and conditions of the cardiometabolic syndrome has been proposed [142,143]. Recently, Pravenec et al. [142] using conplastic rat strains with the same nuclear genome but dissimilar mitochondrial genomes encoding oxidative phosphorylation proteins of differing amino acid composition showed differences in glucose and glycogen metabolism; two risk factors for type 2 diabetes. Whether mtDNA variants are linked to risk factors associated with steatosis, hepatitis, and fibrosis/cirrhosis is a stimulating concept and one that clearly requires further study. Taken together, these findings indicate that genetic factors may contribute to the pathobiology of alcohol and non-alcohol induced steatohepatitis and/or fibrosis, however additional investigations with larger populations are needed to solidify these linkages.

Are Antioxidants Feasible Treatments for Fatty Liver Disease?

While abstinence and weight loss are critically important for ensuring successful outcomes from alcohol and obesity induced liver diseases, respectively, numerous therapeutic agents have been investigated for treatment of these liver diseases (Figure 3). Unfortunately, no proven drug therapies have emerged as being highly effective. This section will provide a brief overview of some of the newer agents that show promise in treatment of fatty liver diseases.

An external file that holds a picture, illustration, etc.
Object name is nihms46170f3.jpg
Proposed therapeutic agents for the treatment of fatty liver diseases

Corticosteriods, pentoxifylline, propylthiouracil, and anti-TNFα have all been used to treat patients with alcoholic hepatitis with some success; however, issues regarding timing of administration in relation to alcohol exposure and efficacy need to be addressed through more randomized clinical trials [144]. Similarly, insulin sensitizing drugs such as metformin and the thiazolidinediones show promise in treating fatty liver disease. Metformin was shown to prevent alcohol induced liver injury in mice [145]. Interestingly, the protective effect of metformin in this study did not correlate to AMP kinase activation, but via prevention of plasminogen activator inhibitor (PAI)-I upregulation by alcohol [145]. Rosiglitazone and pioglitazone, thiazolidinedione peroxisomal proliferator-activated receptor (PPAR)-γ agonists, decrease insulin resistance and have been shown to decrease TNF-α levels and inflammation in rodent models of NASH [146150]. Similarly, the PPAR-γ agonist pioglitazone prevents alcohol induced liver injury through interactions with the c-Met signaling pathway resulting in decreased triglyceride synthesis [151]. It should also be mentioned that treatment with the PPAR-α agonist WY14,643 prevented alcohol induced steatosis via up-regulation of β-oxidation activity [152]. However caution should be exercised when using these medications because recent reports have demonstrated mitochondrial abnormalities associated with rosiglitazone therapy in both experimental [153] and human patients [154] with NAFLD.

Both traditional and newly recognized antioxidants have been proposed to slow the progression, decrease the severity, and/or reverse pathology from fatty liver diseases presumably through their ability to decrease oxidative stress in tissues. And, like the therapeutic agents described above, results using antioxidants have been varied with some antioxidants showing positive effects and others not. In both adults and children with NASH, treatment with α-tocopherol (vitamin E) has been shown to improve serum liver enzymes [155157]. Similarly, NASH patients treated with vitamin E (400 IU/day) and pioglitazone for 6 months showed significant improvement in liver histology over vitamin E treatment alone [158]. While results with α-tocopherol are promising, recent reports have suggested that γ-tocopherol may be a more effective therapeutic agent in treating diseases involving oxidant stress and inflammation due to its potent anti-inflammatory activity [159,160]. Furthermore, Murphy and colleagues have developed novel strategies to “target” traditional antioxidants, like vitamin E, ubiquinone, and lipoic acid to the mitochondrion by attaching them to a lipophilic triphenylphosphonium moiety [161163]. This modification allows the antioxidant to accumulate several-hundred fold in the mitochondrion in response to the mitochondrial membrane potential where it can exert maximum benefit. Indeed, mitochondria-targeted ubiquinone (a.k.a. MitoQ) was shown to decrease cardiac ischemia-reperfusion injury [164]. In contrast, mitochondria-targeted vitamin E (a.k.a. MitoE) caused neurotoxicity in a hypoxic-ischemia model of striatal injury [165]. Thus, additional studies are required to demonstrate the safety and efficacy of these novel mitochondrially targeted antioxidants in diseases where mitochondrial damage and oxidant stress are proposed to play a role in pathology.

Numerous other antioxidants show efficacy in treating fatty liver diseases and include N-acetylcysteine [166], zinc [167,168], silymarin [169], and ursodeoxycholic acid [170], just to name a few. Please refer to the following excellent review articles for additional information on antioxidant therapy for treatment of fatty liver diseases from multiple causes [144,171174].

Defects in methionine metabolism occur in response to both alcohol and non-alcohol induced liver disease and are postulated to contribute to the disease process. One key metabolite of the methionine metabolic pathway is S-adenosylmethionine (SAM), which is the main methyl donor for biological reactions and serves as a precursor in glutathione and polyamine synthesis [175].

In mammalian cells SAM is synthesized by the enzyme methionine adenosylmethytransferase (MAT). Alcohol and other hepatotoxicants decrease hepatic levels of SAM due to decreased activity of the hepatocyte specific form of MAT [176] and methionine synthase [177]. Studies have shown that NO inactivates rat liver MAT in vivo by S-nitrosation of cysteine 121, which leads to decreased synthesis and depletion of hepatic SAM [178,179]. SAM depletion correlates with DNA hypomethylation and strand breaks [176] and several parameters of oxidative stress including decreased GSH and increased lipid peroxidation [180]. Accordingly, exogenous administration of SAM in animal models of alcohol toxicity has been shown to be protective [82,181,182]. SAM supplementation was also shown to significantly improve the outcome of alcoholic cirrhotic patients [183]. Emerging evidence suggests that the mitochondrion may be an important target for the benefits afforded by SAM in the treatment of fatty liver disease.

Studies by Mato and colleagues showed that MAT1A mice, which are deficient in SAM, spontaneously develop steatohepatitis and hepatocellular carcinoma demonstrating that SAM depletion primes the liver for progression to more severe injury [184,185]. Mitochondria isolated from liver of MAT1A− /− mice also have decreased levels of the mitochondrial encoded subunits for cytochrome c oxidase and reduced membrane potential [186]. This finding is consistent with the concept that SAM plays an essential role in the maintenance and proper function of mitochondria and the oxidative phosphorylation system. Early studies showed that co-administration of SAM during alcohol feeding maintained mitochondrial GSH transport and GSH levels, which preserved ATP levels and membrane potential [76,77]. Recently, we have extended these earlier studies and shown that SAM supplementation prevents alcohol-dependent decreases in mitochondrial respiration, increases in superoxide production, mtDNA damage, and iNOS induction [82]. Similarly, SAM prevented alcohol-associated losses in both nuclear and mitochondrial encoded subunits of cytochrome c oxidase, which preserved oxidase activity [82]. These results are supported by observations that depletion of mitochondrial SAM levels sensitizes HepG2 cells to TNF-α cytotoxicity [187].

In addition to SAM, the methionine pathway metabolite betaine has been shown to prevent hepatotoxicity in animal models of alcohol [188,189] and diet [190] induced steatosis, as well as improve serum liver enzymes and liver histology in NASH patients [191]. Betaine, a natural antioxidant, functions as an osmolyte and a methyl donor for the remethylation of homocysteine, “the bad thiol” [192], back to methionine [193]. Hyperhomocysteinemia is associated with steatosis presumably through induction of endoplasmic reticulum stress and activation of SREBPs leading to increased hepatic synthesis and uptake of triglycerides and cholesterol [194]. Indeed, hyperhomocysteinemia may be a suitable predictor of NASH [195]. Preliminary data demonstrates that betaine supplementation prevented chronic alcohol-induced steatosis and losses in the respiratory chain complexes, particularly subunits of Complex I and IV [196]. Taken together, these data reveal a novel molecular mechanism through which SAM and betaine may work to prevent one key component of alcohol and obesity-induced liver disease; mitochondria dysfunction.

Summary

Strong evidence indicates that the pathogenesis of chronic alcohol and obesity/type 2 diabetes induced fatty liver disease is linked to mitochondrial dysfunction. The initial formation of an inflammatory response and increased production of ROS/RNS associated with the metabolic alterations induced by these conditions leads to modifications of the mitochondrial genome and proteome. These alterations result in loss of mitochondrial respiration, the inability to maintain sufficient ATP concentrations, and a further enhancement of ROS/RNS production. Indeed, the role of oxidative stress in these disease processes has been demonstrated in numerous experimental models. Thus, the goal of current and future investigations will be to provide a thorough characterization of the molecular mechanisms involved in the pathogenesis of fatty liver diseases with particular emphasis on defining the role of mitochondrial oxidative stress in the disease process. A more complete understanding of the ROS/RNS mediated effects on the spectrum of alcohol and obesity induced liver disease will facilitate the development of new mitochondrially targeted molecular medicines for treatment.

Acknowledgments

This work was funded by NIH/NIAAA grants AA15172 and DK73775 awarded to SMB. Ms. Adrienne L. King is supported by a Research Supplement to Promote Diversity in Health-Related Research linked to parent grant AA15172.

Department of Environmental Health Sciences, Center for Free Radical Biology, University of Alabama at Birmingham
*Corresponding Author: Shannon M. Bailey, PhD, Associate Professor, Department of Environmental Health Sciences, Center for Free Radical Biology, University of Alabama at Birmingham, Ryals Building, Room 623, 1530 3 Avenue South, Birmingham, AL 35294 USA, Phone: 205-934-7070, Fax: 205-975-6341, Email: ude.bau@yeliabs
Publisher's Disclaimer

Abstract

Fatty liver disease associated with chronic alcohol consumption or obesity/type 2 diabetes has emerged as a serious public health problem. Steatosis, accumulation of triglyceride in hepatocytes, is now recognized as a critical “first-hit” in the pathogenesis of liver disease. It is proposed that steatosis “primes” the liver to progress to more severe liver pathologies when individuals are exposed to subsequent metabolic and/or environmental stressors or “second-hits”. Genetic risk factors can also influence the susceptibility and severity of fatty liver disease. Furthermore, oxidative stress, disrupted nitric oxide (NO) signaling, and mitochondrial dysfunctional are proposed to be key molecular events that accelerate or worsen steatosis and initiate progression to steatohepatitis and fibrosis. This review article will discuss the following topics regarding the pathobiology and molecular mechanisms responsible for fatty liver disease: 1) the “two-hit” or “multi-hit” hypothesis; 2) the role of mitochondrial bioenergetic defects and oxidant stress; 3) interplay between NO and mitochondria in fatty liver disease; 4) genetic risk factors and oxidative stress responsive genes; and 5) the feasibility of antioxidants for treatment.

Keywords: Liver, alcohol, obesity, steatosis, mitochondria, oxidative stress, nitric oxide, antioxidants, proteomics, AFLD, ASH, NAFLD, NASH
Abstract

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Footnotes

References

  • 1. Centers for Disease Control PreventionAlcohol-attributable death and years of potential life lost - United States, 2001. Morb Mortal Wkly Rep. 2004;53:866–70.[PubMed][Google Scholar]
  • 2. Bellentani S, Saccoccio G, Costa G, Tiribelli C, Manenti F, Sodde M, Saveria Croce L, Sasso F, Pozzato G, Cristianini G, Brandi G. Drinking habits as cofactors of risk for alcohol induced liver damage. The Dionysos Study Group. Gut. 1997;41:845–50.
  • 3. Day CPGenes or environment to determine alcoholic liver disease and non-alcoholic fatty liver disease. Liver Int. 2006;26:1021–8.[PubMed][Google Scholar]
  • 4. Marrero JA, Fontana RJ, Fu S, Conjeevaram HS, Su GL, Lok ASAlcohol, tobacco and obesity are synergistic risk factors for hepatocellular carcinoma. J Hepatol. 2005;42:218–24.[PubMed][Google Scholar]
  • 5. Carmiel-Haggai M, Cederbaum AI, Nieto NBinge ethanol exposure increases liver injury in obese rats. Gastroenterology. 2003;125:1818–33.[PubMed][Google Scholar]
  • 6. Ludwig J, Viggiano TR, McGill DB, Oh BJNonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin Proc. 1980;55:434–8.[PubMed][Google Scholar]
  • 7. Mehta K, Van Thiel DH, Shah N, Mobarhan SNonalcoholic fatty liver disease: pathogenesis and the role of antioxidants. Nutr Rev. 2002;60:289–93.[PubMed][Google Scholar]
  • 8. Ruhl CE, Everhart JEEpidemiology of nonalcoholic fatty liver. Clin Liver Dis. 2004;8:501–19.[PubMed][Google Scholar]
  • 9. Papandreou D, Rousso I, Mavromichalis IUpdate on non-alcoholic fatty liver disease in children. Clin Nutr. 2007;26:409–15.[PubMed][Google Scholar]
  • 10. Bunout DNutritional and metabolic effects of alcoholism: their relationship with alcoholic liver disease. Nutrition. 1999;15:583–9.[PubMed][Google Scholar]
  • 11. Howard AA, Arnsten JH, Gourevitch MNEffect of alcohol consumption on diabetes mellitus: a systematic review. Ann Intern Med. 2004;140:211–9.[PubMed][Google Scholar]
  • 12. van de Wiel ADiabetes mellitus and alcohol. Diabetes Metab Res Rev. 2004;20:263–7.[PubMed][Google Scholar]
  • 13. Begriche K, Igoudjil A, Pessayre D, Fromenty BMitochondrial dysfunction in NASH: causes, consequences and possible means to prevent it. Mitochondrion. 2006;6:1–28.[PubMed][Google Scholar]
  • 14. Farrell GC, Larter CZNonalcoholic fatty liver disease: from steatosis to cirrhosis. Hepatology. 2006;43:S99–S112.[PubMed][Google Scholar]
  • 15. Feldstein AE, Werneburg NW, Canbay A, Guicciardi ME, Bronk SF, Rydzewski R, Burgart LJ, Gores GJFree fatty acids promote hepatic lipotoxicity by stimulating TNF-alpha expression via a lysosomal pathway. Hepatology. 2004;40:185–94.[PubMed][Google Scholar]
  • 16. Feldstein AE, Werneburg NW, Li Z, Bronk SF, Gores GJBax inhibition protects against free fatty acid-induced lysosomal permeabilization. Am J Physiol Gastrointest Liver Physiol. 2006;290:G1339–46.[Google Scholar]
  • 17. Wei Y, Wang D, Pagliassotti MJSaturated fatty acid-mediated endoplasmic reticulum stress and apoptosis are augmented by trans-10, cis-12-conjugated linoleic acid in liver cells. Mol Cell Biochem. 2007;303:105–13.[PubMed][Google Scholar]
  • 18. Wei Y, Wang D, Topczewski F, Pagliassotti MJSaturated fatty acids induce endoplasmic reticulum stress and apoptosis independently of ceramide in liver cells. Am J Physiol Endocrinol Metab. 2006;291:E275–81.[PubMed][Google Scholar]
  • 19. Joshi-Barve S, Barve SS, Amancherla K, Gobejishvili L, Hill D, Cave M, Hote P, McClain CJPalmitic acid induces production of proinflammatory cytokine interleukin-8 from hepatocytes. Hepatology. 2007;46:823–30.[PubMed][Google Scholar]
  • 20. Landar A, Shiva S, Levonen AL, Oh JY, Zaragoza C, Johnson MS, Darley-Usmar VMInduction of the permeability transition and cytochrome c release by 15-deoxy-Delta12,14-prostaglandin J2 in mitochondria. Biochem J. 2006;394:185–95.[Google Scholar]
  • 21. Landar A, Zmijewski JW, Dickinson DA, Le Goffe C, Johnson MS, Milne GL, Zanoni G, Vidari G, Morrow JD, Darley-Usmar VMInteraction of electrophilic lipid oxidation products with mitochondria in endothelial cells and formation of reactive oxygen species. Am J Physiol Heart Circ Physiol. 2006;290:H1777–87.[PubMed][Google Scholar]
  • 22. Fromenty B, Robin MA, Igoudjil A, Mansouri A, Pessayre DThe ins and outs of mitochondrial dysfunction in NASH. Diabetes Metab. 2004;30:121–38.[PubMed][Google Scholar]
  • 23. Kang L, Chen X, Sebastian BM, Pratt BT, Bederman IR, Alexander JC, Previs SF, Nagy LEChronic ethanol and triglyceride turnover in white adipose tissue in rats: inhibition of the anti-lipolytic action of insulin after chronic ethanol contributes to increased triglyceride degradation. J Biol Chem. 2007;282:28465–73.[PubMed][Google Scholar]
  • 24. He L, Marecki JC, Serrero G, Simmen FA, Ronis MJ, Badger TMDose Dependent Effects of Alcohol on Insulin Signaling: Partial Explanation for Biphasic Alcohol Impact on Human Health. Mol Endocrinol. 2007[PubMed][Google Scholar]
  • 25. He L, Simmen FA, Mehendale HM, Ronis MJ, Badger TM. Chronic ethanol intake impairs insulin signaling in rats by disrupting Akt association with the cell membrane. Role of TRB3 in inhibition of Akt/protein kinase B activation. J Biol Chem. 2006;281:11126–34.[PubMed]
  • 26. You M, Fischer M, Deeg MA, Crabb DWEthanol induces fatty acid synthesis pathways by activation of sterol regulatory element-binding protein (SREBP) J Biol Chem. 2002;277:29342–7.[PubMed][Google Scholar]
  • 27. You M, Matsumoto M, Pacold CM, Cho WK, Crabb DWThe role of AMP-activated protein kinase in the action of ethanol in the liver. Gastroenterology. 2004;127:1798–808.[PubMed][Google Scholar]
  • 28. Tomita K, Tamiya G, Ando S, Kitamura N, Koizumi H, Kato S, Horie Y, Kaneko T, Azuma T, Nagata H, Ishii H, Hibi TAICAR, an AMPK activator, has protective effects on alcohol-induced fatty liver in rats. Alcohol Clin Exp Res. 2005;29:240S–5S.[PubMed][Google Scholar]
  • 29. Xu A, Wang Y, Keshaw H, Xu LY, Lam KS, Cooper GJThe fat-derived hormone adiponectin alleviates alcoholic and nonalcoholic fatty liver diseases in mice. J Clin Invest. 2003;112:91–100.[Google Scholar]
  • 30. You M, Considine RV, Leone TC, Kelly DP, Crabb DWRole of adiponectin in the protective action of dietary saturated fat against alcoholic fatty liver in mice. Hepatology. 2005;42:568–77.[Google Scholar]
  • 31. Winder WW, Hardie DGAMP-activated protein kinase, a metabolic master switch: possible roles in type 2 diabetes. Am J Physiol. 1999;277:E1–10.[PubMed][Google Scholar]
  • 32. Yeon JE, Califano S, Xu J, Wands JR, De La Monte SMPotential role of PTEN phosphatase in ethanol-impaired survival signaling in the liver. Hepatology. 2003;38:703–14.[PubMed][Google Scholar]
  • 33. Cidad P, Almeida A, Bolanos JPInhibition of mitochondrial respiration by nitric oxide rapidly stimulates cytoprotective GLUT3-mediated glucose uptake through 5′-AMP-activated protein kinase. Biochem J. 2004;384:629–36.[Google Scholar]
  • 34. Hutchinson DS, Csikasz RI, Yamamoto DL, Shabalina IG, Wikstrom P, Wilcke M, Bengtsson TDiphenylene iodonium stimulates glucose uptake in skeletal muscle cells through mitochondrial complex I inhibition and activation of AMP-activated protein kinase. Cell Signal. 2007;19:1610–20.[PubMed][Google Scholar]
  • 35. Zou MH, Kirkpatrick SS, Davis BJ, Nelson JS, Wiles WGt, Schlattner U, Neumann D, Brownlee M, Freeman MB, Goldman MH. Activation of the AMP-activated protein kinase by the anti-diabetic drug metformin in vivo. Role of mitochondrial reactive nitrogen species. J Biol Chem. 2004;279:43940–51.[PubMed]
  • 36. Pritchard MT, McMullen MR, Stavitsky AB, Cohen JI, Lin F, Medof ME, Nagy LEDifferential contributions of C3, C5, and decay-accelerating factor to ethanol-induced fatty liver in mice. Gastroenterology. 2007;132:1117–26.[Google Scholar]
  • 37. Bykov I, Jauhiainen M, Olkkonen VM, Saarikoski ST, Ehnholm C, Junnikkala S, Vakeva A, Lindros KO, Meri SHepatic gene expression and lipid parameters in complement C3(− /− ) mice that do not develop ethanol-induced steatosis. J Hepatol. 2007;46:907–14.[PubMed][Google Scholar]
  • 38. Yamaguchi K, Yang L, McCall S, Huang J, Yu XX, Pandey SK, Bhanot S, Monia BP, Li YX, Diehl AMInhibiting triglyceride synthesis improves hepatic steatosis but exacerbates liver damage and fibrosis in obese mice with nonalcoholic steatohepatitis. Hepatology. 2007;45:1366–74.[PubMed][Google Scholar]
  • 39. Damelin LH, Coward S, Kirwan M, Collins P, Selden C, Hodgson HJFat-loaded HepG2 spheroids exhibit enhanced protection from Pro-oxidant and cytokine induced damage. J Cell Biochem. 2007;101:723–34.[PubMed][Google Scholar]
  • 40. Bailey SMA review of the role of reactive oxygen and nitrogen species in alcohol-induced mitochondrial dysfunction. Free Radic Res. 2003;37:585–96.[PubMed][Google Scholar]
  • 41. Cunningham CC, Coleman WB, Spach PIThe effects of chronic ethanol consumption on hepatic mitochondrial energy metabolism. Alcohol Alcohol. 1990;25:127–36.[PubMed][Google Scholar]
  • 42. Hoek JBMitochondrial energy metabolism in chronic alcoholism. Curr Topic Bioenerg. 1994;17:197–241.[PubMed][Google Scholar]
  • 43. Coleman WB, Cunningham CCEffects of chronic ethanol consumption on the synthesis of polypeptides encoded by the hepatic mitochondrial genome. Biochim Biophys Acta. 1990;1019:142–50.[PubMed][Google Scholar]
  • 44. Cahill A, Stabley GJ, Wang X, Hoek JBChronic ethanol consumption causes alterations in the structural integrity of mitochondrial DNA in aged rats. Hepatology. 1999;30:881–8.[Google Scholar]
  • 45. Cahill A, Wang X, Hoek JBIncreased oxidative damage to mitochondrial DNA following chronic ethanol consumption. Biochem Biophys Res Commun. 1997;235:286–90.[PubMed][Google Scholar]
  • 46. Venkatraman A, Landar A, Davis AJ, Chamlee L, Sanderson T, Kim H, Page G, Pompilius M, Ballinger S, Darley-Usmar V, Bailey SMModification of the mitochondrial proteome in response to the stress of ethanol-dependent hepatotoxicity. J Biol Chem. 2004;279:22092–101.[PubMed][Google Scholar]
  • 47. Cahill A, Cunningham CCEffects of chronic ethanol feeding on the protein composition of mitochondrial ribosomes. Electrophoresis. 2000;21:3420–6.[PubMed][Google Scholar]
  • 48. Patel VB, Cunningham CCAltered hepatic mitochondrial ribosome structure following chronic ethanol consumption. Arch Biochem Biophys. 2002;398:41–50.[PubMed][Google Scholar]
  • 49. Moon KH, Hood BL, Kim BJ, Hardwick JP, Conrads TP, Veenstra TD, Song BJInactivation of oxidized and S-nitrosylated mitochondrial proteins in alcoholic fatty liver of rats. Hepatology. 2006;44:1218–30.[PubMed][Google Scholar]
  • 50. Venkatraman A, Landar A, Davis AJ, Ulasova E, Page G, Murphy MP, Darley-Usmar V, Bailey SMOxidative modification of hepatic mitochondria protein thiols: effect of chronic alcohol consumption. Am J Physiol Gastrointest Liver Physiol. 2004;286:G521–G7.[PubMed][Google Scholar]
  • 51. Chavin KD, Yang S, Lin HZ, Chatham J, Chacko VP, Hoek JB, Walajtys-Rode E, Rashid A, Chen CH, Huang CC, Wu TC, Lane MD, Diehl AMObesity induces expression of uncoupling protein-2 in hepatocytes and promotes liver ATP depletion. J Biol Chem. 1999;274:5692–700.[PubMed][Google Scholar]
  • 52. Perez-Carreras M, Del Hoyo P, Martin MA, Rubio JC, Martin A, Castellano G, Colina F, Arenas J, Solis-Herruzo JADefective hepatic mitochondrial respiratory chain in patients with nonalcoholic steatohepatitis. Hepatology. 2003;38:999–1007.[PubMed][Google Scholar]
  • 53. Garcia-Ruiz I, Rodriguez-Juan C, Diaz-Sanjuan T, del Hoyo P, Colina F, Munoz-Yague T, Solis-Herruzo JAUric acid and anti-TNF antibody improve mitochondrial dysfunction in ob/ob mice. Hepatology. 2006;44:581–91.[PubMed][Google Scholar]
  • 54. Yang SQ, Lin HZ, Lane MD, Clemens M, Diehl AMObesity increases sensitivity to endotoxin liver injury: implications for the pathogenesis of steatohepatitis. Proc Natl Acad Sci U S A. 1997;94:2557–62.[Google Scholar]
  • 55. Yang SQ, Lin HZ, Mandal AK, Huang J, Diehl AMDisrupted signaling and inhibited regeneration in obese mice with fatty livers: implications for nonalcoholic fatty liver disease pathophysiology. Hepatology. 2001;34:694–706.[PubMed][Google Scholar]
  • 56. Cortez-Pinto H, Chatham J, Chacko VP, Arnold C, Rashid A, Diehl AMAlterations in liver ATP homeostasis in human nonalcoholic steatohepatitis: a pilot study. JAMA. 1999;282:1659–64.[PubMed][Google Scholar]
  • 57. Miyamoto KSWFHepatic adenine nucleotide metabolism measured in vivo in rats fed ethanol and a high fat-low protein diet. Hepatology. 1988;8:53–60.[PubMed][Google Scholar]
  • 58. Takahashi H, Geoffrion Y, Butler KW, French SW. In vivo hepatic energy metabolism during the progression of alcoholic liver disease: a noninvasive P nuclear magnetic resonance study in rats. Hepatology. 1990;11:65–73.[PubMed]
  • 59. Hoek JB, Cahill A, Pastorino JGAlcohol and mitochondria: a dysfunctional relationship. Gastroenterology. 2002;122:2049–63.[Google Scholar]
  • 60. Gao D, Wei C, Chen L, Huang J, Yang S, Diehl AMOxidative DNA damage and DNA repair enzyme expression are inversely related in murine models of fatty liver disease. Am J Physiol Gastrointest Liver Physiol. 2004;287:G1070–7.[PubMed][Google Scholar]
  • 61. Yang S, Zhu H, Li Y, Lin H, Gabrielson K, Trush MA, Diehl AMMitochondrial adaptations to obesity-related oxidant stress. Arch Biochem Biophys. 2000;378:259–68.[PubMed][Google Scholar]
  • 62. Bailey SM, Cunningham CCAcute and chronic ethanol increases reactive oxygen species generation and decreases viability in fresh, isolated rat hepatocytes. Hepatology. 1998;28:1318–26.[PubMed][Google Scholar]
  • 63. Fernandez-Checa JC, Kaplowitz N, Garcia-Ruiz C, Colell A, Miranda M, Mari M, Ardite E, Morales AGSH transport in mitochondria: defense against TNF-induced oxidative stress and alcohol-induced defect. Am J Physiol Gastrointest Liver Physiol. 1997;273:G7–17.[PubMed][Google Scholar]
  • 64. Mantena SK, King AL, Andringa KK, Landar A, Darley-Usmar V, Bailey SMNovel interactions of mitochondria and reactive oxygen/nitrogen species in alcohol mediated liver diseases. World J Gastroenterol. 2007;13:4967–73.[Google Scholar]
  • 65. Han D, Canali R, Rettori D, Kaplowitz NEffect of glutathione depletion on sites and topology of superoxide and hydrogen peroxide production in mitochondria. Mol Pharmacol. 2003;64:1136–44.[PubMed][Google Scholar]
  • 66. Lambert AJ, Brand MDSuperoxide production by NADH:ubiquinone oxidoreductase (complex I) depends on the pH gradient across the mitochondrial inner membrane. Biochem J. 2004;382:511–7.[Google Scholar]
  • 67. Hinokio Y, Suzuki S, Komatu K, Ohtomo M, Onoda M, Matsumoto M, Hirai S, Sato Y, Akai H, Abe K, et al A new mitochondrial DNA deletion associated with diabetic amyotrophy, diabetic myoatrophy and diabetic fatty liver. Muscle Nerve. 1995;3:S142–9.[PubMed][Google Scholar]
  • 68. Kawahara H, Fukura M, Tsuchishima M, Takase SMutation of mitochondrial DNA in livers from patients with alcoholic hepatitis and nonalcoholic steatohepatitis. Alcohol Clin Exp Res. 2007;31:S54–60.[PubMed][Google Scholar]
  • 69. Robin MA, Sauvage I, Grandperret T, Descatoire V, Pessayre D, Fromenty BEthanol increases mitochondrial cytochrome P450 2E1 in mouse liver and rat hepatocytes. FEBS Lett. 2005;579:6895–902.[PubMed][Google Scholar]
  • 70. Nordblom GD, Coon MJHydrogen peroxide formation and stoichiometry of hydroxylation reactions catalyzed by highly purified liver microsomal cytochrome P-450. Arch Biochem Biophys. 1977;180:343–7.[PubMed][Google Scholar]
  • 71. Adam-Vizi VProduction of reactive oxygen species in brain mitochondria: contribution by electron transport chain and non-electron transport chain sources. Antioxid Redox Signal. 2005;7:1140–9.[PubMed][Google Scholar]
  • 72. Tretter L, Adam-Vizi VAlpha-ketoglutarate dehydrogenase: a target and generator of oxidative stress. Philos Trans R Soc Lond B Biol Sci. 2005;360:2335–45.[Google Scholar]
  • 73. Tretter L, Takacs K, Hegedus V, Adam-Vizi VCharacteristics of alpha-glycerophosphate-evoked H2O2 generation in brain mitochondria. J Neurochem. 2007;100:650–63.[PubMed][Google Scholar]
  • 74. Garcia-Ruiz C, Fernandez-Checa JCMitochondrial glutathione: hepatocellular survival-death switch. J Gastroenterol Hepatol. 2006;21 Suppl 3:S3–6.[PubMed][Google Scholar]
  • 75. Garcia-Ruiz C, Morales A, Ballesta A, Rodes J, Kaplowitz N, Fernandez-Checa JCEffect of chronic ethanol feeding on glutathione and functional integrity of mitochondria in periportal and perivenous rat hepatocytes. J Clin Invest. 1994;94:193–201.[Google Scholar]
  • 76. Colell A, Garcia-Ruiz C, Morales A, Ballesta A, Ookhtens M, Rodes J, Kaplowitz N, Fernandez-Checa JCTransport of reduced glutathione in hepatic mitochondria and mitoplasts from ethanol-treated rats: effect of membrane physical properties and S-adenosyl-L-methionine. Hepatology. 1997;26:699–708.[PubMed][Google Scholar]
  • 77. Garcia-Ruiz C, Morales A, Colell A, Ballesta A, Rodes J, Kaplowitz N, Fernandez-Checa JCFeeding S-adenosyl-L-methionine attenuates both ethanol-induced depletion of mitochondrial glutathione and mitochondrial dysfunction in periportal and perivenous rat hepatocytes. Hepatology. 1995;21:207–14.[PubMed][Google Scholar]
  • 78. Colell A, Garcia-Ruiz C, Miranda M, Ardite E, Mari M, Morales A, Corrales F, Kaplowitz N, Fernandez-Checa JCSelective glutathione depletion of mitochondria by ethanol sensitizes hepatocytes to tumor necrosis factor. Gastroenterology. 1998;115:1541–51.[PubMed][Google Scholar]
  • 79. Pastorino JG, Hoek JBEthanol potentiates tumor necrosis factor-alpha cytotoxicity in hepatoma cells and primary rat hepatocytes by promoting induction of the mitochondrial permeability transition. Hepatology. 2000;31:1141–52.[PubMed][Google Scholar]
  • 80. Mari M, Caballero F, Colell A, Morales A, Caballeria J, Fernandez A, Enrich C, Fernandez-Checa JC, Garcia-Ruiz CMitochondrial free cholesterol loading sensitizes to TNF- and Fas-mediated steatohepatitis. Cell Metab. 2006;4:185–98.[PubMed][Google Scholar]
  • 81. Bailey SM, Patel VB, Young TA, Asayama K, Cunningham CCChronic ethanol consumption alters the glutathione/glutathione peroxidase-1 system and protein oxidation status in rat liver. Alcohol Clin Exp Res. 2001;25:726–33.[PubMed][Google Scholar]
  • 82. Bailey SM, Robinson G, Pinner A, Chamlee L, Ulasova E, Pompilius M, Page GP, Chhieng D, Jhala N, Landar A, Kharbanda KK, Ballinger S, Darley-Usmar VS-adenosylmethionine prevents chronic alcohol-induced mitochondrial dysfunction in the rat liver. Am J Physiol Gastrointest Liver Physiol. 2006;291:G857–67.[PubMed][Google Scholar]
  • 83. Deaciuc IV, Fortunato F, D’Souza NB, Hill DB, Schmidt J, Lee EY, McClain CJModulation of capase-3 activity and Fas ligand mRNA expression in rat liver cells in vivo by alcohol and lipopolysaccharide. Alcohol Clin Exp Res. 1999;23:349–56.[PubMed][Google Scholar]
  • 84. Sastre J, Minana JB, Alguacil P, Malet G, Gomez-Cambronero L, Marin JA, Vina JR, Vina J. Chronic ethanol feeding causes oxidative stress in rat liver mitochondria. Prevention by S-adenosyl methionine. Free Radic Res. 1999;30:325–7.[PubMed]
  • 85. Mari M, Cederbaum AICYP2E1 overexpression in HepG2 cells induces glutathione synthesis by transcriptional activation of gamma-glutamylcysteine synthetase. J Biol Chem. 2000;275:15563–71.[PubMed][Google Scholar]
  • 86. Dickinson DA, Moellering DR, Iles KE, Patel RP, Levonen AL, Wigley A, Darley-Usmar VM, Forman HJCytoprotection against oxidative stress and the regulation of glutathione synthesis. Biol Chem. 2003;384:527–37.[PubMed][Google Scholar]
  • 87. Gutierrez J, Ballinger SW, Darley-Usmar VM, Landar AFree radicals, mitochondria, and oxidized lipids: the emerging role in signal transduction in vascular cells. Circ Res. 2006;99:924–32.[PubMed][Google Scholar]
  • 88. Oz HS, Im HJ, Chen TS, de Villiers WJ, McClain CJGlutathione-enhancing agents protect against steatohepatitis in a dietary model. J Biochem Mol Toxicol. 2006;20:39–47.[Google Scholar]
  • 89. Nobili V, Pastore A, Gaeta LM, Tozzi G, Comparcola D, Sartorelli MR, Marcellini M, Bertini E, Piemonte FGlutathione metabolism and antioxidant enzymes in patients affected by nonalcoholic steatohepatitis. Clin Chim Acta. 2005;355:105–11.[PubMed][Google Scholar]
  • 90. Venkatraman A, Shiva S, Wigley A, Ulasova E, Chhieng D, Bailey SM, Darley-Usmar VThe role of iNOS in alcohol-dependent hepatoxicity and mitochondrial dysfunction. Hepatology. 2004;40:565–73.[PubMed][Google Scholar]
  • 91. Osei SY, Ahima RS, Fabry ME, Nagel RL, Bank NImmunohistochemical localization of hepatic nitric oxide synthase in normal and transgenic sickle cell mice: the effect of hypoxia. Blood. 1996;88:3583–8.[PubMed][Google Scholar]
  • 92. Buttery LD, Evans TJ, Springall DR, Carpenter A, Cohen J, Polak JMImmunochemical localization of inducible nitric oxide synthase in endotoxin-treated rats. Lab Invest. 1994;71:755–64.[PubMed][Google Scholar]
  • 93. Kuo PC, Alfrey EJ, Abe KY, Huie P, Sibley RK, Dafoe DCCellular localization and effect of nitric oxide synthesis in a rat model of orthotopic liver transplantation. Transplantation. 1996;61:305–12.[PubMed][Google Scholar]
  • 94. Wei CL, Khoo HE, Lee KH, Hon WMDifferential expression and localization of nitric oxide synthases in cirrhotic livers of bile duct-ligated rats. Nitric Oxide. 2002;7:91–102.[PubMed][Google Scholar]
  • 95. Koeppel TA, Mihaljevic N, Kraenzlin B, Loehr M, Jesenofsky R, Post S, Palma PEnhanced iNOS gene expression in the steatotic rat liver after normothermic ischemia. Eur Surg Res. 2007;39:303–11.[PubMed][Google Scholar]
  • 96. Radi RNitric oxide, oxidants, and protein tyrosine nitration. Proc Natl Acad Sci U S A. 2004;101:4003–8.[Google Scholar]
  • 97. Radi R, Cassina A, Hodara R, Quijano C, Castro LPeroxynitrite reactions and formation in mitochondria. Free Radic Biol Med. 2002;33:1451–64.[PubMed][Google Scholar]
  • 98. Lee JH, Yang ES, Park JW. Inactivation of NADP+-dependent isocitrate dehydrogenase by peroxynitrite. Implications for cytotoxicity and alcohol-induced liver injury. J Biol Chem. 2003;278:51360–71.[PubMed]
  • 99. Brookes PS, Kraus DW, Shiva S, Doeller JE, Barone MC, Patel RP, Lancaster JR, Jr, Darley-Usmar VControl of mitochondrial respiration by NO*, effects of low oxygen and respiratory state. J Biol Chem. 2003;278:31603–9.[PubMed][Google Scholar]
  • 100. Poderoso JJ, Lisdero C, Schopfer F, Riobo N, Carreras MC, Cadenas E, Boveris AThe regulation of mitochondrial oxygen uptake by redox reactions involving nitric oxide and ubiquinol. J Biol Chem. 1999;274:37709–16.[PubMed][Google Scholar]
  • 101. Torres J, Darley-Usmar V, Wilson MTInhibition of cytochrome c oxidase in turnover by nitric oxide: mechanism and implications for control of respiration. Biochem J. 1995;312(Pt 1):169–73.[Google Scholar]
  • 102. Nisoli E, Clementi E, Moncada S, Carruba MOMitochondrial biogenesis as a cellular signaling framework. Biochem Pharmacol. 2004;67:1–15.[PubMed][Google Scholar]
  • 103. Venkatraman A, Shiva S, Davis AJ, Bailey SM, Brookes PS, Darley-Usmar VChronic alcohol consumption increases the senstivity of rat liver mitochondrial respiration to inhibition by nitric oxide. Hepatology. 2003;38:141–7.[PubMed][Google Scholar]
  • 104. Thayer WS, Rubin EMolecular alterations in the respiratory chain of rat liver after chronic ethanol consumption. J Biol Chem. 1981;256:6090–7.[PubMed][Google Scholar]
  • 105. Shiva S, Oh JY, Landar AL, Ulasova E, Venkatraman A, Bailey SM, Darley-Usmar VMNitroxia: The pathological consequence of dysfunction in the nitric oxide-cytochrome c oxidase signaling pathway. Free Radic Biol Med. 2005;38:297–306.[PubMed][Google Scholar]
  • 106. McKim SE, Gabele E, Isayama F, Lambert JC, Tucker LM, Wheeler MD, Connor HD, Mason RP, Doll MA, Hein DW, Arteel GEInducible nitric oxide synthase is required in alcohol-induced liver injury: studies with knockout mice. Gastroenterology. 2003;125:1834–44.[PubMed][Google Scholar]
  • 107. Liu J, Waalkes MPNitric oxide and chemically induced hepatotoxicity: beneficial effects of the liver-selective nitric oxide donor, V-PYRRO/NO. Toxicology. 2005;208:289–97.[PubMed][Google Scholar]
  • 108. Duranski MR, Elrod JW, Calvert JW, Bryan NS, Feelisch M, Lefer DJGenetic overexpression of eNOS attenuates hepatic ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2006;291:H2980–6.[PubMed][Google Scholar]
  • 109. Rivera-Chavez FA, Toledo-Pereyra LH, Dean RE, Crouch L, Ward PAExogenous and endogenous nitric oxide but not iNOS inhibition improves function and survival of ischemically injured livers. J Invest Surg. 2001;14:267–73.[PubMed][Google Scholar]
  • 110. de Oliveira CP, Simplicio FI, de Lima VM, Yuahasi K, Lopasso FP, Alves VA, Abdalla DS, Carrilho FJ, Laurindo FR, de Oliveira MGOral administration of S-nitroso-N-acetylcysteine prevents the onset of non alcoholic fatty liver disease in rats. World J Gastroenterol. 2006;12:1905–11.[Google Scholar]
  • 111. de Oliveira CP, Stefano JT, de Lima VM, de Sa SV, Simplicio FI, de Mello ES, Correa-Giannella ML, Alves VA, Laurindo FR, de Oliveira MG, Giannella-Neto D, Carrilho FJHepatic gene expression profile associated with non-alcoholic steatohepatitis protection by S-nitroso-N-acetylcysteine in ob/ob mice. J Hepatol. 2006;45:725–33.[PubMed][Google Scholar]
  • 112. Lang JD, Jr, Teng X, Chumley P, Crawford JH, Isbell TS, Chacko BK, Liu Y, Jhala N, Crowe DR, Smith AB, Cross RC, Frenette L, Kelley EE, Wilhite DW, Hall CR, Page GP, Fallon MB, Bynon JS, Eckhoff DE, Patel RPInhaled NO accelerates restoration of liver function in adults following orthotopic liver transplantation. J Clin Invest. 2007;117:2583–91.[Google Scholar]
  • 113. Gladwin MT, Raat NJ, Shiva S, Dezfulian C, Hogg N, Kim-Shapiro DB, Patel RPNitrite as a vascular endocrine nitric oxide reservoir that contributes to hypoxic signaling, cytoprotection, and vasodilation. Am J Physiol Heart Circ Physiol. 2006;291:H2026–35.[PubMed][Google Scholar]
  • 114. Duranski MR, Greer JJ, Dejam A, Jaganmohan S, Hogg N, Langston W, Patel RP, Yet SF, Wang X, Kevil CG, Gladwin MT, Lefer DJCytoprotective effects of nitrite during in vivo ischemia-reperfusion of the heart and liver. J Clin Invest. 2005;115:1232–40.[Google Scholar]
  • 115. Karaa A, Kamoun WS, Clemens MGChronic ethanol sensitizes the liver to endotoxin via effects on endothelial nitric oxide synthase regulation. Shock. 2005;24:447–54.[PubMed][Google Scholar]
  • 116. Wang X, Abdel-Rahman AAEffect of chronic ethanol administration on hepatic eNOS activity and its association with caveolin-1 and calmodulin in female rats. Am J Physiol Gastrointest Liver Physiol. 2005;289:G579–85.[PubMed][Google Scholar]
  • 117. Nanji AA, Greenberg SS, Tahan SR, Fogt F, Loscalzo J, Hossein Sadrzadeh SM, Xie J, Stamler JSNitric oxide production in experimental alcoholic liver disease: role in protection from injury. Gastroenterology. 1995;109:899–907.[PubMed][Google Scholar]
  • 118. Nanji AA, Jokelainen K, Lau GK, Rahemtulla A, Tipoe GL, Polavarapu R, Lalani ENArginine reverses ethanol-induced inflammatory and fibrotic changes in liver despite continued ethanol administration. J Pharmacol Exp Ther. 2001;299:832–9.[PubMed][Google Scholar]
  • 119. Haynes V, Elfering S, Traaseth N, Giulivi CMitochondrial nitric-oxide synthase: enzyme expression, characterization, and regulation. J Bioenerg Biomembr. 2004;36:341–6.[PubMed][Google Scholar]
  • 120. Haynes V, Elfering SL, Squires RJ, Traaseth N, Solien J, Ettl A, Giulivi CMitochondrial nitric-oxide synthase: role in pathophysiology. IUBMB Life. 2003;55:599–603.[PubMed][Google Scholar]
  • 121. Shiva S, Huang Z, Grubina R, Sun J, Ringwood LA, MacArthur PH, Xu X, Murphy E, Darley-Usmar VM, Gladwin MTDeoxymyoglobin is a nitrite reductase that generates nitric oxide and regulates mitochondrial respiration. Circ Res. 2007;100:654–61.[PubMed][Google Scholar]
  • 122. Wilfred de Alwis NM, Day CPGenetics of alcoholic liver disease and nonalcoholic fatty liver disease. Semin Liver Dis. 2007;27:44–54.[PubMed][Google Scholar]
  • 123. Stickel F, Osterreicher CHThe role of genetic polymorphisms in alcoholic liver disease. Alcohol Alcohol. 2006;41:209–24.[PubMed][Google Scholar]
  • 124. Zintzaras E, Stefanidis I, Santos M, Vidal FDo alcohol-metabolizing enzyme gene polymorphisms increase the risk of alcoholism and alcoholic liver disease? Hepatology. 2006;43:352–61.[PubMed][Google Scholar]
  • 125. Degoul F, Sutton A, Mansouri A, Cepanec C, Degott C, Fromenty B, Beaugrand M, Valla D, Pessayre DHomozygosity for alanine in the mitochondrial targeting sequence of superoxide dismutase and risk for severe alcoholic liver disease. Gastroenterology. 2001;120:1468–74.[PubMed][Google Scholar]
  • 126. Nahon P, Sutton A, Pessayre D, Rufat P, Degoul F, Ganne-Carrie N, Ziol M, Charnaux N, N’Kontchou G, Trinchet JC, Gattegno L, Beaugrand MGenetic dimorphism in superoxide dismutase and susceptibility to alcoholic cirrhosis, hepatocellular carcinoma, and death. Clin Gastroenterol Hepatol. 2005;3:292–8.[PubMed][Google Scholar]
  • 127. Sutton A, Imbert A, Igoudjil A, Descatoire V, Cazanave S, Pessayre D, Degoul FThe manganese superoxide dismutase Ala16Val dimorphism modulates both mitochondrial import and mRNA stability. Pharmacogenet Genomics. 2005;15:311–9.[PubMed][Google Scholar]
  • 128. Sutton A, Khoury H, Prip-Buus C, Cepanec C, Pessayre D, Degoul FThe Ala16Val genetic dimorphism modulates the import of human manganese superoxide dismutase into rat liver mitochondria. Pharmacogenetics. 2003;13:145–57.[PubMed][Google Scholar]
  • 129. Sutton A, Nahon P, Pessayre D, Rufat P, Poire A, Ziol M, Vidaud D, Barget N, Ganne-Carrie N, Charnaux N, Trinchet JC, Gattegno L, Beaugrand MGenetic polymorphisms in antioxidant enzymes modulate hepatic iron accumulation and hepatocellular carcinoma development in patients with alcohol-induced cirrhosis. Cancer Res. 2006;66:2844–52.[PubMed][Google Scholar]
  • 130. Koch O, Farre S, De Leo ME, Palozza P, Palazzotti B, Borrelo S, Palombini G, Cravero A, Galeotti TRegulation of manganese superoxide dismutase (MnSOD) in chronic experimental alcoholism: effects of vitamin E-supplemented and -deficient diets. Alcohol Alcohol. 2000;35:159–63.[PubMed][Google Scholar]
  • 131. Koch OR, De Leo ME, Borrello S, Palombini G, Galeotti TEthanol treatment up-regulates the expression of mitochondrial manganese superoxide dismutase in rat liver. Biochem Biophys Res Commun. 1994;201:1356–65.[PubMed][Google Scholar]
  • 132. Stewart SF, Leathart JB, Chen Y, Daly AK, Rolla R, Vay D, Mottaran E, Vidali M, Albano E, Day CPValine-alanine manganese superoxide dismutase polymorphism is not associated with alcohol-induced oxidative stress or liver fibrosis. Hepatology. 2002;36:1355–60.[PubMed][Google Scholar]
  • 133. Namikawa C, Shu-Ping Z, Vyselaar JR, Nozaki Y, Nemoto Y, Ono M, Akisawa N, Saibara T, Hiroi M, Enzan H, Onishi SPolymorphisms of microsomal triglyceride transfer protein gene and manganese superoxide dismutase gene in non-alcoholic steatohepatitis. J Hepatol. 2004;40:781–6.[PubMed][Google Scholar]
  • 134. Hines IN, Wheeler MD. Recent advances in alcoholic liver disease III. Role of the innate immune response in alcoholic hepatitis. Am J Physiol Gastrointest Liver Physiol. 2004;287:G310–4.[PubMed]
  • 135. Thurman RG. Mechanisms of hepatic toxicity II. Alcoholic liver injury involves activation of Kupffer cells by endotoxin. Am J Physiol Gastrointest Liver Physiol. 1998;275:G605–G11.[PubMed]
  • 136. Jarvelainen HA, Orpana A, Perola M, Savolainen VT, Karhunen PJ, Lindros KOPromoter polymorphism of the CD14 endotoxin receptor gene as a risk factor for alcoholic liver disease. Hepatology. 2001;33:1148–53.[PubMed][Google Scholar]
  • 137. Day CPCD14 promoter polymorphism associated with risk of NASH. J Hepatol. 2002;36:21.[PubMed][Google Scholar]
  • 138. Baldini M, Lohman IC, Halonen M, Erickson RP, Holt PG, Martinez FDA Polymorphism* in the 5′ flanking region of the CD14 gene is associated with circulating soluble CD14 levels and with total serum immunoglobulin E. Am J Respir Cell Mol Biol. 1999;20:976–83.[PubMed][Google Scholar]
  • 139. Leathart JB, Day CP, Daly AKNo association between functional SNP’s in the endotoxin receptors CD14 and TLR4 and alcoholic liver disease (ALD): is endotoxin important in the pathogenesis of ALD in humans? Hepatology. 2001;34:459A.[PubMed][Google Scholar]
  • 140. Martins A, Cortez-Pinto H, Machado M, Goncalves MS, Soren S, Marques-Vidal P, de Moura MC, Camilo MEAre genetic polymorphisms of tumour necrosis factor alpha, interleukin-10, CD14 endotoxin receptor or manganese superoxide dismutase associated with alcoholic liver disease? Eur J Gastroenterol Hepatol. 2005;17:1099–104.[PubMed][Google Scholar]
  • 141. Dixon JB, Bhathal PS, Jonsson JR, Dixon AF, Powell EE, O’Brien PEPro-fibrotic polymorphisms predictive of advanced liver fibrosis in the severely obese. J Hepatol. 2003;39:967–71.[PubMed][Google Scholar]
  • 142. Pravenec M, Hyakukoku M, Houstek J, Zidek V, Landa V, Mlejnek P, Miksik I, Dudova-Mothejzikova K, Pecina P, Vrbacky M, Drahota Z, Vojtiskova A, Mracek T, Kazdova L, Oliyarnyk O, Wang J, Ho C, Qi N, Sugimoto K, Kurtz TDirect linkage of mitochondrial genome variation to risk factors for type 2 diabetes in conplastic strains. Genome Res. 2007;17:1319–26.[Google Scholar]
  • 143. Wallace DCA mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev Genet. 2005;39:359–407.[Google Scholar]
  • 144. Tome S, Lucey MRReview article: current management of alcoholic liver disease. Aliment Pharmacol Ther. 2004;19:707–14.[PubMed][Google Scholar]
  • 145. Bergheim I, Guo L, Davis MA, Lambert JC, Beier JI, Duveau I, Luyendyk JP, Roth RA, Arteel GEMetformin prevents alcohol-induced liver injury in the mouse: Critical role of plasminogen activator inhibitor-1. Gastroenterology. 2006;130:2099–112.[Google Scholar]
  • 146. Hernandez R, Teruel T, de Alvaro C, Lorenzo MRosiglitazone ameliorates insulin resistance in brown adipocytes of Wistar rats by impairing TNF-alpha induction of p38 and p42/p44 mitogen-activated protein kinases. Diabetologia. 2004;47:1615–24.[PubMed][Google Scholar]
  • 147. Iwata M, Haruta T, Usui I, Takata Y, Takano A, Uno T, Kawahara J, Ueno E, Sasaoka T, Ishibashi O, Kobayashi MPioglitazone ameliorates tumor necrosis factor-alpha-induced insulin resistance by a mechanism independent of adipogenic activity of peroxisome proliferator--activated receptor-gamma. Diabetes. 2001;50:1083–92.[PubMed][Google Scholar]
  • 148. Murase K, Odaka H, Suzuki M, Tayuki N, Ikeda HPioglitazone time-dependently reduces tumour necrosis factor-alpha level in muscle and improves metabolic abnormalities in Wistar fatty rats. Diabetologia. 1998;41:257–64.[PubMed][Google Scholar]
  • 149. Leclercq IA, Lebrun VA, Starkel P, Horsmans YJIntrahepatic insulin resistance in a murine model of steatohepatitis: effect of PPARgamma agonist pioglitazone. Lab Invest. 2007;87:56–65.[PubMed][Google Scholar]
  • 150. Tahan V, Eren F, Avsar E, Yavuz D, Yuksel M, Emekli E, Imeryuz N, Celikel C, Uzun H, Haklar G, Tozun NRosiglitazone Attenuates Liver Inflammation in a Rat Model of Nonalcoholic Steatohepatitis. Dig Dis Sci. 2007[PubMed][Google Scholar]
  • 151. Tomita K, Azuma T, Kitamura N, Nishida J, Tamiya G, Oka A, Inokuchi S, Nishimura T, Suematsu M, Ishii HPioglitazone prevents alcohol-induced fatty liver in rats through up-regulation of c-Met. Gastroenterology. 2004;126:873–85.[PubMed][Google Scholar]
  • 152. Fischer M, You M, Matsumoto M, Crabb DWPeroxisome proliferator-activated receptor alpha (PPARalpha) agonist treatment reverses PPARalpha dysfunction and abnormalities in hepatic lipid metabolism in ethanol-fed mice. J Biol Chem. 2003;278:27997–8004.[PubMed][Google Scholar]
  • 153. Garcia-Ruiz I, Rodriguez-Juan C, Diaz-Sanjuan T, Martinez MA, Munoz-Yague T, Solis-Herruzo JAEffects of rosiglitazone on the liver histology and mitochondrial function in ob/ob mice. Hepatology. 2007;46:414–23.[PubMed][Google Scholar]
  • 154. Caldwell SH, Patrie JT, Brunt EM, Redick JA, Davis CA, Park SH, Neuschwander-Tetri BAThe effects of 48 weeks of rosiglitazone on hepatocyte mitochondria in human nonalcoholic steatohepatitis. Hepatology. 2007[PubMed][Google Scholar]
  • 155. Harrison SA, Torgerson S, Hayashi P, Ward J, Schenker SVitamin E and vitamin C treatment improves fibrosis in patients with nonalcoholic steatohepatitis. Am J Gastroenterol. 2003;98:2485–90.[PubMed][Google Scholar]
  • 156. Hasegawa T, Yoneda M, Nakamura K, Makino I, Terano APlasma transforming growth factor-beta1 level and efficacy of alpha-tocopherol in patients with non-alcoholic steatohepatitis: a pilot study. Aliment Pharmacol Ther. 2001;15:1667–72.[PubMed][Google Scholar]
  • 157. Lavine JEVitamin E treatment of nonalcoholic steatohepatitis in children: a pilot study. J Pediatr. 2000;136:734–8.[PubMed][Google Scholar]
  • 158. Sanyal AJ, Mofrad PS, Contos MJ, Sargeant C, Luketic VA, Sterling RK, Stravitz RT, Shiffman ML, Clore J, Mills ASA pilot study of vitamin E versus vitamin E and pioglitazone for the treatment of nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2004;2:1107–15.[PubMed][Google Scholar]
  • 159. Devaraj S, Jialal IFailure of vitamin E in clinical trials: is gamma-tocopherol the answer? Nutr Rev. 2005;63:290–3.[PubMed][Google Scholar]
  • 160. Jiang Q, Ames BNGamma-tocopherol, but not alpha-tocopherol, decreases proinflammatory eicosanoids and inflammation damage in rats. FASEB J. 2003;17:816–22.[PubMed][Google Scholar]
  • 161. Brown SE, Ross MF, Sanjuan-Pla A, Manas AR, Smith RA, Murphy MPTargeting lipoic acid to mitochondria: synthesis and characterization of a triphenylphosphonium-conjugated alpha-lipoyl derivative. Free Radic Biol Med. 2007;42:1766–80.[PubMed][Google Scholar]
  • 162. Cocheme HM, Kelso GF, James AM, Ross MF, Trnka J, Mahendiran T, Asin-Cayuela J, Blaikie FH, Manas AR, Porteous CM, Adlam VJ, Smith RA, Murphy MPMitochondrial targeting of quinones: therapeutic implications. Mitochondrion. 2007;7 Suppl:S94–102.[PubMed][Google Scholar]
  • 163. Murphy MP, Smith RATargeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu Rev Pharmacol Toxicol. 2007;47:629–56.[PubMed][Google Scholar]
  • 164. Adlam VJ, Harrison JC, Porteous CM, James AM, Smith RA, Murphy MP, Sammut IATargeting an antioxidant to mitochondria decreases cardiac ischemia-reperfusion injury. FASEB J. 2005;19:1088–95.[PubMed][Google Scholar]
  • 165. Covey MV, Murphy MP, Hobbs CE, Smith RA, Oorschot DEEffect of the mitochondrial antioxidant, Mito Vitamin E, on hypoxic-ischemic striatal injury in neonatal rats: a dose-response and stereological study. Exp Neurol. 2006;199:513–9.[PubMed][Google Scholar]
  • 166. Ronis MJ, Butura A, Sampey BP, Shankar K, Prior RL, Korourian S, Albano E, Ingelman-Sundberg M, Petersen DR, Badger TMEffects of N-acetylcysteine on ethanol-induced hepatotoxicity in rats fed via total enteral nutrition. Free Radic Biol Med. 2005;39:619–30.[Google Scholar]
  • 167. Kang YJ, Zhou ZZinc prevention and treatment of alcoholic liver disease. Mol Aspects Med. 2005;26:391–404.[PubMed][Google Scholar]
  • 168. Zhou Z, Wang L, Song Z, Saari JT, McClain CJ, Kang YJZinc supplementation prevents alcoholic liver injury in mice through attenuation of oxidative stress. Am J Pathol. 2005;166:1681–90.[Google Scholar]
  • 169. Song Z, Deaciuc I, Song M, Lee DY, Liu Y, Ji X, McClain CSilymarin protects against acute ethanol-induced hepatotoxicity in mice. Alcohol Clin Exp Res. 2006;30:407–13.[Google Scholar]
  • 170. Lindor KD, Kowdley KV, Heathcote EJ, Harrison ME, Jorgensen R, Angulo P, Lymp JF, Burgart L, Colin PUrsodeoxycholic acid for treatment of nonalcoholic steatohepatitis: results of a randomized trial. Hepatology. 2004;39:770–8.[PubMed][Google Scholar]
  • 171. Cave M, Deaciuc I, Mendez C, Song Z, Joshi-Barve S, Barve S, McClain CNonalcoholic fatty liver disease: predisposing factors and the role of nutrition. J Nutr Biochem. 2007;18:184–95.[PubMed][Google Scholar]
  • 172. Comar KM, Sterling RKReview article: Drug therapy for non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2006;23:207–15.[PubMed][Google Scholar]
  • 173. Federico A, Trappoliere M, Loguercio CTreatment of patients with non-alcoholic fatty liver disease: current views and perspectives. Dig Liver Dis. 2006;38:789–801.[PubMed][Google Scholar]
  • 174. Reuben AAlcohol and the liver. Curr Opin Gastroenterol. 2007;23:283–91.[PubMed][Google Scholar]
  • 175. Mato JM, Alvarez L, Ortiz P, Pajares MAS-adenosylmethionine synthesis: molecular mechanisms and clinical implications. Pharmacol Ther. 1997;73:265–80.[PubMed][Google Scholar]
  • 176. Lu SC, Huang ZZ, Yang H, Mato JM, Avila MA, Tsukamoto HChanges in methionine adenosyltransferase and S-adenosylmethionine homeostasis in alcoholic rat liver. Am J Physiol Gastrointest Liver Physiol. 2000;279:G178–85.[PubMed][Google Scholar]
  • 177. Barak AJ, Beckenhauer HC, Tuma DJ. Methionine synthase. a possible prime site of the ethanolic lesion in liver. Alcohol. 2002;26:65–7.[PubMed]
  • 178. Perez-Mato I, Castro C, Ruiz FA, Corrales FJ, Mato JMMethionine adenosyltransferase S-nitrosylation is regulated by the basic and acidic amino acids surrounding the target thiol. J Biol Chem. 1999;274:17075–9.[PubMed][Google Scholar]
  • 179. Ruiz F, Corrales FJ, Miqueo C, Mato JMNitric oxide inactivates rat hepatic methionine adenosyltransferase In vivo by S-nitrosylation. Hepatology. 1998;28:1051–7.[PubMed][Google Scholar]
  • 180. Lieber CSS-adenosyl-L-methionine: its role in the treatment of liver disorders. Am J Clin Nutr. 2002;76(suppl):1183S–7S.[PubMed][Google Scholar]
  • 181. Garcia-Ruiz C, Colell A, Morales A, Kaplowitz N, Fernandez-Checa JCRole of oxidative stress generated from the mitochondrial electron transport chain and mitochondrial glutahione status in loss of mitochondrial function and activation of transcription factor nuclear factor-kB: studies with isolated mitochondria and rat hepatocytes. Mol Pharmacol. 1995;48:825–34.[PubMed][Google Scholar]
  • 182. Lieber CS, DeCarli LM, Mak KM, Kim CI, Leo MAAttenuation of alcohol-induced hepatic fibrosis by polyunsaturated lecithin. Hepatology. 1990;12:1390–8.[PubMed][Google Scholar]
  • 183. Mato JM, Camara J, Fernandez de Paz J, Caballeria L, Coll S, Caballero A, Garcia-Buey L, Beltran J, Benita V, Caballeria J, Sola R, Moreno-Otero R, Barrao F, Martin-Duce A, Correa JA, Pares A, Barrao E, Garcia-Magaz I, Puerta JL, Moreno J, Boissard G, Ortiz P, Rodes JS-adenosylmethionine in alcoholic liver cirrhosis: a randomized, placebo-controlled, double-blind, multicenter clinical trial. J Hepatol. 1999;30:1081–9.[PubMed][Google Scholar]
  • 184. Lu SC, Alvarez L, Huang ZZ, Chen L, An W, Corrales FJ, Avila MA, Kanel G, Mato JMMethionine adenosyltransferase 1A knockout mice are predisposed to liver injury and exhibit increased expression of genes involved in proliferation. Proc Natl Acad Sci U S A. 2001;98:5560–5.[Google Scholar]
  • 185. Martinez-Chantar ML, Corrales FJ, Martinez-Cruz LA, Garcia-Trevijano ER, Huang ZZ, Chen L, Kanel G, Avila MA, Mato JM, Lu SCSpontaneous oxidative stress and liver tumors in mice lacking methionine adenosyltransferase 1A. FASEB J. 2002;16:1292–4.[PubMed][Google Scholar]
  • 186. Santamaria E, Avila MA, Latasa MU, Rubio A, Martin-Duce A, Lu SC, Mato JM, Corrales FJFunctional proteomics of nonalcoholic steatohepatitis: mitochondrial proteins as targets of S-adenosylmethionine. Proc Natl Acad Sci U S A. 2003;100:3065–70.[Google Scholar]
  • 187. Song Z, Zhou Z, Song M, Uriarte S, Chen T, Deaciuc I, McClain CJAlcohol-induced S-adenosylhomocysteine accumulation in the liver sensitizes to TNF hepatotoxicity: possible involvement of mitochondrial S-adenosylmethionine transport. Biochem Pharmacol. 2007;74:521–31.[Google Scholar]
  • 188. Ji C, Kaplowitz NBetaine decreases hyperhomocysteinemia, endoplasmic reticulum stress, and liver injury in alcohol-fed mice. Gastroenterology. 2003;124:1488–99.[PubMed][Google Scholar]
  • 189. Kharbanda KK, Mailliard ME, Baldwin CR, Beckenhauer HC, Sorrell MF, Tuma DJBetaine attenuates alcoholic steatosis by restoring phosphatidylcholine generation via the phosphatidylethanolamine methyltransferase pathway. J Hepatol. 2007;46:314–21.[PubMed][Google Scholar]
  • 190. Song Z, Deaciuc I, Zhou Z, Song M, Chen T, Hill D, McClain CJInvolvement of AMP-activated Protein Kinase in Beneficial Effects of Betaine on High-Sucrose Diet-Induced Hepatic Steatosis. Am J Physiol Gastrointest Liver Physiol. 2007[Google Scholar]
  • 191. Abdelmalek MF, Angulo P, Jorgensen RA, Sylvestre PB, Lindor KDBetaine, a promising new agent for patients with nonalcoholic steatohepatitis: results of a pilot study. Am J Gastroenterol. 2001;96:2711–7.[PubMed][Google Scholar]
  • 192. Mato JM, Lu SCHomocysteine, the bad thiol. Hepatology. 2005;41:976–9.[PubMed][Google Scholar]
  • 193. Craig SABetaine in human nutrition. Am J Clin Nutr. 2004;80:539–49.[PubMed][Google Scholar]
  • 194. Werstuck GH, Lentz SR, Dayal S, Hossain GS, Sood SK, Shi YY, Zhou J, Maeda N, Krisans SK, Malinow MR, Austin RCHomocysteine-induced endoplasmic reticulum stress causes dysregulation of the cholesterol and triglyceride biosynthetic pathways. J Clin Invest. 2001;107:1263–73.[Google Scholar]
  • 195. Gulsen M, Yesilova Z, Bagci S, Uygun A, Ozcan A, Ercin CN, Erdil A, Sanisoglu SY, Cakir E, Ates Y, Erbil MK, Karaeren N, Dagalp KElevated plasma homocysteine concentrations as a predictor of steatohepatitis in patients with non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 2005;20:1448–55.[PubMed][Google Scholar]
  • 196. King AL, Eccleston HE, Andringa KK, Tuma DJ, Kharbanda KK, Bailey SMS-Adenosylmethionine and betaine prevent chronic alcohol associated losses in hepatic mitochondria respiratory complex content. Free Radic Biol Med. 2006;41:S23.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.