Skip to main content
Journal of Clinical and Translational Research logoLink to Journal of Clinical and Translational Research
. 2017 Feb 12;3(1):212–232. doi: 10.18053/jctres.03.2017S1.006

Role of nonalcoholic fatty liver disease as risk factor for drug-induced hepatotoxicity

Julie Massart 1,, Karima Begriche 2,, Caroline Moreau 2,3, Bernard Fromenty 2,*
PMCID: PMC5500243  NIHMSID: NIHMS872033  PMID: 28691103

Abstract

Background: Obesity is often associated with nonalcoholic fatty liver disease (NAFLD), which refers to a large spectrum of hepatic lesions including fatty liver, nonalcoholic steatohepatitis (NASH) and cirrhosis. Different investigations showed or suggested that obesity and NAFLD are able to increase the risk of hepatotoxicity of different drugs. Some of these drugs could induce more frequently an acute hepatitis in obese individuals whereas others could worsen pre-existing NAFLD.

Aim: The main objective of the present review was to collect the available information regarding the role of NAFLD as risk factor for drug-induced hepatotoxicity. For this purpose, we performed a data-mining analysis using different queries including drug-induced liver injury (or DILI), drug-induced hepatotoxicity, fatty liver, nonalcoholic fatty liver disease (or NAFLD), steatosis and obesity. The main data from the collected articles are reported in this review and when available, some pathophysiological hypotheses are put forward.

Relevance for patients: Drugs that could pose a potential risk in obese patients include compounds belonging to different pharmacological classes such as acetaminophen, halothane, methotrexate, rosiglitazone, stavudine and tamoxifen. For some of these drugs, experimental investigations in obese rodents confirmed the clinical observations and unveiled different pathophysiological mechanisms which could explain why these pharmaceuticals are particularly hepatotoxic in obesity and NAFLD. Other drugs such as pentoxifyl-line, phenobarbital and omeprazole might also pose a risk but more investigations are required to determine whether this risk is significant or not. Because obese people often take several drugs for the treatment of different obesity-related diseases such as type 2 diabetes, hyperlipidemia and coronary heart disease, it is urgent to identify the main pharmaceuticals that can cause acute hepatitis on a fatty liver background or induce NAFLD worsening.

Keywords: obesity, nonalcoholic fatty liver disease, drug, liver, toxicity, drug-induced liver injury, hepatotoxicity, cytochrome P450, mitochondria, acetaminophen, halothane, isoflurane, methotrexate, tamoxifen, pentoxifylline, rosiglitazone, stavudine

1. Introduction

It is currently estimated that more than 350 drugs of the modern pharmacopoeia can induce liver injury with different clinical presentations such as hepatic cytolysis and cholestasis [1,2]. Although severe drug-induced liver injury (DILI) is a rather rare event, it can require orthotopic liver transplantation, or lead to the death of the patient [2,3]. In addition, DILI can result in the withdrawal of drugs from the market, thus causing massive financial losses [4]. Thus, DILI is a major issue for public health and pharmaceutical companies.

Besides drugs characteristics such as daily dose, chemical structure and liver metabolism, different predisposing factors inherent to the patients are known to enhance the risk of DILI [1,5]. Such factors include for instance polymorphisms in genes coding for xenobiotic-metabolizing enzymes (XMEs), underlying genetic mitochondrial diseases and pre-existing liver diseases such as alcoholic liver diseases and viral hepatitis [4,6]. Another liver disease that is increasingly recognized to favor DILI is nonalcoholic fatty liver disease (NAFLD), at least for some drugs such as acetaminophen, halothane, some antiretroviral drugs and methotrexate [7,8]. The main objective of the present review is to present the available information regarding the role of NAFLD as risk factor for drug-induced hepatotoxicity. For this purpose, we performed a PubMed search of literature published in the English language using the following terms: "drug-induced liver injury", "drug-induced hepatotoxicity", "fatty liver", "nonalcoholic fatty liver disease", "steatosis" and "obesity". This data-mining analysis was also completed by using Google Scholar®. Before presenting the collected data concerning the present topic, some major features of NAFLD pathophysiology will be briefly recalled. Such information is important to understand why this frequent liver disease can enhance the risk of hepatotoxicity of some drugs.

2. Pathophysiology of NAFLD

NAFLD refers to a large spectrum of hepatic lesions linked to obesity including fatty liver, nonalcoholic steatohepatitis (NASH) and cirrhosis. NASH is characterized not only by fatty liver (also referred to as hepatic steatosis) but also by necroinflammation, some fibrosis and the presence of apoptotic hepatocytes. Importantly, most obese patients present simple fatty liver but this lesion progresses in the long term to NASH in only 10 to 20% of patients. Several genetic polymorphisms could explain, at least in part, why fatty liver progresses to NASH only in a subset of obese patients [9,10].

There is now strong evidence that insulin resistance (IR) in skeletal muscle and white adipose tissue (WAT) plays a key role in the pathogenesis of fatty liver linked to obesity [9,11]. IR in muscle is characterized by impaired glucose uptake and glycogen synthesis, thus favoring glucose utilization for hepatic de novo lipogenesis (DNL), while IR in WAT favors triacylglycerol (TAG) lipolysis. This uncontrolled lipolysis leads to the release in the circulation of large amounts of non-esterified fatty acids (NEFAs), which enter the liver in a concentration-dependent manner. In addition, NEFAs are synthesized more actively in liver because IR-associated hyperinsulinemia favors hepatic DNL. Indeed, high plasma insulin concentrations increase hepatic levels of sterol regulatory elementbinding protein 1c (SREBP1c) and peroxisome proliferator-activated receptor γ (PPARγ), two transcription factors controlling the expression of key enzymes involved in NEFA and TAG synthesis. When type 2 diabetes (T2D) occurs in obese individuals, hyperglycemia can also contributes to fatty liver by activating carbohydrate responsive element-binding protein (ChREBP), a transcription factor that enhances the expression of several glycolytic and lipogenic enzymes [9,12].

The pathogenesis of the progression of fatty liver to NASH is complex and could involve different events including overproduction of reactive oxygen species (ROS), reduced ROS detoxification, mitochondrial dysfunction, endoplasmic reticulum (ER) stress and increased release of pro-inflammatory and profibrogenic cytokines [9,11]. Some of these events could result from the direct toxicity of lipid derivatives (also referred to as lipotoxicity) on different metabolic pathways and hepatocellular constituents [9,13]. Notably, ROS overproduction during NAFLD could mainly occur within mitochondria, in particular at the level of complexes I and III of the mitochondrial respiratory chain (MRC), and of some enzymes of the fatty acid oxidation (FAO) pathway [9,14,15]. Another significant source of ROS in fatty liver could be the cytochrome P450 2E1 (CYP2E1), a XME located not only within the ER but also within mitochondria [16,17]. Thus, several mitochondrial enzymes are probably involved in ROS overproduction during NAFLD.

Numerous studies carried out in rodents and humans have reported higher hepatic CYP2E1 expression and/or activity in obesity and NAFLD [18-21]. A role of enhanced CYP2E1 activity in NAFLD pathogenesis is supported by different experimental investigations [18,22,23]. It is still unclear why CYP2E1 activity is increased in obesity and NAFLD, even though some studies suggested the role of disturbed intra-hepatic insulin signalling and/or accumulation of some endogenous derivatives such as ketone bodies and saturated fatty acids [18,20,24]. The activity of other CYPs such as CYP2C9 and CYP2D6 has also been reported to be enhanced during obesity and NAFLD [7,25]. However, higher CYP2C9 and CYP2D6 activity has most probably no significant role in NAFLD progression because these CYP isoforms do not produce ROS during their catalytic cycles, in contrast to CYP2E1 [18]. Contrary to these CYPs, hepatic CYP3A4 expression and activity are lower during obesity and NAFLD [24-26]. This might disturb the pharmacologic and safety profiles of some of the numerous drugs metabolized by this enzyme [27].

3. NAFLD and drug-induced hepatotoxicity

There is growing evidence that NAFLD can increase the risk and/or the severity of liver injury induced by different drugs. Practitioners should bear this information in mind because obese patients are consuming on average more drugs than non-obese individuals [7,28]. Indeed, obesity is often associated with different diseases such as T2D, hyperlipidemia, coronary heart disease and osteoarthritis that need to be treated. Moreover, obese patients can suffer from chronic diseases not necessarily related to obesity such as viral infections (i.e. HIV and HCV) and cancers. Notably, treatment of the afore-mentioned diseases requires long-term drug administration, which can enhance the risk of adverse effects including hepatotoxicity [29,30].

Obese patients are not only more likely to be exposed to different types of pharmaceuticals, but their diseased liver is also more vulnerable to some toxicological insults. Actually, hepatotoxicity in obese patients with NAFLD could occur as two distinct clinical settings [7]. For some drugs, an acute hepatitis could happen more frequently and could be more severe than for healthy patients. For some other drugs, an aggravation of the pre-existing NAFLD could occur, with for instance an accelerated transition from fatty liver to NASH.

3.1. Acute hepatitis in NAFLD

Numerous drugs can induce acute hepatitis [31,32], which can be sometimes severe and require the patient’s hospitalization. Indeed, acute cytolysis can involve an important proportion of hepatocytes, thus leading to hepatic failure that requires orthotopic liver transplantation [33,34]. Notably, it has been reported that subjects with drug-induced acute liver failure (ALF) were on average overweight but the body mass index (BMI) did not predict poor ALF outcome [33]. However, this study did not determine whether high BMI predicted ALF induced by some specific drugs. Actually, higher risk of acute hepatitis in obesity and NAFLD has been documented (or suspected) only with a few drugs, as listed in Table 1 and below. However, one can expect that this list will grow in the future because practitioners and researchers are increasingly aware about this issue.

Table 1. Drugs and acute hepatitis in obesity and nonalcoholic fatty liver disease.

Drug(s) Pharmacological class Comments
Acetaminophen (APAP) Analgesic and antipyretic Several experimental and clinical studies strongly suggest that acute APAP hepatotoxicity could be more frequent and more severe in obesity and NAFLD [8,49,50,54]. This is probably because these diseases are often associated with higher hepatic CYP2E1 activity, which generates greater levels of the toxic NAPQI metabolite. However, some obese individuals do not present high CYP2E1 activity and thus they may not be at risk for APAP-induced liver injury
Amiodarone Antiarrhythmic and antianginal One clinical study reported that the frequency of amiodarone-induced hepatotoxicity did not appear to be greater in patients suffering from the metabolic syndrome [97]. Further investigations would be needed in order to confirm this observation
Fosinopril Antihypertensive Only one study reported an increased risk of fasinopril-induced hepatitis in patients with NAFLD [94]. The exact mechanism of this greater risk is currently unknown. Further investigations would be needed to confirm this clinical observation
Halothane and isoflurane Volatile anesthetics Several clinical studies strongly suggest that obese patients could be particularly at risk for halothane and isoflurane-induced acute hepatotoxicity [18,59-63]. This is probably because obesity is often associated with higher hepatic CYP2E1 activity, which generates greater levels of the toxic acyl chloride metabolites. However, some obese individuals do not present high CYP2E1 activity and thus they may not be at risk for halothane and isoflurane-induced liver injury
Losartan Antihypertensive Only one study reported an increased risk of losartan-induced hepatitis in patients with NAFLD [94]. The exact mechanism of this greater risk is currently unknown. Further investigations would be needed to confirm this clinical observation
Methotrexate anticancer and anti-inflammatory (anti-folate) Although several clinical studies suggest that methotrexate could induce acute hepatitis more frequently in obese patients, this drug is mostly suspected to aggravate NAFLD (see Table 2)
Omeprazole Antiulcer (proton pump inhibitor) Only one study reported an increased risk of omeprazole-induced hepatitis in patients with NAFLD [94]. The exact mechanism of this greater risk is currently unknown. Further investigations would be needed to confirm this clinical observation
Piperacillin/tazobactam Antibiotics Only one study reported an increased risk of piperacillin/tazobactam-induced hepatitis in patients with NAFLD [94]. The exact mechanism of this greater risk is currently unknown. Further investigations would be needed to confirm this clinical observation
Statins Hypolipidemic drugs Several clinical studies suggested that the risk of statin-induced hepatotoxicity is not higher in patients with NAFLD [95,96].
Telithromycin Antibiotic Only one study reported an increased risk of telithromycin-induced hepatitis in patients with NAFLD [94]. The exact mechanism of this greater risk is currently unknown. Further investigations would be needed to confirm this clinical observation
Ticlopidine Antithrombotic (inhibitor of platelet aggregation) Only one study reported an increased risk of ticlopidine-induced hepatitis in patients with NAFLD [94]. The exact mechanism of this greater risk is currently unknown. Further investigations would be needed to confirm this clinical observation

Theoretically, drug-induced acute hepatitis could occur more frequently in NAFLD because drugs are expected to be more cytotoxic in this new metabolic environment characterized by reduced ATP synthesis and higher ROS levels (Figure 1) [9,35,36]. In other words, for the same drug exposure, fatty liver will be more likely to be damaged as compared to normal liver. During NAFLD, higher ROS levels can be the consequence of both ROS overproduction by different cellular components (respiratory chain, CYP2E1 and peroxisomes) and reduced antioxidant defenses (glutathione, glutathione peroxidases, superoxide dismutases..) [9,37]. When NASH occurs, the pro-inflammatory environment with high tumor necrosis factor-α (TNFα) production could also sensitize the liver to drug-induced acute cytotoxicity (Figure 1) [38,39]. In addition to these mechanisms, drug cytotoxicity could be favored by other events such as the production of higher levels of CYP-generated toxic metabolites, as discussed below for acetaminophen and other pharmaceuticals. Finally, it must be pointed out that NAFLD might not increase the susceptibility of acute liver injury for all hepatotoxic drugs. This is most probably because the NAFLD-associated alteration of XME activities will not necessarily be associated with higher levels of the toxic compounds, which can be either the parent drugs and/or CYP-generated reactive metabolites.

Figure 1. Mechanisms whereby NASH could increase the susceptibility of drug-induced acute liver injury. NASH is associated with increased CYP2E1 expression and activity, reduced MRC activity and inflammation. These events lead to ROS overproduction, reduced ATP synthesis and increased production of pro-inflammatory cytokines such as TNFα, which can favor the occurrence of drug-induced acute liver injury.

Figure 1

3.1.1. Acetaminophen

In this section, we will briefly provide the most important information related to acetaminophen-induced liver injury in obesity and NAFLD, as we recently published a detailed review on the topic [8].

The painkiller acetaminophen (APAP), also known as paracetamol, is mainly metabolized in the liver into harmless glucuronide and sulfate conjugates. However, a small amount of APAP is oxidized to the highly toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI) by several cytochromes P450, in particular CYP2E1 and CYP3A4. Hepatic CYP2E1 activity is increased not only in NAFLD, as previously mentioned, but also in other pathophysiological states including alcoholic liver disease, malnutrition and diabetes [18,40]. Thus, these diseases are expected to favor the generation of higher intrahepatic levels of NAPQI (Figure 2).

Figure 2. Increased hepatotoxicity induced by acetaminophen and halothane in obesity and NAFLD. Obesity and NAFLD are associated with higher hepatic cytochrome P450 2E1 (CYP2E1) activity, which is responsible for a greater biotransformation of acetaminophen and halothane to the highly reactive metabolites N-acetyl-p-benzoquinone imine and trifluoroacetyl chloride, respectively. These reactive metabolites bind to glutathione (GSH) and key cellular targets, in particular at the mitochondrial level. These deleterious events induce mitochondrial dysfunction, reduced ATP synthesis and oxidative stress, thus triggering hepatic cytolysis.

Figure 2

Although NAPQI is usually safely detoxified by hepatic reduced glutathione (GSH) when APAP is taken at the maximum recommended dose (i.e. 4 grams per day), high levels of NAPQI after an acute overdose consistently induce a fall in the GSH stores, thus promoting the covalent binding of free NAPQI to different cellular proteins, in particular at the mitochondrial level. This leads to profound oxidative phosphorylation (OXPHOS) impairment and ATP shortage, massive hepatocellular necrosis and ALF [8,41]. Activation of c-jun N-terminal kinase (JNK) could also be involved in APAP-induced liver injury, although some studies did not support such pathological role [24,42].

Besides the context of APAP overdose, it is noteworthy that therapeutic dose of APAP can induce mild to moderate hepatic cytolysis [21,43], and even fulminant hepatitis in a few patients [44,45]. However, it is noteworthy that no well-controlled study has reported ALF in individuals treated with recommended doses of APAP. Hence, such rare therapeutic misadventures seem to occur preferentially in patients with predisposing factors such as malnutrition, co-medication with different drugs, alcoholic liver disease and NAFLD [8,44,46]. Some of these factors can not only enhance liver CYP2E1 activity, as previously recalled, but they can also significantly reduce the intrahepatic stores of GSH [8,47,48].

NAFLD could increase the risk of APAP-induced hepatotoxicity either after an overdose or at therapeutic dosage. However, this risk might not concern all obese patients but only a subset of obese patients, as discussed below. Two large retrospective studies reported that NAFLD could be a risk factor for APAP-induced acute liver injury (ALI) after APAP overdose [49,50]. In these studies, patients with pre-existent NAFLD and hospitalized for APAP overdose had a 4-to 7-fold higher prevalence of ALI as compared to those without NAFLD [49,50]. Interestingly, these studies showed that the risk of severe ALI after APAP overdose was increased about similarly with NAFLD and alcoholic liver disease [49,50]. A recent study reported that therapeutic doses of APAP significantly increased plasma transaminases (i.e. ALT and AST) in morbidly obese patients but not in non-obese individuals [21]. Increased levels of transaminases were more than three times the reference values in some obese patients [21]. Moreover, it has been reported a case of acute fulminant hepatitis in an obese female treated by the maximal recommended dose of APAP for 10 days [44]. In contrast to these studies, the susceptibility to APAP-induced ALI was reported to be similar, or even lower, in obese patients compared to non-obese individuals [51,52]. However, one of these studies showed that obese patients had significantly poorer outcomes after ALF [51]. Although all these clinical studies are very informative, it is noteworthy that some confounding factors were not taken into account, such as different psychological and genetic factors.

Several studies in obese rodents showed that obesity and NAFLD are actually not sufficient by themselves to favor APAP-induced hepatotoxicity [8,53,54]. In contrast, our investigations in ob/ob and db/db obese mice clearly indicated that greater APAP hepatotoxicity was correlated with higher hepatic CYP2E1 activity but not with the importance of fat accumulation in liver [54]. Thus, taking into account these data and other studies, we recently proposed that the occurrence of APAP-induced hepatotoxicity in an obese individual will depend on a subtle balance between metabolic factors that can be protective (i.e. lower absorption rate, higher volume of distribution, reduced CYP3A4 activity and increased APAP glucuronidation in liver) and others that can enhance the hepatic production of NAPQI (CYP2E1 induction) or lessen the detoxification rate of this toxic metabolite (lower GSH stores) [8].

The high variability of hepatic CYP2E1 activity in obese individuals could also explain why all these subjects are apparently not at risk for APAP-induced hepatotoxicity. Although different clinical investigations consistently showed that liver CYP2E1 activity is statistically greater in obesity and NAFLD, this activity is highly variable in obese patients [21,55-58]. In particular, whereas some obese patients present very high hepatic CYP2E1 activity, others are in the range of non-obese individuals [21,55-58]. Further investigations would be needed to determine whether some simple biomarkers could predict high hepatic CYP2E1 activity in obese individuals. Indeed, such biomarkers might be interesting to identify the patients for whom APAP should be avoided, in particular in long-term treatment. Finally, variability in hepatic GSH levels might also exist in obese individuals. Indeed, GSH stores could be lower in patients with NASH, as compared to individuals with simple fatty liver, because NASH is consistently associated with higher oxidative stress [8,9]. Consequently, greater oxidative stress and lower liver GSH at baseline could increase the threshold of APAP toxicity in certain obese patients [8].

3.1.2. Halothane and isoflurane

Halothane and isoflurane are two volatile halogenated anesthetics that have been reported to induce ALI more frequently in obese individuals [18,59-63]. Moreover, obesity seems to be associated with poor prognosis during halothane and isoflurane-induced hepatitis [60,61]. However, it is noteworthy that the above-mentioned investigations were based on relatively small number of patients and thus larger studies would be needed. Besides obesity, other factors such as female gender and repeated exposures to these halogenated anesthetics could also enhance the risk and severity of hepatotoxicity [61,63,64]. In the most severe cases, halothane and isoflurane are able to induce massive liver necrosis and fulminant hepatic failure that can require orthotopic liver transplantation, or even lead to the death of the patients [59,61,63]. These anesthetics can also induce cholestasis and steatosis in some patients [31,59,61].

Both halothane and isoflurane are predominantly oxidized in the liver by CYP2E1. Importantly, this CYP2E1-mediated oxidation generates electrophilic acyl chloride reactive metabolites able to covalently bind to a large array of cellular proteins [18,65,66]. Regarding halothane, covalent binding of the trifluoroacetyl chloride metabolite to key cellular targets is deemed to be involved in liver injury by triggering an immune-mediated reaction but also by inducing mitochondrial dysfunction, GSH depletion and oxidative stress [67-71]. Similar mechanisms of hepatotoxicity have been proposed for isoflurane [65,72]. Thus, taking into account the key role of CYP2E1 in the pathogenesis of halogenated anesthetic-induced hepatotoxicity, it is likely that the greater risk of liver injury in obese patients treated with halothane or isoflurane might be the consequence of the higher CYP2E1 activity commonly observed in obese patients (Figure 2) [7,18]. In keeping with this assumption, plasma levels of trifluoroacetic acid (i.e. a metabolite generated from trifluoroacetyl chloride) tended to be higher in morbidly obese patients compared to lean individuals [73]. Another study performed in morbidly obese individuals reported higher halothane biotransformation to the ionic fluoride metabolite [74]. Unfortunately, unlike the painkiller APAP, no investigations have been carried out in experimental NAFLD models in order to confirm that higher hepatic CYP2E1 activity is responsible for increased halothane hepatotoxicity in obesity. Finally, it should be pointed out that the higher risk of halothane and isoflurane-induced hepatotoxicity might not concern all obese patients but only a subset of patients who present the highest hepatic CYP2E1 activity, as previously discussed for APAP.

3.1.3. Methotrexate

Methotrexate (MTX) is a folate antagonist used in the treatment of some malignancies and different autoimmune and inflammatory diseases such as psoriasis, psoriatic arthritis and rheumatoid arthritis. Supplementation with folic acid (or folinic acid) is recommended in treated patients in order to reduce the risk of MTX-induced adverse effects including hepatotoxicity [75,76]. Notably, MTX-induced liver toxicity is mostly associated with its long-term use for the treatment of inflammatory diseases [77,78]. Although some cases of acute hepatitis with marked increase in plasma transaminases and hepatocellular necrosis have been reported in a few patients treated with high doses of intravenous MTX [79,80], this drug mostly induces chronic forms of liver lesion such as steatosis, steatohepatitis, fibrosis and even cirrhosis [31,78,80,81-83]. Psoriatic patients appear to have a higher risk for MTX-induced liver fibrosis. Indeed, several studies reported the presence of hepatic fibrosis in at least 20% of the treated psoriatic patients, although a majority of these cases corresponded to mild fibrosis [77,84,85].

Different studies reported that obesity and T2D could increase the risk of MTX-induced hepatotoxicity as assessed by increased plasma transaminases, or the presence of hepatic alterations such as steatohepatitis, fibrosis or cirrhosis [81,85,86-90]. However, when considering the investigations reporting only plasma transaminases [87,90], it is unclear whether MTX-induced transaminase elevation was actually linked to the occurrence of an acute hepatitis or a chronic form of liver injury such as steatohepatitis, or alternatively, to the worsening of pre-existing NAFLD. Because MTX-induced acute hepatitis seems to be an uncommon adverse event, it is likely that most cases of transaminase elevation during MTX treatment in obese and diabetic patients might be linked to the occurrence of a chronic form of liver injury, or to an aggravation of NAFLD. Hence, the issue of MTX-induced aggravation of pre-existing NAFLD and its possible pathophysiology will be discussed below. Finally, it is noteworthy that other factors seem to increase the risk of MTX-induced hepatotoxicity including cumulative dose and duration of MTX therapy, lack of folate supplementation, hyperlipidemia, alcohol abuse and genetic factors [87,90-93].

3.1.4. Other drugs

In a 6-year prospective study carried out in 74 patients with NAFLD and 174 with HCV-related chronic hepatitis, it has been found that NAFLD was an independent risk of DILI with an odds ratio of almost 4 [94]. In this study, ALI was observed in obese patients treated with fosinopril and losartan (antihypertensive agents), piperacillin/tazobactam and telithromycin (antibiotics), omeprazole (proton pump inhibitor) and ticlopidine (inhibitor of platelet aggregation) [94]. Although this report is interesting, prospective studies on larger population are needed in order to determine whether NAFLD is a bona fide independent risk of hepatotoxicity induced by these drugs.

Other investigations suggested that some drugs might not induce more frequently acute hepatitis in obese patients with NAFLD. These drugs include statins [95,96] and amiodarone [97]. However, these investigations were retrospective and further studies are warranted to confirm the innocuousness of these drugs in obese patients. Nonetheless, these reports suggest that obesity and NAFLD might not increase the risk of acute hepatitis for all hepatotoxic drugs, as previously mentioned.

3.2. Aggravation of pre-existing NAFLD

Clinical and experimental investigations indicate that chronic administration of some drugs can aggravate pre-existing NAFLD. Actually, the fact that NAFLD can be aggravated by long-term exposure to some xenobiotics is not restricted to drugs. For instance, different experimental and clinical studies have consistently shown that chronic alcohol abuse is particularly harmful for the steatotic liver and can severely worsen NAFLD [98-103].

As previously mentioned, NAFLD refers to a large spectrum of hepatic lesions including fatty liver, NASH and cirrhosis. Hence, aggravation of NAFLD can be secondary to an exacerbation of the intrahepatic accumulation of fat and/or a faster progression of the disease, for instance through a quicker transition from fatty liver to NASH. Conceptually, xenobiotics could exacerbate hepatic fat accretion by increasing DNL, limiting very low-density lipoprotein (VLDL) excretion and/or reducing mitochondrial FAO (Figure 3) [7,104]. Furthermore, a more rapid transition towards NASH could primarily involve ROS overproduction by the dysfunctional MRC, ER stress, lower antioxidant defences and higher expression of pro-inflammatory cytokines such as TNFα (Figure 3) [6,7]. Interestingly, almost all the above-mentioned events seem to be involved in NAFLD aggravation induced by chronic ethanol intoxication [101-103].

Figure 3. Mechanisms whereby some drugs could aggravate fatty liver and promote its progression to NASH in obese patients. Drug-induced worsening of fatty liver could be secondary to reduced VLDL secretion, increased lipogenesis and impaired mitochondrial FAO. The progression of fatty liver to NASH could be triggered by lower ATP production, higher mitochondrial ROS production, ER stress and inflammation. Notably, drug-induced inhibition of MRC could be a common mechanism leading to impaired FAO, reduced ATP levels and higher ROS production.

Figure 3

Actually, impairment of the MRC activity could be a central mechanism whereby some xenobiotics including drugs are able to worsen NAFLD (Figure 3) [6,7]. Indeed, MRC inhibition could not only contribute to the aggravation of fatty liver via the secondary impairment of mitochondrial FAO but also to the occurrence of necrosis secondary to ATP deficiency. In addition, MRC impairment is leading to higher mitochondrial ROS production, which could favor hepatic fibrosis [105,106]. Interestingly, several drugs discussed in the following sections were reported to inhibit the MRC activity. Table 2 summarizes the main data collected for these drugs.

Table 2. Drugs and aggravation of pre-existing NAFLD.

Drug(s) Pharmacological class Comments
Acetaminophen (APAP) Analgesic and antipyretic Some experimental studies in rodents suggest that chronic treatment with therapeutic doses of APAP could have deleterious effects on liver function and carbohydrate homeostasis [113,115,116]. Although it is currently unclear whether fatty liver might be particularly affected by long-term APAP treatment, further investigations are warranted in order to determine whether APAP can be safely prescribed for prolonged periods in obese people
Corticosteroids Anti-inflammatory Experimental studies showed that corticosteroids aggravated fatty liver in obese rodents [121,122]. Some experimental data also suggested that these drugs could also trigger NASH [121]. Hence, corticosteroids should be used with caution in obese patients
Anticancer Irinotecan Clinical studies reported that irinotecan administration before liver resection could induce steatohepatitis more frequently in obese individuals [126,127]
Methotrexate (MTX) anticancer and anti-inflammatory (anti-folate) Several clinical investigations suggested that MTX might aggravate NAFLD, in particular hepatic fibrosis [81,129,130]
Pentoxifylline (PTX) Hemorrheologic agent with anti-TNF properties PTX might aggravate NAFLD in a few patients [166,167]. Investigations in obese mice suggested that PTX-induced worsening of fatty liver could be secondary to hyperglycemia and higher hepatic DNL [171]
Phenobarbital Antiepileptic Experimental investigations showed that phenobarbital exacerbated hepatic lipid deposition in obese rats [180]. No data are available for obese patients
Raloxifene Anti-osteoporotic (selective estrogen receptor modulator) A case report described a case of NASH aggravation in a female patient treated with raloxifene [215]. Further investigations would be needed in order to determine whether raloxifene could pose a potential risk in obese patients
Rosiglitazone Antidiabetic (PPARγ agonist) Worsening of necroinflammation, perisinusoidal fibrosis and steatosis was observed in some obese patients treated with rosiglitazone [195,197,198]. Experimental investigations performed in different murine models of obesity and T2D confirmed that rosiglitazone was able to exacerbate hepatic steatosis and liver TAG accumulation [200-205]. Some experimental data also suggested that necroinflammation could also be worsened by rosiglitazone [200,202]
Stavudine (d4T) Antiretroviral (NRTI) Some clinical investigations suggested that d4T could induce fatty liver disease or worsen NAFLD in some patients, although further investigations would be needed to ascertain NAFLD aggravation [119,154-156]. Other studies also suggested that the risk of d4T-induced lactic acidosis could be higher in overweight or obese female patients [159-162]. Other NRTIs such as didanosine (ddI) might also aggravate NAFLD in obese patients [156].
Tamoxifen Anticancer (selective estrogen receptor modulator) Some clinical studies showed that obesity enhanced the risk of tamoxifen-induced steatohepatitis [211,213]. However, further investigations are needed in order to determine whether tamoxifen is able to worsen pre-existing NAFLD in obese patients
Tetracycline Antibiotic Experimental investigations showed that tetracycline exacerbated hepatic TAG deposition and triggered steatohepatitis in obese mice [231]. However, this interesting observation is not clinically relevant because tetracycline is no longer prescribed in patients

3.2.1. Acetaminophen (APAP)

As previously mentioned, therapeutic doses of APAP can induce liver injury in some patients. About one third of treated patients could have a significant increase in their plasma transaminases after 1 or 2 weeks of APAP treatment [43,107]. Although extremely rare, fulminant hepatitis have been reported in patients after several days of APAP treatment at recommended dosage [44,45]. Moreover, long-term intake of APAP has been reported to induce chronic liver injury in a few treated patients, such as cirrhosis and granulomatous hepatitis [31,108]. Interestingly, some data suggest that chronic APAP ingestion could significantly reduce GSH stores, at least in some treated patients [109,110]. Lastly, therapeutic doses of APAP could also have detrimental effects on the kidney and the cardiovascular system [111,112]. Thus, therapeutic doses of APAP might be at risk for some individuals, in particular during chronic treatment. Accordingly, several investigations in rodents showed that long-term treatment with therapeutic doses of APAP was able to induce oxidative stress and cellular injury in different tissues [113-115]. Interestingly, a single low dose of APAP in mice induced reversible mitochondrial dysfunction and steatosis in liver, but this study did not determine the effects of repeated administration of such low APAP dose [116].

There is currently no information as to whether NAFLD might be aggravated in some patients by therapeutic doses of APAP. However, recent experimental studies performed in our laboratory suggested that APAP administration might not be safe in NAFLD (Begriche et al., manuscript in preparation). Indeed, a 1-month treatment with therapeutic doses of APAP significantly enhanced plasma aspartate aminotransferase (AST) activity by 45% in mice fed a high-fat diet (HFD), whereas this increase was only 19% in mice fed a normal diet. Furthermore, HFD obese mice treated by APAP presented a higher hepatic mRNA expression of several proteins responsive to cellular stress including γ-glutamylcysteine synthetase (γ-GCS), tribbles pseudokinase 3 (Trib3) and X-box binding protein 1 (Xbp1). However, chronic APAP treatment concomitantly induced in obese mice a significant reduction of liver TAG levels associated with lower plasma insulin and reduced liver expression of different genes involved in DNL including SREBP1c, stearoyl-coenzyme A desaturase 1 (SCD1) and glycerol-3-phosphate acyltransferase (GPAT). In contrast to insulin, blood glucose was not significantly changed by APAP. Other studies in mice reported the effect of chronic treatment with therapeutic doses of APAP on glucose homeostasis and insulin resistance. In two studies from the same group, APAP was reported to alleviate impaired glucose intolerance and hyperinsulinemia in HFD obese mice [117,118]. However, long-term administration of APAP induced glucose intolerance and hepatic insulin resistance in mice fed a chow diet [115]. Furthermore, APAP decreased serum insulin and β-cell relative volume in these mice [115]. Clearly, further investigations are warranted in order to determine whether APAP can be safely prescribed for chronic treatment, in particular in patients suffering from obesity and related metabolic disorders.

3.2.2. Corticosteroids

These drugs are used in numerous diseases for their antiinflammatory and anti-allergic effects. However, one of the main adverse effects of this drug class is the occurrence of metabolic and endocrine disturbances and in particular obesity, hyperlipidemia, steatosis, steatohepatitis, IR and diabetes [6,7,119,120]. In two independent studies carried out in HFD obese rodents, long-term treatment with corticosteroids (corticosterone or dexamethasone) significantly worsened hepatic steatosis [121,122]. In one of these studies, plasma ALT, total bilirubin and liver collagen content were significantly enhanced in HFD obese rats treated with corticosterone whereas no such disturbances were observed in untreated HFD rats [121]. Thus, in the context of obesity, corticosteroids might not only aggravate fatty liver but might also trigger NASH. Interestingly, corticosteroids are able to disturb mitochondrial FAO, MRC activity and oxidative phosphorylation in liver [123-125]. However, further investigations would be required to determine whether mitochondrial dysfunction is involved in NAFLD worsening in corticosteroid-treated rodents [121,122].

3.2.3. Irinotecan

This topoisomerase 1 inhibitor, also known as CPT-11, is mainly used for the treatment of advanced colorectal cancer. Irinotecan is also used before resection of hepatic colorectal metastases. Two independent clinical studies reported that irinotecan administration before liver resection could induce steatohepatitis (sometimes severe) in treated patients and that the risk of liver injury appeared to be greater in obese individuals [126,127]. Although the reason of this increased susceptibility is currently unknown, recent experimental investigations in mice showed that obesity was associated with higher plasma levels of the toxic irinotecan metabolite, SN-38 [128].

3.2.4. Methotrexate

As previously mentioned, it is likely that most cases of transaminase elevation in obese and diabetic patients treated with MTX could be linked to the occurrence of a chronic form of liver injury such as steatohepatitis, or to an aggravation of pre-existing NAFLD. Although this issue is still unclear, an aggravation of NAFLD is supported by a clinical study carried out in a small group (n=20) of obese and/or diabetic patients for whom serial liver biopsies were performed [81]. In this study, fibrosis was aggravated in three-quarter of the treated patients, whereas steatosis and necroinflammation were worsened in the remaining individuals [81]. Thus, although these data indicated that all histological components of NAFLD could be worsened by MTX, it seems that this drug could more frequently aggravate hepatic fibrosis. It is noteworthy that other studies using non-invasive methods such as ultrasound elastography also suggested that MTX might aggravate liver fibrosis in obese patients [129,130]. However, a serial assessment of hepatic fibrosis was not performed in these studies. Interestingly, recent investigations in a rat model of steatohepatitis induced by a methionine choline-deficient (MCD) diet showed that a single injection of MTX was able to induce liver fibrosis in the MCD rats whereas no fibrosis was observed in rats fed with a control diet [131,132]. Although these data are interesting, it should be underlined that the MCD model of NASH does not reflect the metabolic context of obesity because the MCD diet induces a significant loss of body weight and lower glycemia [9,133].

Although the exact mechanism whereby MTX could aggravate NASH is still unknown, one attractive hypothesis is mitochondrial dysfunction (Figure 3). Indeed, MTX-induced mitochondrial dysfunction has been shown in different experimental models [104,134,135]. In particular, our recent investigations in isolated mouse liver mitochondria showed that relatively low concentrations of MTX (~40-50 μM) were able to inhibit oxygen consumption with glutamate/malate or palmitoyl-L-carnitine/malate as respiratory substrates [104]. These mitochondrial effects might explain why MTX is able to induce hepatic steatosis, necrosis and fibrosis, as already discussed.

A previous study in patients with MTX-induced liver fibrosis reported a hepatic overexpression of different genes encoding complement components including C3, C5 and C8a [136]. Interestingly, activation of the complement system could play a role in the progression of NAFLD [137,138] and in the occurrence of fibrosis in other liver diseases [139,140]. Thus, MTX might aggravate NAFLD and especially fibrosis by further activating the complement system in liver. Although MTX-induced mitochondrial dysfunction and complement activation might be attractive hypotheses, it is clear that further investigations are needed to decipher the mechanism(s) whereby MTX is able to worsen NAFLD in obese patients.

3.2.5. Nucleoside reverse transcriptase inhibitors

The nucleoside reverse transcriptase inhibitors (NRTIs) are efficient drugs used for the treatment of HIV infection. NRTIs include abacavir (ABC), didanosine (ddI), lamivudine (3TC), stavudine (d4T) and zidovudine (AZT). These drugs can cause various adverse effects, sometimes severe and fatal, including lactic acidosis, bone marrow suppression, myopathy, pancreatitis and liver injury [141-143]. NRTI-induced hepatotoxicity includes hepatic cytolysis, microvesicular and macrovacuolar steatosis, steatohepatitis and cirrhosis [104,141-143]. This hepatic toxicity is particularly observed with d4T, ddI and AZT [31,143]. These latter NRTIs can also induce lipoatrophy, which is characterized by a loss of WAT in different body areas [141,144,145]. Notably, NRTI-induced lipoatrophy could promote secondary IR in some tissues including WAT and muscle [146,147]. It is acknowledged that most NRTI-induced adverse effects are caused by depletion of mitochondrial DNA (mtDNA), a small genome encoding for 13 proteins mandatory for MRC activity and OXPHOS [6,141,143]. Importantly, mtDNA depletion-induced MRC alteration in turn leads to an impairment of mitochondrial FAO and tricarboxylic acid (TCA) cycle, responsible for hepatic steatosis and lactic acidosis, respectively [6,141,148-150]. However, it is noteworthy that NRTI-induced toxic effects not related to mtDNA depletion were also reported in different experimental investigations [141,151-153].

Some NRTIs, in particular d4T and ddI, are suspected to induce fatty liver disease or worsen NAFLD in treated patients [119,154-156]. Several different mechanisms, which are not mutually exclusive, might be involved in these deleterious effects. First, NRTI-induced impairment of hepatic MRC activity could be involved, as previously discussed (Figure 3). Second, NRTIs could also induce hepatic lipid deposition by inducing ER stress and inhibiting autophagy activity [157,158]. Third, NRTI could promote fatty liver by way of lipoatrophy-related IR [154,155], in a similar way than for obesity-induced IR [9,11].

It is noteworthy that several studies suggested that the risk of d4T-induced lactic acidosis could be higher in overweight or obese female patients [159-162]. Moreover, a high BMI might increase the risk of fatal lactic acidosis [163]. Although the exact mechanisms underlying this higher risk are currently unknown, it is conceivable that d4T treatment might have significantly exacerbated NAFLD-associated mitochondrial dysfunction in these obese individuals, thus triggering severe TCA cycle deficiency and lactic acidosis. Unfortunately, these investigations did not determine whether d4T also worsen fatty liver or NASH in these patients.

3.2.6. Pentoxifylline

The methylxanthine derivative pentoxifylline (PTX) is a nonselective phosphodiesterase inhibitor commonly prescribed for peripheral vascular disorders and intermittent claudication by improving blood flow and circulation. Because PTX is also an inhibitor of TNFα synthesis, it has also been tested in different diseased states involving this pro-inflammatory cytokine such as alcoholic hepatitis and NASH. However, recent studies suggested that PTX could not be efficient for the treatment of alcoholic hepatitis [164,165]. Regarding NASH, although some studies reported a beneficial effect of PTX treatment [166-168], others did not show any efficacy of this drug compared to placebo [169,170]. Moreover, steatosis, inflammation and fibrosis worsened in a few patients treated with PTX, although it was unclear whether this was related to the treatment or other factors [166,167].

Investigations carried out in wild-type (i.e. lean) mice and obese and diabetic ob/ob mice treated for 3 weeks with PTX showed that this drug aggravated fatty liver in ob/ob mice, whereas liver lipid accumulation was not observed in PTX-treated lean mice [171]. In addition, PTX further enhanced plasma ALT activity in obese mice, thus reflecting aggravation of hepatic cytolysis [171]. Another study showed that a 2-week treatment with PTX significantly increased hepatic TAG levels in mice fed a MCD diet [172]. Investigations in ob/ob mice also suggested that PTX could promote DNL in liver through a ChREBP-dependent pathway, possibly activated by PTX-induced hyperglycemia [171]. Moreover, additional investigations suggested that this hyperglycemia could be secondary to higher intestinal glucose absorption due to increased jejunal expression of the glucose transporter GLUT2 [171]. Interestingly, the methylxanthine caffeine is suspected to exaggerate post-prandial hyperglycemia in patients with T2D [173,174]. Altogether, these data suggest that long-term treatment with PTX might aggravate NAFLD in some patients, in particular in those suffering from T2D, by increasing post-prandial glycemia and hepatic DNL [171].

3.2.7. Phenobarbital

Phenobarbital is a barbiturate derivative used for the treatment of epilepsy. Notably, phenobarbital is a potent inducer of different hepatic CYPs including CYP2B, CYP2C and CYP3A, in particular by activating the nuclear constitutive androstane receptor (CAR) [175,176]. Some studies also showed that phenobarbital is able to enhance CYP2E1 activity [177-179], but the mechanism of such induction is still unknown. Experimental investigations showed that phenobarbital exacerbated hepatic lipid deposition in HFD obese rats, whereas this antiepileptic drug did not induce lipid accumulation in lean rats [180]. Although the mechanism whereby phenobarbital aggravated hepatic steatosis was not determined in this study, some data might suggest a role of CYP2E1 induction. Indeed, investigations in different murine models support the notion that CYP2E1 favors hepatic lipogenesis and lipid deposition [23,181-184]. Thus, phenobarbital-induced aggravation of fatty liver in HFD obese rats [180] might have been induced, at least in part, by the exacerbation of NAFLD-related CYP2E1 induction. Alternatively, phenobarbital might have exacerbated fatty liver by activating CAR [6]. However, the role of CAR in drug-induced steatosis is still unclear and could depend on the duration of CAR activation and/or the nature of the activating molecule [6].

3.2.8. Rosiglitazone

This thiazolidinedione (TZD) derivative is a PPARγ agonist used for the treatment of T2D. Indeed, TZDs enhance insulin sensitivity by different mechanisms including reduction of circulating free fatty acids, increased adiponectin secretion and stimulation of FAO and energy expenditure [185,186]. Rosiglitazone belongs to the same pharmacological class as troglitazone, which has been withdrawn from the market in 2000 because of the occurrence of several cases of severe (sometimes fatal) liver injury [187,188]. Importantly, several studies suggested that mitochondrial dysfunction could play a major role in troglitazone-induced hepatotoxicity [189,190].

Although rosiglitazone is safer than troglitazone, several cases of hepatotoxicity, sometimes severe, were reported in treated patients [31,191,192]. Interestingly, rosiglitazone also induces mitochondrial dysfunction in particular by inhibiting the activity of different MRC complexes [190,193]. However, the extent of rosiglitazone-induced inhibition of MRC complexes is less compared to troglitazone [190,193]. Pioglitazone, another antidiabetic PPARγ agonist, presents a hepatotoxicity profile and an ability to impair mitochondrial dysfunction that seems about similar to rosiglitazone [31,190]. Besides hepatotoxicity, a major concern with the use of rosiglitazone is its possible association with increased risk of cardiac diseases including congestive heart failure [186]. Rosiglitazone-induced cardiotoxicity could also involve mitochondrial dysfunction [186].

Because of their beneficial effects on insulin sensitivity, rosiglitazone and pioglitazone have been tested in the treatment of NASH [186,194]. Although pioglitazone showed beneficial effects on NASH progression, rosiglitazone efficiency was less consistent, especially during long-term treatment [194-196]. In addition, worsening of necroinflammation, perisinusoidal fibrosis and steatosis was observed in some patients [195,197]. In another study, rosiglitazone treatment was interrupted in an obese woman with severe steatosis because of a significant rise in serum ALT and AST [198]. Interestingly, increased hepatic expression of different pro-inflammatory genes (i.e. TLR4, IL-8 and CCL2) was reported in patients treated with rosiglitazone [199].

Several experimental investigations carried out in different murine models of obesity and T2D consistently reported that rosiglitazone was able to exacerbate hepatic steatosis and liver TAG accumulation [200-205]. Interestingly, two of these studies reported that aggravation of steatosis was associated with increased circulating ALT levels, thus suggesting that necroinflammation could also be worsened by rosiglitazone [200,202]. Although the exact mechanism(s) whereby rosiglitazone is able to aggravate liver steatosis in obese and diabetic mice is still unknown, some data suggest an exacerbation of hepatic DNL [200], possibly mediated by the high expression of PPARγ already present in the obese liver [202,205]. Further impairment of MRC complex I activity and exacerbation of oxidative stress might have contributed to the possible worsening of hepatic necroinflammation in rosiglitazone-treated obese mice [202].

3.2.9. Tamoxifen

Tamoxifen is a selective estrogen receptor modulator used for the treatment of estrogen receptor positive breast cancer. Tamoxifen can induce different types of chronic liver lesion including steatosis, steatohepatitis, fibrosis, cirrhosis and hepatocellular carcinoma [31,78,83,206]. Whereas steatosis can be detected in at least one third of the treated patients [207,208], tamoxifen-induced steatohepatitis and cirrhosis seems to be uncommon [209-211]. Similarly, tamoxifen-induced acute hepatitis seems to be rare [31].

Several studies reported that obesity increased the risk of tamoxifen hepatotoxicity as assessed by increased plasma transaminases, or the presence of hepatic histologic alterations [211-213]. Two of these studies showed that obesity more specifically enhanced the risk of tamoxifen-induced steatohepatitis [211,213]. Interestingly, in a study reporting three cases of tamoxifen-induced steatohepatitis, all patients were over-weight, or obese [214]. However, it is noteworthy that one of the above-cited study suggested that tamoxifen did not worsen pre-existing NASH in a small subgroup of patients [211]. Hence, further investigations in larger series of obese patients undergoing serial liver biopsies would be needed in order to confirm these data. This could be clinically relevant because a previous study reported a case of NASH aggravation in a female patient treated with raloxifene [215], a selective estrogen receptor modulator used to treat osteoporosis.

The mechanism whereby tamoxifen could induce steato-hepatitis more frequently in obese individuals is currently unknown, although mitochondrial dysfunction could be involved (Figure 3). Indeed, tamoxifen is able to impair MRC activity in isolated rodent liver mitochondria via a direct inhibition of several MRC complexes [104,216-218]. One of these studies also showed that chronic tamoxifen treatment in mice induced mtDNA depletion and impairment of different MRC complexes in liver [216]. Because mtDNA encodes 13 MRC polypeptides [143], mtDNA depletion could also contribute to the impairment of MRC activity observed in mice treated by tamoxifen, in addition to its direct inhibitory effect on the MRC [216]. Interestingly, studies carried out in rodents showed that tamoxifen induced oxidative stress in liver, including at the mitochondrial level [219-221]. However, a single dose of tamoxifen was administered in these studies and further investigations with chronic tamoxifen treatment are required.

As previously mentioned, numerous investigations pointed to the occurrence of mitochondrial dysfunction in NAFLD. In particular, lower mitochondrial respiration with MRC substrates and reduced activity of MRC complexes have consistently been found in different rodent models of NAFLD, including in rodents with simple fatty liver or mild NASH [9,222]. Hence, it is tempting to speculate that tamoxifen might exacerbate MRC impairment in obese patients and trigger steatohepatitis in individuals with the more severe mitochondrial dysfunction and the highest ROS overproduction.

3.2.10. Tetracycline

Tetracycline is a broad-spectrum antibiotic prescribed to treat different bacterial infections. However, the clinical use of tetracycline declined because the high doses required to treat infections induced numerous adverse effects. In particular, tetracycline has been responsible for several severe cases of hepatotoxicity characterized by microvesicular steatosis and fulminant liver failure [143,223]. Experimental investigations suggested that tetracycline-induced inhibition of mitochondrial FAO could be an important mechanism leading to hepatic steatosis [224-226]. A recent study suggested that one mechanism of tetracycline-induced impairment of mitochondrial FAO could be secondary to the oxidative inhibition of long-chain acyl-CoA dehydrogenase, a key enzyme of the mitochondrial FAO pathway [227]. Other investigations showed that tetracycline was able to inhibit mitochondrial protein synthesis [228-230]. The latter effect could worsen mitochondrial dysfunction during long-term tetracycline treatment because all the mtDNA-encoded proteins are mandatory for MRC activity and OXPHOS [143], as previously mentioned.

A previous study carried out in mice showed that tetracycline aggravated NAFLD in HFD obese mice [231]. Interestingly, tetracycline not only exacerbated hepatic TAG deposition but also induced steatohepatitis in obese mice, whereas no sign of tetracycline-induced hepatotoxicity was observed in lean mice [231]. Liver expression of TNFα and IL1β (two pro-inflammatory cytokines) and that of α-smooth muscle actin (α-SMA, a marker of fibrosis) were enhanced only in obese mice treated by tetracycline [231]. Unfortunately, the mechanism whereby tetracycline worsened NAFLD in obese mice was not assessed in this study. Further investigations will be needed in order to determine whether tetracycline-induced mitochondrial dysfunction and oxidative stress could be involved. It will also be interesting to assess the hepatic effects of other tetracycline derivatives in obese animals. Indeed, derivatives such as doxycycline, minocycline and rolitetracycline are also able to induce mitochondrial dysfunction [230,232,233]. Finally, it is noteworthy that tetracycline derivatives present other metabolic effects that could promote hepatic lipid accumulation including impairment of TAG secretion and stimulation of DNL [232,234,235].

4. General discussion and remaining issues

After reading this review, the reader might have the feeling that most of the data regarding DILI in obesity and NAFLD do not rely on solid evidence. This is right because this medical issue has been identified relatively recently and thus, only a few data have been collected thanks to a handful of clinical and experimental investigations. The sad news is that one cannot expect to have a clear picture of the situation in the next few years because there are more than 350 potential hepatotoxic drugs currently on the market [1,2], without mentioning the numerous herbals and dietary supplements that can also induce liver injury [236-238]. Importantly, many drugs, herbals and dietary supplements are taken by obese patients in order to treat different associated disorders (hyperlipidemia, T2D, hypertension), or to induce weight loss. Clearly, more investigations are needed in order to determine what are the drugs (or other compounds) that can induce liver injury in obese patients and also to decipher the involved mechanisms.

The issue of DILI in obesity is also complicated by the fact that an aggravation of NAFLD secondary to a treatment could be clinically silent in some obese individuals. Indeed, normal levels of plasma ALT and AST can be observed in a significant number of patients with NAFLD, even in those with NASH and hepatic fibrosis [239,240]. Moreover, when NAFLD is associated with increased transaminase levels, such elevation is in general modest and cannot discriminate simple steatosis from NASH [241,242]. Thus, caregivers should keep in mind that drug-induced NAFLD aggravation will not necessarily be associated with a worsening of liver enzymes and that other investigations could be needed to monitor hepatic function and histology.

Two different strategies can be implemented to collect further information regarding DILI in obesity and NAFLD. Notably, these strategies are not mutually exclusive.

First, prospective clinical studies might be designed but one should keep in mind that DILI is a rare event. Indeed, it is estimated that this adverse effect occurs at the most in about 1 per 10,000 individuals who take the drug [237,243]. Thus, it will be difficult for some drugs to draw valid conclusion because the very low number of DILI cases will preclude to statistically ascertain that obesity and/or NAFLD actually increases the risk of hepatotoxicity. Furthermore, as previously mentioned, numerous drugs are potentially hepatotoxic and thus it is difficult to a priori select one particular drug (or a drug class) that will be specifically studied in patients. Despite these caveats, it would be interesting to carry out prospective studies with drugs with high toxicity potential (e.g. methotrexate, tamoxifen, NRTIs) in order to confirm or not their liability in obese patients.

A second strategy is to perform experimental investigations in order to identify the drugs that might be more hepatotoxic in obesity and NAFLD. Although studies in obese rodents can be informative [53,54,121,171,202,204,231], such in vivo investigations are not amenable for the screening of a large number of drugs. In contrast, in vitro investigations appear to be the best strategy to rapidly screen a large number of drugs. In the past ten years, numerous cellular models of NAFLD have been developed by researchers willing to study the different patho-physiological consequences related to hepatocellular fat accumulation, including XME expression and drug-induced cytotoxicity [244-252]. However, most of these studies used human hepatoma cell lines (e.g. HuH7, HepG2) that do not have the full repertoire of XMEs, or rodent hepatocytes that do not have the same profile of drug metabolism than human hepatocytes. Moreover, all these studies were performed in cells incubated with fatty acids for a short duration of time (from a few hours to 2 or 3 days). This is probably too short to induce most of the hepatocellular alterations or adaptations secondary to lipid accumulation. Recently, we set up a cellular model of NAFLD by using HepaRG cells incubated with stearic acid for 7 days [24]. Importantly, CYP2E1 activity was increased whereas CYP3A4 activity was decreased in these stearate-loaded cells [24], thus reproducing what has been consistently observed in obese patients with NAFLD [8,18,25,26]. In addition, our cellular model was successfully used to demonstrate that increased APAP cytotoxicity in stearate-loaded cells was due to higher CYP2E1 activity [24]. Hence, we believe that such cellular model can be valuable to screen a great number of potentially hepatotoxic drugs. For drugs presenting significantly higher cytotoxicity in steatotic cells compared to non-steatotic cells, further studies in obese rodents might be warranted before considering clinical investigations. Even if drug metabolism presents significant differences between rodent and human, such investigations could be interesting in order to determine whether the in vitro effects can be reproduced in an in vivo situation.

Experimental investigations also offer the possibility to perform mechanistic investigations. By using rodent and cellular models of NAFLD, it will be for instance interesting to determine whether mitochondrial dysfunction is a central mechanism involved in drug-induced aggravation of NAFLD (Figure 3). Notably, almost all drugs that have been shown or suspected to worsen NAFLD are able to impair mitochondrial function, although the exact mechanisms of such mitochondrial dysfunction greatly differ from one drug to another. As already mentioned, drug-induced mitochondrial dysfunction might explain not only why hepatic fat can further accumulate but also why necroinflammation and fibrosis can be worsened in some patients. However, although numerous drugs are known to be mitochondriotoxic [4,6,104,253-255], it seems unlikely that NAFLD aggravation could be observed with all these drugs. Thus, additional mechanisms unrelated to mitochondrial dysfunction might also be involved such as induction of ER stress, JNK activation, impairment of autophagy and the accumulation of certain lipid intermediates since these events could play a significant role in NAFLD progression [256-258]. Finally, it would be also interesting to determine whether the drugs shown or suspected to worsen NAFLD are able to reduce the expression and activity of patatin-like phospholipase domain containing 3 (PNPLA3), a lipase which appears to play a major role in the occurrence of fatty liver and its progression to steatohepatitis [259,260].

Acknowledgements

We are grateful to INSERM (Institut National de la Recherche et de la Santé Médicale) for its constant financial support.

Glossary

ABBREVIATIONS

Abbreviation Expansion
ALF

acute liver failure;

ALI

acute liver injury;

ALT

alanine aminotransferase;

APAP

acetaminophen;

AST

aspartate aminotransferase;

AZT

zidovudine;

BMI

body mass index;

CAR

constitutive androstane receptor;

ChREBP

carbohydrate responsive element-binding protein;

CYP

cytochrome P450;

ddI

didanosine;

DILI

drug-induced liver injury;

DNL

de novo lipogenesis;

d4T

stavudine;

ER

endoplasmic reticulum;

FAO

fatty acid oxidation;

GSH

glutathione;

HFD

high-fat diet;

IR

insulin resistance;

MCD

methionine choline-deficient;

MRC

mitochondrial respiratory chain;

MTX

methotrexate;

mtDNA

mitochondrial DNA;

NAFLD

nonalcoholic fatty liver disease;

NAPQI

N-acetyl-p-benzoquinone imine;

NASH

nonalcoholic steatohepatitis;

NEFA

non-esterified fatty acids;

NRTI

nucleoside reverse transcriptase inhibitor;

OXPHOS

oxidative phosphorylation;

PPAR

peroxisome proliferator-activated receptor;

PTX

pentoxifylline;

ROS

reactive oxygen species;

SREBP-1c

sterol regulatory element-binding protein-1c;

TAG

triacylglycerol;

TCA

tricarboxylic acid;

T2D

type 2 diabetes;

TNFα

tumor necrosis factor-α;

TZD

thiazolidinedione;

VLDL

very low-density lipoprotein;

XME

xenobiotic-metabolizing enzyme;

WAT

white adipose tissue.

Disclosure

The authors do not have any conflict of interest to report regarding this work.

References

  • [1].Weng Z, Wang K, Li H, Shi Q. A comprehensive study of the association between drug hepatotoxicity and daily dose, liver metabolism, and lipophilicity using 975 oral medications. Oncotarget. 2015;6:17031–17038. doi: 10.18632/oncotarget.4400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Björnsson ES, Hoofnagle JH. Categorization of drugs implicated in causing liver injury: Critical assessment based on published case reports. Hepatology. 2016;63:590–603. doi: 10.1002/hep.28323. [DOI] [PubMed] [Google Scholar]
  • [3].Robles-Diaz M, Lucena MI, Kaplowitz N, Stephens C, Medina-Caliz I, Gonzalez-Jimenez A, Ulzurrun E, Gonzalez AF, Fernandez MC, Romero-Gomez M, Jimenez-Perez M, Bruguera M, Prieto M, Bessone F, Hernandez N, Arrese M, Andrade RJ. Use of Hy’s law and a new composite algorithm to predict acute liver failure in patients with drug-induced liver injury. Gastroenterology. 2014;147:109–118. doi: 10.1053/j.gastro.2014.03.050. [DOI] [PubMed] [Google Scholar]
  • [4].Labbe G, Pessayre D, Fromenty B. Drug-induced liver injury through mitochondrial dysfunction: mechanisms and detection during preclinical safety studies. Fundam Clin Pharmacol. 2008;22:335–353. doi: 10.1111/j.1472-8206.2008.00608.x. [DOI] [PubMed] [Google Scholar]
  • [5].Webb GJ, Adams DH. Modeling idiosyncrasy: a novel animal model of drug-induced liver injury. Hepatology. 2015;61:1124–1126. doi: 10.1002/hep.27617. [DOI] [PubMed] [Google Scholar]
  • [6].Begriche K, Massart J, Robin MA, Borgne-Sanchez A, Fromenty B. Drug-induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and deleterious consequences for the liver. J Hepatol. 2011;54:773–794. doi: 10.1016/j.jhep.2010.11.006. [DOI] [PubMed] [Google Scholar]
  • [7].Fromenty B. Drug-induced liver injury in obesity. J Hepatol. 2013;58:824–826. doi: 10.1016/j.jhep.2012.12.018. [DOI] [PubMed] [Google Scholar]
  • [8].Michaut A, Moreau C, Robin MA, Fromenty B. Acetaminophen-induced liver injury in obesity and nonalcoholic fatty liver disease. Liver Int. 2014;34:e171–e179. doi: 10.1111/liv.12514. [DOI] [PubMed] [Google Scholar]
  • [9].Begriche K, Massart J, Robin MA, Bonnet F, Fromenty B. Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology. 2013;58:1497–1507. doi: 10.1002/hep.26226. [DOI] [PubMed] [Google Scholar]
  • [10].Xu R, Tao A, Zhang S, Deng Y, Chen G. Association between patatin-like phospholipase domain containing 3 gene (PNPLA3) polymorphisms and nonalcoholic fatty liver disease: a HuGE review and meta-analysis. Sci Rep. 2015;5:9284. doi: 10.1038/srep09284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Hardy T, Oakley F, Anstee QM, Day CP. Nonalcoholic fatty liver disease: pathogenesis and disease spectrum. Annu Rev Pathol. 2016;11:451–496. doi: 10.1146/annurev-pathol-012615-044224. [DOI] [PubMed] [Google Scholar]
  • [12].Poupeau A, Postic C. Cross-regulation of hepatic glucose metabolism via ChREBP and nuclear receptors. Biochim Biophys Acta. 2011;1812:995–1006. doi: 10.1016/j.bbadis.2011.03.015. [DOI] [PubMed] [Google Scholar]
  • [13].Hirsova P, Ibrahim SH, Gores GJ, Malhi H. Lipotoxic lethal and sublethal stress signaling in hepatocytes: relevance to NASH pathogenesis. J Lipid Res. 2016;57:1758–1770. doi: 10.1194/jlr.R066357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Satapati S, Kucejova B, Duarte JA, Fletcher JA, Reynolds L, Sunny NE, He T, Nair LA, Livingston K, Fu X, Merritt ME, Sherry AD, Malloy CR, Shelton JM, Lambert J, Parks EJ, Corbin I, Magnuson MA, Browning JD, Burgess SC. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J Clin Invest. 2015;125:4447–4462. doi: 10.1172/JCI82204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Kakimoto PA, Tamaki FK, Cardoso AR, Marana SR, Kowaltowski AJ. H2O2 release from the very long chain acyl-CoA dehydrogenase. Redox Biol. 2015;4:375–380. doi: 10.1016/j.redox.2015.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Knockaert L, Fromenty B, Robin MA. Mechanisms of mitochondrial targeting of cytochrome P450 2E1: physiopathological role in liver injury and obesity. FEBS J. 2011;278:4252–4260. doi: 10.1111/j.1742-4658.2011.08357.x. [DOI] [PubMed] [Google Scholar]
  • [17].Hartman JH, Martin HC, Caro AA, Pearce AR, Miller GP. Subcellular localization of rat CYP2E1 impacts metabolic efficiency toward common substrates. Toxicology. 2015;338:47–58. doi: 10.1016/j.tox.2015.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Aubert J, Begriche K, Knockaert L, Robin MA, Fromenty B. Increased expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: mechanisms and pathophysiological role. Clin Res Hepatol Gastroenterol. 2011;35:630–637. doi: 10.1016/j.clinre.2011.04.015. [DOI] [PubMed] [Google Scholar]
  • [19].Weltman MD, Farrell GC, Hall P, Ingelman-Sundberg M, Liddle C. Hepatic cytochrome P450 2E1 is increased in patients with nonalcoholic steatohepatitis. Hepatology. 1998;27:128–133. doi: 10.1002/hep.510270121. [DOI] [PubMed] [Google Scholar]
  • [20].Chalasani N, Gorski JC, Asghar MS, Asghar A, Foresman B, Hall SD, Crabb DW. Hepatic cytochrome P450 2E1 activity in nondiabetic patients with nonalcoholic steatohepatitis. Hepatology. 2003;37:544–550. doi: 10.1053/jhep.2003.50095. [DOI] [PubMed] [Google Scholar]
  • [21].van Rongen A, Välitalo PA, Peeters MY, Boerma D, Huisman FW, van Ramshorst B, van Dongen EP, van den Anker JN, Knibbe CA. Morbidly obese patients exhibit increased CYP2E1-mediated oxidation of acetaminophen. Clin Pharmacokinet. 2016;55:833–847. doi: 10.1007/s40262-015-0357-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Schattenberg JM, Wang Y, Singh R, Rigoli RM, Czaja MJ. Hepatocyte CYP2E1 overexpression and steatohepatitis lead to impaired hepatic insulin signaling. J Biol Chem. 2005;280:9887–9894. doi: 10.1074/jbc.M410310200. [DOI] [PubMed] [Google Scholar]
  • [23].Kathirvel E, Morgan K, French SW, Morgan TR. Overexpression of liver-specific cytochrome P4502E1 impairs hepatic insulin signaling in a transgenic mouse model of nonalcoholic fatty liver disease. Eur J Gastroenterol Hepatol. 2009;21:973–83. doi: 10.1097/MEG.0b013e328328f461. [DOI] [PubMed] [Google Scholar]
  • [24].Michaut A, Le Guillou D, Moreau C, Bucher S, McGill MR, Martinais S, Gicquel T, Morel I, Robin MA, Jaeschke H, Fromenty B. A cellular model to study drug-induced liver injury in nonalcoholic fatty liver disease: Application to acetaminophen. Toxicol Appl Pharmacol. 2016;292:40–55. doi: 10.1016/j.taap.2015.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Brill MJ, Diepstraten J, van Rongen A, van Kralingen S, van den Anker JN, Knibbe CA. Impact of obesity on drug metabolism and elimination in adults and children. Clin Pharmacokinet. 2012;51:277–304. doi: 10.2165/11599410-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • [26].Woolsey SJ, Mansell SE, Kim RB, Tirona RG, Beaton MD. CYP3A activity and expression in nonalcoholic fatty liver disease. Drug Metab Dispos. 2015;43:1484–1490. doi: 10.1124/dmd.115.065979. [DOI] [PubMed] [Google Scholar]
  • [27].Guengerich FP. Cytochrome P-450 3A4: regulation and role in drug metabolism. Annu Rev Pharmacol Toxicol. 1999;39:1–17. doi: 10.1146/annurev.pharmtox.39.1.1. [DOI] [PubMed] [Google Scholar]
  • [28].Finkelstein EA, Trogdon JG, Cohen JW, Dietz W. Annual medical spending attributable to obesity: payer-and service-specific estimates. Health Aff. 2009;28:w822–w831. doi: 10.1377/hlthaff.28.5.w822. [DOI] [PubMed] [Google Scholar]
  • [29].ter Hofstede HJ, de Marie S, Foudraine NA, Danner SA, Brinkman K. Clinical features and risk factors of lactic acidosis following long-term antiretroviral therapy: 4 fatal cases. Int J STD AIDS. 2000;11:611–616. doi: 10.1258/0956462001916498. [DOI] [PubMed] [Google Scholar]
  • [30].Wehling M. Non-steroidal anti-inflammatory drug use in chronic pain conditions with special emphasis on the elderly and patients with relevant comorbidities: management and mitigation of risks and adverse effects. Eur J Clin Pharmacol. 2014;70:1159–1172. doi: 10.1007/s00228-014-1734-6. [DOI] [PubMed] [Google Scholar]
  • [31].Biour M, Ben Salem C, Chazouillères O, Grangé JD, Serfaty L, Poupon R. Drug-induced liver injury: fourteenth updated edition of the bibliographic database of liver injuries and related drugs. Gastroenterol Clin Biol. 2004;28:720–759. doi: 10.1016/s0399-8320(04)95062-2. [DOI] [PubMed] [Google Scholar]
  • [32].Chalasani N, Bonkovsky HL, Fontana R, Lee W, Stolz A, Talwalkar J, Reddy KR, Watkins PB, Navarro V, Barnhart H, Gu J, Serrano J. Features and outcomes of 899 patients with drug-induced liver injury: The DILIN prospective study. Gastroenterology. 2015;148:1340–1352. doi: 10.1053/j.gastro.2015.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Reuben A, Koch DG, Lee WM. Drug-induced acute liver failure: results of a U.S. multicenter, prospective study. Hepatology. 2010;52:2065–2076. doi: 10.1002/hep.23937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Aithal GP, Watkins PB, Andrade RJ, Larrey D, Molokhia M, Takikawa H, Hunt CM, Wilke RA, Avigan M, Kaplowitz N, Bjornsson E, Daly AK. Case definition and phenotype standardization in drug-induced liver injury. Clin Pharmacol Ther. 2011;89:806–815. doi: 10.1038/clpt.2011.58. [DOI] [PubMed] [Google Scholar]
  • [35].Cortez-Pinto H, Chatham J, Chacko VP, Arnold C, Rashid A, Diehl AM. Alterations in liver ATP homeostasis in human nonalcoholic steatohepatitis: a pilot study. JAMA. 1999;282:1659–1664. doi: 10.1001/jama.282.17.1659. [DOI] [PubMed] [Google Scholar]
  • [36].Koliaki C, Roden M. Hepatic energy metabolism in human diabetes mellitus, obesity and non-alcoholic fatty liver disease. Mol Cell Endocrinol. 2013;379:35–42. doi: 10.1016/j.mce.2013.06.002. [DOI] [PubMed] [Google Scholar]
  • [37].Serviddio G, Bellanti F, Vendemiale G. Free radical biology for medicine: learning from nonalcoholic fatty liver disease. Free Radic Biol Med. 2013;65:952–968. doi: 10.1016/j.freeradbiomed.2013.08.174. [DOI] [PubMed] [Google Scholar]
  • [38].Shaw PJ, Hopfensperger MJ, Ganey PE, Roth RA. Lipopolysaccharide and trovafloxacin coexposure in mice causes idiosyncrasy-like liver injury dependent on tumor necrosis factor-alpha. Toxicol Sci. 2007;100:259–266. doi: 10.1093/toxsci/kfm218. [DOI] [PubMed] [Google Scholar]
  • [39].Ramm S, Mally A. Role of drug-independent stress factors in liver injury associated with diclofenac intake. Toxicology. 2013;312:83–96. doi: 10.1016/j.tox.2013.08.002. [DOI] [PubMed] [Google Scholar]
  • [40].Song BJ, Akbar M, Jo I, Hardwick JP, Abdelmegeed MA. Translational implications of the alcohol-metabolizing enzymes, including cytochrome P450-2E1, in alcoholic and non-alcoholic liver disease. Adv Pharmacol. 2015;74:303–372. doi: 10.1016/bs.apha.2015.04.002. [DOI] [PubMed] [Google Scholar]
  • [41].Maes M, Vinken M, Jaeschke H. Experimental models of hepatotoxicity related to acute liver failure. Toxicol Appl Pharmacol. 2016;290:86–97. doi: 10.1016/j.taap.2015.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Du K, Xie Y, McGill MR, Jaeschke H. Pathophysiological significance of c-jun N-terminal kinase in acetaminophen hepatotoxicity. Expert Opin Drug Metab Toxicol. 2015;11:1769–1779. doi: 10.1517/17425255.2015.1071353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Watkins PB, Kaplowitz N, Slattery JT, Colonese CR, Colucci SV, Stewart PW, Harris SC. Aminotransferase elevations in healthy adults receiving 4 grams of acetaminophen daily: a randomized controlled trial. JAMA. 2006;296:87–93. doi: 10.1001/jama.296.1.87. [DOI] [PubMed] [Google Scholar]
  • [44].Forget P, Wittebole X, Laterre PF. Therapeutic dose of acetaminophen may induce fulminant hepatitis in the presence of risk factors: a report of two cases. Br J Anaesth. 2009;103:899–900. doi: 10.1093/bja/aep322. [DOI] [PubMed] [Google Scholar]
  • [45].Savino F, Lupica MM, Tarasco V, Locatelli E, Garazzino S, Tovo PA. Fulminant hepatitis after 10 days of acetaminophen treatment at recommended dosage in an infant. Pediatrics. 2011;127:e494–e497. doi: 10.1542/peds.2010-1965. [DOI] [PubMed] [Google Scholar]
  • [46].Claridge LC, Eksteen B, Smith A, Shah T, Holt AP. Acute liver failure after administration of paracetamol at the maximum recommended daily dose in adults. BMJ. 2010;341:c6764. doi: 10.1136/bmj.c6764. [DOI] [PubMed] [Google Scholar]
  • [47].Neuman MG, French SW, French BA, Seitz HK, Cohen LB, Mueller S, Osna NA, Kharbanda KK, Seth D, Bautista A, Thompson KJ, McKillop IH, Kirpich IA, McClain CJ, Bataller R, Nanau RM, Voiculescu M, Opris M, Shen H, Tillman B, Li J, Liu H, Thomes PG, Ganesan M, Malnick S. Alcoholic and non-alcoholic steatohepatitis. Exp Mol Pathol. 2014;97:492–510. doi: 10.1016/j.yexmp.2014.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Shi EC, Fisher R, McEvoy M, Vantol R, Rose M, Ham JM. Factors influencing hepatic glutathione concentrations: a study in surgical patients. Clin Sci. 1982;62:279–283. doi: 10.1042/cs0620279. [DOI] [PubMed] [Google Scholar]
  • [49].Nguyen GC, Sam J, Thuluvath PJ. Hepatitis C is a predictor of acute liver injury among hospitalizations for acetaminophen overdose in the United States: a nationwide analysis. Hepatology. 2008;48:1336–1341. doi: 10.1002/hep.22536. [DOI] [PubMed] [Google Scholar]
  • [50].Myers RP, Shaheen AA. Hepatitis C, alcohol abuse, and unintentional overdoses are risk factors for acetaminophen-related hepatotoxicity. Hepatology. 2009;49:1399–1400. doi: 10.1002/hep.22798. [DOI] [PubMed] [Google Scholar]
  • [51].Rutherford A, Davern T, Hay JE, Murray NG, Hassanein T, Lee WM, Chung RT. Influence of high body mass index on outcome in acute liver failure. Clin Gastroenterol Hepatol. 2006;4:1544–1549. doi: 10.1016/j.cgh.2006.07.014. [DOI] [PubMed] [Google Scholar]
  • [52].Radosevich JJ, Patanwala AE, Erstad BL. Hepatotoxicity in obese versus nonobese patients with acetaminophen poisoning who are treated with intravenous N-acetylcysteine. Am J Ther 2016. 2016;23:e714–e719. doi: 10.1097/01.mjt.0000434043.62372.00. [DOI] [PubMed] [Google Scholar]
  • [53].Blouin RA, Dickson P, McNamara PJ, Cibull M, McClain C. Phenobarbital induction and acetaminophen hepatotoxicity: resistance in the obese Zucker rodent. J Pharmacol Exp Ther. 1987;243:565–570. [PubMed] [Google Scholar]
  • [54].Aubert J, Begriche K, Delannoy M, Morel I, Pajaud J, Ribault C, Lepage S, McGill MR, Lucas-Clerc C, Turlin B, Robin MA, Jaeschke H, Fromenty B. Differences in early acetaminophen hepatotoxicity between obese ob/ob and db/db mice. J Pharmacol Exp Ther. 2012;342:676–687. doi: 10.1124/jpet.112.193813. [DOI] [PubMed] [Google Scholar]
  • [55].O’Shea D, Davis SN, Kim RB, Wilkinson GR. Effect of fasting and obesity in humans on the 6-hydroxylation of chlorzoxazone: a putative probe of CYP2E1 activity. Clin Pharmacol Ther. 1994;56:359–367. doi: 10.1038/clpt.1994.150. [DOI] [PubMed] [Google Scholar]
  • [56].Lucas D, Farez C, Bardou LG, Vaisse J, Attali JR, Valensi P. Cytochrome P450 2E1 activity in diabetic and obese patients as assessed by chlorzoxazone hydroxylation. Fundam Clin Pharmacol. 1998;12:553–558. doi: 10.1111/j.1472-8206.1998.tb00985.x. [DOI] [PubMed] [Google Scholar]
  • [57].Emery MG, Fisher JM, Chien JY, Kharasch ED, Dellinger EP, Kowdley KV, Thummel KE. CYP2E1 activity before and after weight loss in morbidly obese subjects with nonalcoholic fatty liver disease. Hepatology. 2003;38:428–435. doi: 10.1053/jhep.2003.50342. [DOI] [PubMed] [Google Scholar]
  • [58].Wang Z, Hall SD, Maya JF, Li L, Asghar A, Gorski JC. Diabetes mellitus increases the in vivo activity of cytochrome P450 2E1 in humans. Br J Clin Pharmacol. 2003;55:77–85. doi: 10.1046/j.1365-2125.2003.01731.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Qizilbash AH. Halothane hepatitis. Can Med Assoc J. 1973;108:171–177. [PMC free article] [PubMed] [Google Scholar]
  • [60].Moult PJ, Sherlock S. Halothane-related hepatitis. A clinical study of twenty-six cases. Q J Med. 1975;44:99–114. [PubMed] [Google Scholar]
  • [61].Brunt EM, White H, Marsh JW, Holtmann B, Peters MG. Fulminant hepatic failure after repeated exposure to isoflurane anesthesia: a case report. Hepatology. 1991;13:1017–1021. [PubMed] [Google Scholar]
  • [62].Gunaratnam NT, Benson J, Gandolfi AJ, Chen M. Suspected isoflurane hepatitis in an obese patient with a history of halothane hepatitis. Anesthesiology. 1995;83:1361–1364. doi: 10.1097/00000542-199512000-00030. [DOI] [PubMed] [Google Scholar]
  • [63].Eghtesadi-Araghi P, Sohrabpour A, Vahedi H, Saberi-Firoozi M. Halothane hepatitis in Iran: a review of 59 cases. World J Gastroenterol. 2008;14:5322–5326. doi: 10.3748/wjg.14.5322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Böttiger LE, Dalén E, Hallén B. Halothane-induced liver damage: an analysis of the material reported to the Swedish Adverse Drug Reaction Committee, 1966-1973. Acta Anaesthesiol Scand. 1976;20:40–46. doi: 10.1111/j.1399-6576.1976.tb05007.x. [DOI] [PubMed] [Google Scholar]
  • [65].Kharasch ED, Hankins DC, Cox K. Clinical isoflurane metabolism by cytochrome P450 2E1. Anesthesiology. 1999;90:766–771. doi: 10.1097/00000542-199903000-00019. [DOI] [PubMed] [Google Scholar]
  • [66].Park BK, Kitteringham NR, O’Neill PM. Metabolism of fluorine-containing drugs. Annu Rev Pharmacol Toxicol. 2001;41:443–470. doi: 10.1146/annurev.pharmtox.41.1.443. [DOI] [PubMed] [Google Scholar]
  • [67].Becker GL. Fatty acid lessens halothane’s inhibition of energy metabolism in isolated hepatocytes. Anesth Analg. 1990;70:22–28. doi: 10.1213/00000539-199001000-00005. [DOI] [PubMed] [Google Scholar]
  • [68].Gut J, Christen U, Huwyler J. Mechanisms of halothane toxicity: novel insights. Pharmacol Ther. 1993;58:133–155. doi: 10.1016/0163-7258(93)90047-h. [DOI] [PubMed] [Google Scholar]
  • [69].Kinugawa H, Kawahara M, Amakata Y, Maeda T. Halothane-induced cytotoxicity in isolated rat hepatocytes: an electron microscopic study. In Vivo. 1993;7:497–501. [PubMed] [Google Scholar]
  • [70].Dansette PM, Bonierbale E, Minoletti C, Beaune PH, Pessayre D, Mansuy D. Drug-induced immunotoxicity. Eur J Drug Metab Pharmacokinet. 1998;23:443–451. doi: 10.1007/BF03189993. [DOI] [PubMed] [Google Scholar]
  • [71].Brasil LJ, San-Miguel B, Kretzmann NA, Amaral JL, Zettler CG, Marroni N, Gonzalez-Gallego J, Tunon MJ. Halothane induces oxidative stress and NF-kappaB activation in rat liver: protective effect of propofol. Toxicology. 2006;227:53–61. doi: 10.1016/j.tox.2006.07.013. [DOI] [PubMed] [Google Scholar]
  • [72].Kim H, Oh E, Im H, Mun J, Yang M, Khim JY, Lee E, Lim SH, Kong MH, Lee M, Sul D. Oxidative damages in the DNA, lipids, and proteins of rats exposed to isofluranes and alcohols. Toxicology. 2006;220:169–178. doi: 10.1016/j.tox.2005.12.010. [DOI] [PubMed] [Google Scholar]
  • [73].Bentley JB, Vaughan RW, Gandolfi AJ, Cork RC. Halothane biotransformation in obese and nonobese patients. Anesthesiology. 1982;57:94–97. doi: 10.1097/00000542-198208000-00005. [DOI] [PubMed] [Google Scholar]
  • [74].Young SR, Stoelting RK, Peterson C, Madura JA. Anesthetic biotransformation and renal function in obese patients during and after methoxyflurane or halothane anesthesia. Anesthesiology. 1975;42:451–457. doi: 10.1097/00000542-197504000-00015. [DOI] [PubMed] [Google Scholar]
  • [75].Prey S, Paul C. Effect of folic or folinic acid supplementation on methotrexate-associated safety and efficacy in inflammatory disease: a systematic review. Br J Dermatol. 2009;160:622–628. doi: 10.1111/j.1365-2133.2008.08876.x. [DOI] [PubMed] [Google Scholar]
  • [76].Romao VC, Lima A, Bernardes M, Canhao H, Fonseca JE. Three decades of low-dose methotrexate in rheumatoid arthritis: can we predict toxicity? Immunol Res. 2014;60:289–310. doi: 10.1007/s12026-014-8564-6. [DOI] [PubMed] [Google Scholar]
  • [77].West SG. Methotrexate hepatotoxicity. Rheum Dis Clin North Am. 1997;23:883–915. doi: 10.1016/s0889-857x(05)70365-3. [DOI] [PubMed] [Google Scholar]
  • [78].Rabinowich L, Shibolet O. Drug induced steatohepatitis: an uncommon culprit of a common disease. Biomed Res Int. 2015;2015:168905. doi: 10.1155/2015/168905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Banerjee AK, Lakhani S, Vincent M, Selby P. Dose-dependent acute hepatitis associated with administration of high dose methotrexate. Hum Toxicol. 1988;7:561–562. doi: 10.1177/096032718800700608. [DOI] [PubMed] [Google Scholar]
  • [80].Stine JG, Chalasani N. Chronic liver injury induced by drugs: a systematic review. Liver Int. 2015;35:2343–2353. doi: 10.1111/liv.12958. [DOI] [PubMed] [Google Scholar]
  • [81].Langman G, Hall PM, Todd G. Role of non-alcoholic steato-hepatitis in methotrexate-induced liver injury. J Gastroenterol Hepatol. 2001;16:1395–1401. doi: 10.1046/j.1440-1746.2001.02644.x. [DOI] [PubMed] [Google Scholar]
  • [82].Ramachandran R, Kakar S. Histological patterns in drug-induced liver disease. J Clin Pathol. 2009;62:481–492. doi: 10.1136/jcp.2008.058248. [DOI] [PubMed] [Google Scholar]
  • [83].Wang Y, Lin Z, Liu Z, Harris S, Kelly R, Zhang J, Ge W, Chen M, Borlak J, Tong W. A unifying ontology to integrate histological and clinical observations for drug-induced liver injury. Am J Pathol. 2013;182:1180–1187. doi: 10.1016/j.ajpath.2012.12.033. [DOI] [PubMed] [Google Scholar]
  • [84].Ahern MJ, Smith MD, Roberts-Thomson PJ. Methotrexate hepatotoxicity: what is the evidence? Inflamm Res. 1998;47:148–151. doi: 10.1007/s000110050307. [DOI] [PubMed] [Google Scholar]
  • [85].Rosenberg P, Urwitz H, Johannesson A, Ros AM, Lindholm J, Kinnman N, Hultcrantz R. Psoriasis patients with diabetes type 2 are at high risk of developing liver fibrosis during methotrexate treatment. J Hepatol. 2007;46:1111–1118. doi: 10.1016/j.jhep.2007.01.024. [DOI] [PubMed] [Google Scholar]
  • [86].Malatjalian DA, Ross JB, Williams CN, Colwell SJ, Eastwood BJ. Methotrexate hepatotoxicity in psoriatics: report of 104 patients from Nova Scotia, with analysis of risks from obesity, diabetes and alcohol consumption during long term follow-up. Can J Gastroenterol. 1996;10:369–375. doi: 10.1155/1996/213596. [DOI] [PubMed] [Google Scholar]
  • [87].Kent PD, Luthra HS, Michet C. Risk factors for methotrexate-induced abnormal laboratory monitoring results in patients with rheumatoid arthritis. J Rheumatol. 2004;31:1727–1731. [PubMed] [Google Scholar]
  • [88].Montaudié H, Sbidian E, Paul C, Maza A, Gallini A, Aractingi S, Aubin F, Bachelez H, Cribier B, Joly P, Jullien D, Le Maître M, Misery L, Richard MA, Ortonne JP. Methotrexate in psoriasis: a systematic review of treatment modalities, incidence, risk factors and monitoring of liver toxicity. J Eur Acad Dermatol Venereol. 2011;25(2):12–18. doi: 10.1111/j.1468-3083.2011.03991.x. [DOI] [PubMed] [Google Scholar]
  • [89].Yeo CM, Chong VH, Earnest A, Yang WL. Prevalence and risk factors of methotrexate hepatotoxicity in Asian patients with psoriasis. World J Hepatol. 2013;5:275–280. doi: 10.4254/wjh.v5.i5.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Schmajuk G, Miao Y, Yazdany J, Boscardin WJ, Daikh DI, Steinman MA. Identification of risk factors for elevated transaminases in methotrexate users through an electronic health record. Arthritis Care Res. 2014;66:1159–1166. doi: 10.1002/acr.22294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Zachariae H, Kragballe K, Søgaard H. Methotrexate induced liver cirrhosis. Studies including serial liver biopsies during continued treatment. Br J Dermatol. 1980;102:407–412. doi: 10.1111/j.1365-2133.1980.tb06553.x. [DOI] [PubMed] [Google Scholar]
  • [92].Whiting-O’Keefe QE, Fye KH, Sack KD. Methotrexate and histologic hepatic abnormalities: a meta-analysis. Am J Med. 1991;90:711–716. [PubMed] [Google Scholar]
  • [93].Davila-Fajardo CL, Swen JJ, Cabeza Barrera J, Guchelaar HJ. Genetic risk factors for drug-induced liver injury in rheumatoid arthritis patients using low-dose methotrexate. Pharmacogenomics. 2013;14:63–73. doi: 10.2217/pgs.12.183. [DOI] [PubMed] [Google Scholar]
  • [94].Tarantino G, Conca P, Basile V, Gentile A, Capone D, Polichetti G, Leo E. A prospective study of acute drug-induced liver injury in patients suffering from non-alcoholic fatty liver disease. Hepatol Res. 2007;37:410–415. doi: 10.1111/j.1872-034X.2007.00072.x. [DOI] [PubMed] [Google Scholar]
  • [95].Chalasani N, Aljadhey H, Kesterson J, Murray MD, Hall SD. Patients with elevated liver enzymes are not at higher risk for statin hepatotoxicity. Gastroenterology. 2004;126:1287–1292. doi: 10.1053/j.gastro.2004.02.015. [DOI] [PubMed] [Google Scholar]
  • [96].Bays H, Cohen DE, Chalasani N, Harrison SA. An assessment by the Statin Liver Safety Task Force: 2014 update. J Clin Lipidol. 2014;8:S47–S57. doi: 10.1016/j.jacl.2014.02.011. [DOI] [PubMed] [Google Scholar]
  • [97].Mattar W, Juliar B, Gradus-Pizlo I, Kwo PY. Amiodarone hepatotoxicity in the context of the metabolic syndrome and right-sided heart failure. J Gastrointestin Liver Dis. 2009;18:419–423. [PubMed] [Google Scholar]
  • [98].Tsukamoto H, Towner SJ, Ciofalo LM, French SW. Ethanol-induced liver fibrosis in rats fed high fat diet. Hepatology. 1986;6:814–822. doi: 10.1002/hep.1840060503. [DOI] [PubMed] [Google Scholar]
  • [99].Naveau S, Giraud V, Borotto E, Aubert A, Capron F, Chaput JC. Excess weight risk factor for alcoholic liver disease. Hepatology. 1997;25:108–111. doi: 10.1002/hep.510250120. [DOI] [PubMed] [Google Scholar]
  • [100].Hart CL, Morrison DS, Batty GD, Mitchell RJ, Davey Smith G. Effect of body mass index and alcohol consumption on liver disease: analysis of data from two prospective cohort studies. BMJ. 2010;340:c1240. doi: 10.1136/bmj.c1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Xu J, Lai KK, Verlinsky A, Lugea A, French SW, Cooper MP, Ji C, Tsukamoto H. Synergistic steatohepatitis by moderate obesity and alcohol in mice despite increased adiponectin and p-AMPK. J Hepatol. 2011;55:673–682. doi: 10.1016/j.jhep.2010.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Everitt H, Hu M, Ajmo JM, Rogers CQ, Liang X, Zhang R, Yin H, Choi A, Bennett ES, You M. Ethanol administration exacerbates the abnormalities in hepatic lipid oxidation in genetically obese mice. Am J Physiol Gastrointest Liver Physiol. 2013;304:G38–G47. doi: 10.1152/ajpgi.00309.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Tan TC, Crawford DH, Jaskowski LA, Subramaniam VN, Clouston AD, Crane DI, Bridle KR, Anderson GJ, Fletcher LM. Excess iron modulates endoplasmic reticulum stress-associated pathways in a mouse model of alcohol and high-fat diet-induced liver injury. Lab Invest. 2013;93:1295–1312. doi: 10.1038/labinvest.2013.121. [DOI] [PubMed] [Google Scholar]
  • [104].Massart J, Begriche K, Buron N, Porceddu M, Borgne-Sanchez A, Fromenty B. Drug-induced inhibition of mitochondrial fatty acid oxidation and steatosis. Curr Pathobiol Rep. 2013;1:147–157. [Google Scholar]
  • [105].Mitchell C, Robin MA, Mayeuf A, Mahrouf-Yorgov M, Mansouri A, Hamard M, Couton D, Fromenty B, Gilgenkrantz H. Protection against hepatocyte mitochondrial dysfunction delays fibrosis progression in mice. Am J Pathol. 2009;175:1929–1937. doi: 10.2353/ajpath.2009.090332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Rehman H, Liu Q, Krishnasamy Y, Shi Z, Ramshesh VK, Haque K, Schnellmann RG, Murphy MP, Lemasters JJ, Rockey DC, Zhong Z. The mitochondria-targeted antioxidant MitoQ attenuates liver fibrosis in mice. Int J Physiol Pathophysiol Pharmacol. 2016;8:14–27. [PMC free article] [PubMed] [Google Scholar]
  • [107].Winnike JH, Li Z, Wright FA, Macdonald JM, O’Connell TM, Watkins PB. Use of pharmaco-metabonomics for early prediction of acetaminophen-induced hepatotoxicity in humans. Clin Pharmacol Ther. 2010;88:45–51. doi: 10.1038/clpt.2009.240. [DOI] [PubMed] [Google Scholar]
  • [108].Watelet J, Laurent V, Bressenot A, Bronowicki JP, Larrey D, Peyrin-Biroulet L. Toxicity of chronic paracetamol ingestion. Aliment Pharmacol Ther. 2007;26:1543–1544. doi: 10.1111/j.1365-2036.2007.03503.x. [DOI] [PubMed] [Google Scholar]
  • [109].Trenti T, Bertolotti M, Castellana CN, Ferrari A, Pini LA, Sternieri E. Plasma glutathione level in paracetamol daily abuser patients. Changes in plasma cysteine and thiol groups after reduced glutathione administration. Toxicol Lett. 1992;64-65:757–761. doi: 10.1016/0378-4274(92)90258-l. [DOI] [PubMed] [Google Scholar]
  • [110].Emmett M. Acetaminophen toxicity and 5-oxoproline (pyroglutamic acid): a tale of two cycles, one an ATP-depleting futile cycle and the other a useful cycle. Clin J Am Soc Nephrol. 2014;9:191–200. doi: 10.2215/CJN.07730713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Kato H, Fujigaki Y, Inoue R, Asakawa S, Shin S, Shima T, Furunishi J, Higaki M, Tanemoto M, Yamaguchi Y, Hoshimoto K, Uozaki H, Uchida S. Therapeutic dose of acetaminophen as a possible risk factor for acute kidney injury: learning from two healthy young adult cases. Intern Med. 2014;53:1531–1534. doi: 10.2169/internalmedicine.53.1502. [DOI] [PubMed] [Google Scholar]
  • [112].Sudano I, Flammer AJ, Périat D, Enseleit F, Hermann M, Wolfrum M, Hirt A, Kaiser P, Hurlimann D, Neidhart M, Gay S, Holzmeister J, Nussberger J, Mocharla P, Landmesser U, Haile SR, Corti R, Vanhoutte PM, Lüscher TF, Noll G, Ruschitzka F. Acetaminophen increases blood pressure in patients with coronary artery disease. Circulation. 2010;122:1789–1796. doi: 10.1161/CIRCULATIONAHA.110.956490. [DOI] [PubMed] [Google Scholar]
  • [113].Savransky V, Reinke C, Jun J, Bevans-Fonti S, Nanayakkara A, Li J, Myers AC, Torbenson MS, Polotsky VY. Chronic intermittent hypoxia and acetaminophen induce synergistic liver injury in mice. Exp Physiol. 2009;94:228–239. doi: 10.1113/expphysiol.2008.044883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Mast C, Joly C, Savary-Auzeloux I, Remond D, Dardevet D, Papet I. Skeletal muscle wasting occurs in adult rats under chronic treatment with paracetamol when glutathione-dependent detoxification is highly activated. J Physiol Pharmacol. 2014;65:623–631. [PubMed] [Google Scholar]
  • [115].Mobasher MA, de Toro-Martín J, Gonzalez-Rodríguez A, Ramos S, Letzig LG, James LP, Muntané J, Alvarez C, Valverde AM. Essential role of protein-tyrosine phosphatase 1B in the modulation of insulin signaling by acetaminophen in hepatocytes. J Biol Chem. 2014;289:29406–294019. doi: 10.1074/jbc.M113.539189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Hu J, Ramshesh VK, McGill MR, Jaeschke H, Lemasters JJ. Low dose acetaminophen induces reversible mitochondrial dysfunction associated with transient c-Jun N-terminal kinase activation in mouse liver. Toxicol Sci. 2016;150:204–215. doi: 10.1093/toxsci/kfv319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Kendig EL, Schneider SN, Clegg DJ, Genter MB, Shertzer HG. Over-the-counter analgesics normalize blood glucose and body composition in mice fed a high fat diet. Biochem Pharmacol. 2008;76:216–224. doi: 10.1016/j.bcp.2008.05.001. [DOI] [PubMed] [Google Scholar]
  • [118].Shertzer HG, Schneider SN, Kendig EL, Clegg DJ, D’Alessio DA, Genter M B. Acetaminophen normalizes glucose homeostasis in mouse models for diabetes. Biochem Pharmacol. 2008;75:1402–1410. doi: 10.1016/j.bcp.2007.12.003. [DOI] [PubMed] [Google Scholar]
  • [119].Larrain S, Rinella ME. A myriad of pathways to NASH. Clin Liver Dis. 2012;16:525–548. doi: 10.1016/j.cld.2012.05.009. [DOI] [PubMed] [Google Scholar]
  • [120].Rafacho A, Ortsäter H, Nadal A, Quesada I. Glucocorticoid treatment and endocrine pancreas function: implications for glucose homeostasis, insulin resistance and diabetes. J Endocrinol. 2014;223:R49–R62. doi: 10.1530/JOE-14-0373. [DOI] [PubMed] [Google Scholar]
  • [121].D’souza AM, Beaudry JL, Szigiato AA, Trumble SJ, Snook LA, Bonen A, Giacca A, Riddell MC. Consumption of a high-fat diet rapidly exacerbates the development of fatty liver disease that occurs with chronically elevated glucocorticoids. Am J Physiol Gastrointest Liver Physiol. 2012;302:G850–G863. doi: 10.1152/ajpgi.00378.2011. [DOI] [PubMed] [Google Scholar]
  • [122].Poggioli R, Ueta CB, Drigo RA, Castillo M, Fonseca TL, Bianco AC. Dexamethasone reduces energy expenditure and increases susceptibility to diet-induced obesity in mice. Obesity. 2013;21:E415–E420. doi: 10.1002/oby.20338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Kimberg DV, Loud AV, Wiener J. Cortisone-induced alterations in mitochondrial function and structure. J Cell Biol. 1968;37:63–79. doi: 10.1083/jcb.37.1.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Lettéron P, Brahimi-Bourouina N, Robin MA, Moreau A, Feldmann G, Pessayre D. Glucocorticoids inhibit mitochondrial matrix acyl-CoA dehydrogenases and fatty acid beta-oxidation. Am J Physiol. 1997;272:G1141–G1150. doi: 10.1152/ajpgi.1997.272.5.G1141. [DOI] [PubMed] [Google Scholar]
  • [125].Roussel D, Dumas JF, Simard G, Malthièry Y, Ritz P. Kinetics and control of oxidative phosphorylation in rat liver mitochondria after dexamethasone treatment. Biochem J. 2004;382:491–499. doi: 10.1042/BJ20040696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Fernandez FG, Ritter J, Goodwin JW, Linehan DC, Hawkins WG, Strasberg SM. Effect of steatohepatitis associated with irinotecan or oxaliplatin pretreatment on resectability of hepatic colorectal metastases. J Am Coll Surg. 2005;200:845–853. doi: 10.1016/j.jamcollsurg.2005.01.024. [DOI] [PubMed] [Google Scholar]
  • [127].Vauthey JN, Pawlik TM, Ribero D, Wu TT, Zorzi D, Hoff PM, Xiong HQ, Eng C, Lauwers GY, Mino-Kenudson M, Risio M, Muratore A, Capussotti L, Curley SA, Abdalla EK. Chemo-therapy regimen predicts steatohepatitis and an increase in 90-day mortality after surgery for hepatic colorectal metastases. J Clin Oncol. 2006;24:2065–2072. doi: 10.1200/JCO.2005.05.3074. [DOI] [PubMed] [Google Scholar]
  • [128].Mallick P, Shah P, Gandhi A, Ghose R. Impact of obesity on accumulation of the toxic irinotecan metabolite, SN-38, in mice. Life Sci. 2015;139:132–138. doi: 10.1016/j.lfs.2015.08.017. [DOI] [PubMed] [Google Scholar]
  • [129].Laharie D, Seneschal J, Schaeverbeke T, Doutre MS, Longy-Boursier M, Pellegrin JL, Chabrun E, Villars S, Zerbib F, de Lédinghen V. Assessment of liver fibrosis with transient elastography and FibroTest in patients treated with methotrexate for chronic inflammatory diseases: a case-control study. J Hepatol. 2010;53:1035–1040. doi: 10.1016/j.jhep.2010.04.043. [DOI] [PubMed] [Google Scholar]
  • [130].Kim TY, Kim JY, Sohn JH, Lee HS, Bang SY, Kim Y, Kim MY, Jeong WK. Assessment of substantial liver fibrosis by real-time shear wave elastography in methotrexate-treated patients with rheumatoid arthritis. J Ultrasound Med. 2015;34:1621–1630. doi: 10.7863/ultra.15.14.10035. [DOI] [PubMed] [Google Scholar]
  • [131].Hardwick RN, Clarke JD, Lake AD, Canet MJ, Anumol T, Street SM, Merrell MD, Goedken MJ, Snyder SA, Cherrington NJ. Increased susceptibility to methotrexate-induced toxicity in nonalcoholic steatohepatitis. Toxicol Sci. 2014;142:45–55. doi: 10.1093/toxsci/kfu156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Kyriakides M, Hardwick RN, Jin Z, Goedken MJ, Holmes E, Cherrington NJ, Coen M. Systems level metabolic phenotype of methotrexate administration in the context of non-alcoholic steatohepatitis in the rat. Toxicol Sci. 2014;142:105–116. doi: 10.1093/toxsci/kfu160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Jha P, Knopf A, Koefeler H, Mueller M, Lackner C, Hoefler G, Claudel T, Trauner M. Role of adipose tissue in methionine-choline-deficient model of non-alcoholic steatohepatitis (NASH) Biochim Biophys Acta. 2014;1842:959–970. doi: 10.1016/j.bbadis.2014.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Yamamoto N, Lopes LC, Campello AP, Klüppel ML. Methotrexate: studies on cellular metabolism. II-Effects on mitochondrial oxidative metabolism and ion transport. Cell Biochem Funct. 1989;7:129–134. doi: 10.1002/cbf.290070208. [DOI] [PubMed] [Google Scholar]
  • [135].Schumacher JD, Guo GL. Mechanistic review of drug-induced steatohepatitis. Toxicol Appl Pharmacol. 2015;289:40–47. doi: 10.1016/j.taap.2015.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Belinsky GS, Parke AL, Huang Q, Blanchard K, Jayadev S, Stoll R, Rothe M, Achenie LE, Gupta RR, Wu GY, Rosenberg DW. The contribution of methotrexate exposure and host factors on transcriptional variance in human liver. Toxicol Sci. 2007;97:582–594. doi: 10.1093/toxsci/kfm067. [DOI] [PubMed] [Google Scholar]
  • [137].Sreekumar R, Rosado B, Rasmussen D, Charlton M. Hepatic gene expression in histologically progressive nonalcoholic steatohepatitis. Hepatology. 2003;38:244–251. doi: 10.1053/jhep.2003.50290. [DOI] [PubMed] [Google Scholar]
  • [138].Rensen SS, Slaats Y, Driessen A, Peutz-Kootstra CJ, Nijhuis J, Steffensen R, Greve JW, Buurman WA. Activation of the complement system in human nonalcoholic fatty liver disease. Hepatology. 2009;50:1809–1817. doi: 10.1002/hep.23228. [DOI] [PubMed] [Google Scholar]
  • [139].Hillebrandt S, Wasmuth HE, Weiskirchen R, Hellerbrand C, Keppeler H, Werth A, Schirin-Sokhan R, Wilkens G, Geier A, Lorenzen J, Köhl J, Gressner AM, Matern S, Lammert F. Complement factor 5 is a quantitative trait gene that modifies liver fibrogenesis in mice and humans. Nat Genet. 2005;37:835–843. doi: 10.1038/ng1599. [DOI] [PubMed] [Google Scholar]
  • [140].Schmitt J, Roderfeld M, Sabrane K, Zhang P, Tian Y, Mertens JC, Frei P, Stieger B, Weber A, Müllhaupt B, Roeb E, Geier A. Complement factor C5 deficiency significantly delays the progression of biliary fibrosis in bile duct-ligated mice. Biochem Biophys Res Commun. 2012;418:445–450. doi: 10.1016/j.bbrc.2012.01.036. [DOI] [PubMed] [Google Scholar]
  • [141].Igoudjil A, Begriche K, Pessayre D, Fromenty B. Mitochondrial, metabolic and genotoxic effects of antiretroviral nucleo-side reverse-transcriptase inhibitors. Anti-Infect Agents Med Chem. 2006;5:273–292. [Google Scholar]
  • [142].Nunez M. Clinical syndromes and consequences of antiretroviral-related hepatotoxicity. Hepatology. 2010;52:1143–1155. doi: 10.1002/hep.23716. [DOI] [PubMed] [Google Scholar]
  • [143].Schon E, Fromenty B. Alterations of mitochondrial DNA in liver diseases. In: Kaplowitz N, Han D, editors. Mitochondria in Liver Disease. Taylor & Francis; New-York: 2016. pp. 279–309. [Google Scholar]
  • [144].Tien PC, Barrón Y, Justman JE, Hyman C, Cohen MH, Young M, Kovacs A, Cole SR. Antiretroviral therapies associated with lipoatrophy in HIV-infected women. AIDS Patient Care STDS. 2007;21:297–305. doi: 10.1089/apc.2006.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].de Waal R, Cohen K, Maartens G. Systematic review of antiretroviral-associated lipodystrophy: lipoatrophy, but not central fat gain, is an antiretroviral adverse drug reaction. PLoS One. 2013;8:e63623. doi: 10.1371/journal.pone.0063623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Gougeon ML, Pénicaud L, Fromenty B, Leclercq P, Viard JP, Capeau J. Adipocytes targets and actors in the pathogenesis of HIV-associated lipodystrophy and metabolic alterations. Anti-vir Ther. 2004;9:161–177. [PubMed] [Google Scholar]
  • [147].Tien PC, Schneider MF, Cole SR, Levine AM, Cohen M, DeHovitz J, Young M, Justman JE. Antiretroviral therapy exposure and insulin resistance in the Women’s Interagency HIV study. J Acquir Immune Defic Syndr. 2008;49:369–376. doi: 10.1097/qai.0b013e318189a780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Walker UA, Venhoff N, Koch EC, Olschewski M, Schneider J, Setzer B. Uridine abrogates mitochondrial toxicity related to nucleoside analogue reverse transcriptase inhibitors in HepG2 cells. Antivir Ther. 2003;8:463–470. [PubMed] [Google Scholar]
  • [149].Lebrecht D, Vargas-Infante YA, Setzer B, Kirschner J, Walker UA. Uridine supplementation antagonizes zalcitabine-induced microvesicular steatohepatitis in mice. Hepatology. 2007;45:72–79. doi: 10.1002/hep.21490. [DOI] [PubMed] [Google Scholar]
  • [150].Gardner K, Hall PA, Chinnery PF, Payne BA. HIV treatment and associated mitochondrial pathology: review of 25 years of in vitro, animal, and human studies. Toxicol Pathol. 2014;42:811–822. doi: 10.1177/0192623313503519. [DOI] [PubMed] [Google Scholar]
  • [151].Igoudjil A, Abbey-Toby A, Begriche K, Grodet A, Chataigner K, Peytavin G, Maachi M, Colin M, Robin MA, Lettéron P, Feldmann G, Pessayre D, Fromenty B. High doses of stavudine induce fat wasting and mild liver damage without impairing mitochondrial respiration in mice. Antivir Ther. 2007;12:389–400. [PubMed] [Google Scholar]
  • [152].Igoudjil A, Massart J, Begriche K, Descatoire V, Robin MA, Fromenty B. High concentrations of stavudine impair fatty acid oxidation without depleting mitochondrial DNA in cultured rat hepatocytes. Toxicol In Vitro. 2008;22:887–898. doi: 10.1016/j.tiv.2008.01.011. [DOI] [PubMed] [Google Scholar]
  • [153].Nagiah S, Phulukdaree A, Chuturgoon A. Mitochondrial and oxidative stress response in HepG2 cells following acute and prolonged exposure to antiretroviral drugs. J Cell Biochem. 2015;116:1939–1946. doi: 10.1002/jcb.25149. [DOI] [PubMed] [Google Scholar]
  • [154].Guaraldi G, Squillace N, Stentarelli C, Orlando G, D’Amico R, Ligabue G, Fiocchi F, Zona S, Loria P, Esposito R, Palella F. Nonalcoholic fatty liver disease in HIV-infected patients referred to a metabolic clinic: prevalence, characteristics, and predictors. Clin Infect Dis. 2008;47:250–257. doi: 10.1086/589294. [DOI] [PubMed] [Google Scholar]
  • [155].Ingiliz P, Valantin MA, Duvivier C, Medja F, Dominguez S, Charlotte F, Tubiana R, Poynard T, Katlama C, Lombès A, Benhamou Y. Liver damage underlying unexplained transaminase elevation in human immunodeficiency virus-1 mono-infected patients on antiretroviral therapy. Hepatology. 2009;49:436–442. doi: 10.1002/hep.22665. [DOI] [PubMed] [Google Scholar]
  • [156].Lemoine M, Serfaty L, Capeau J. From nonalcoholic fatty liver to nonalcoholic steatohepatitis and cirrhosis in HIV-infected patients: diagnosis and management. Curr Opin Infect Dis. 2012;25:10–16. doi: 10.1097/QCO.0b013e32834ef599. [DOI] [PubMed] [Google Scholar]
  • [157].Foufelle F, Fromenty B. Role of endoplasmic reticulum stress in drug-induced toxicity. Pharmacol Res Perspect. 2016;4:e00211. doi: 10.1002/prp2.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Stankov MV, Panayotova-Dimitrova D, Leverkus M, Vondran FW, Bauerfeind R, Binz A, Behrens GM. Autophagy inhibition due to thymidine analogues as novel mechanism leading to hepatocyte dysfunction and lipid accumulation. AIDS. 2012;26:1995–2006. doi: 10.1097/QAD.0b013e32835804f9. [DOI] [PubMed] [Google Scholar]
  • [159].Geddes R, Knight S, Moosa MY, Reddi A, Uebel K, Sunpath H. A high incidence of nucleoside reverse transcriptase inhibitor (NRTI)-induced lactic acidosis in HIV-infected patients in a South African context. S Afr Med J. 2006;96:722–724. [PubMed] [Google Scholar]
  • [160].Bolhaar MG, Karstaedt AS. A high incidence of lactic acidosis and symptomatic hyperlactatemia in women receiving highly active antiretroviral therapy in Soweto, South Africa. Clin Infect Dis. 2007;45:254–260. doi: 10.1086/518976. [DOI] [PubMed] [Google Scholar]
  • [161].Wester CW, Okezie OA, Thomas AM, Bussmann H, Moyo S, Muzenda T, Makhema J, van Widenfelt E, Musonda R, Novitsky V, Gaolathe T, Ndwapi N, Essex M, Kuritzkes DR, DeGruttola V, Marlink RG. Higher-than-expected rates of lactic acidosis among highly active antiretroviral therapy-treated women in Botswana: preliminary results from a large randomized clinical trial. J Acquir Immune Defic Syndr. 2007;46:318–322. doi: 10.1097/QAI.0b013e3181568e3f. [DOI] [PubMed] [Google Scholar]
  • [162].Wester CW, Eden SK, Shepherd BE, Bussmann H, Novitsky V, Samuels DC, Hendrickson SL, Winkler CA, O’Brien SJ, Essex M, D’Aquila RT, DeGruttola V, Marlink RG. Risk factors for symptomatic hyperlactatemia and lactic acidosis among combination antiretroviral therapy-treated adults in Botswana: results from a clinical trial. AIDS Res Hum Retroviruses. 2012;28:759–765. doi: 10.1089/aid.2011.0303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Coghlan ME, Sommadossi JP, Jhala NC, Many WJ, Saag MS, Johnson VA. Symptomatic lactic acidosis in hospitalized antiretroviral-treated patients with human immunodeficiency virus infection: a report of 12 cases. Clin Infect Dis. 2001;33:1914–1921. doi: 10.1086/323783. [DOI] [PubMed] [Google Scholar]
  • [164].Thursz MR, Richardson P, Allison M, Austin A, Bowers M, Day CP, Downs N, Gleeson D, MacGilchrist A, Grant A, Hood S, Masson S, McCune A, Mellor J, O’Grady J, Patch D, Ratcliffe I, Roderick P, Stanton L, Vergis N, Wright M, Ryder S, Forrest EH. Prednisolone or pentoxifylline for alcoholic hepatitis. N Engl J Med. 2015;372:1619–1628. doi: 10.1056/NEJMoa1412278. [DOI] [PubMed] [Google Scholar]
  • [165].Nguyen TA, DeShazo JP, Thacker LR, Puri P, Sanyal AJ. The worsening profile of alcoholic hepatitis in the United States. Alcohol Clin Exp Res. 2016;40:1295–1303. doi: 10.1111/acer.13069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Adams LA, Zein CO, Angulo P, Lindor KD. A pilot trial of pentoxifylline in nonalcoholic steatohepatitis. Am J Gastroenterol. 2004;99:2365–2368. doi: 10.1111/j.1572-0241.2004.40064.x. [DOI] [PubMed] [Google Scholar]
  • [167].Satapathy SK, Sakhuja P, Malhotra V, Sharma BC, Sarin SK. Beneficial effects of pentoxifylline on hepatic steatosis, fibrosis and necroinflammation in patients with non-alcoholic steato-hepatitis. J Gastroenterol Hepatol. 2007;22:634–638. doi: 10.1111/j.1440-1746.2006.04756.x. [DOI] [PubMed] [Google Scholar]
  • [168].Du J, Ma YY, Yu CH, Li YM. Effects of pentoxifylline on nonalcoholic fatty liver disease: a meta-analysis. World J Gastroenterol. 2014;20:569–577. doi: 10.3748/wjg.v20.i2.569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Van Wagner LB, Koppe SW, Brunt EM, Gottstein J, Gardikiotes K, Green RM, Rinella ME. Pentoxifylline for the treatment of non-alcoholic steatohepatitis: a randomized controlled trial. Ann Hepatol. 2011;10:277–286. [PubMed] [Google Scholar]
  • [170].Baniasadi N, Salajegheh F, Pardakhty A, Seyedmirzaee SM, Hayatbakhsh MM, Nikpoor AR, Mohammadi M. Effects of pentoxifylline on non-alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled trial in iran. Hepat Mon. 2015;15:e32418. doi: 10.5812/hepatmon.32418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Massart J, Robin MA, Noury F, Fautrel A, Lettéron P, Bado A, Eliat PA, Fromenty B. Pentoxifylline aggravates fatty liver in obese and diabetic ob/ob mice by increasing intestinal glucose absorption and activating hepatic lipogenesis. Br J Pharmacol. 2012;165:1361–1374. doi: 10.1111/j.1476-5381.2011.01580.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Koppe SW, Sahai A, Malladi P, Whitington PF, Green RM. Pentoxifylline attenuates steatohepatitis induced by the methionine choline deficient diet. J Hepatol. 2004;41:592–598. doi: 10.1016/j.jhep.2004.06.030. [DOI] [PubMed] [Google Scholar]
  • [173].Lane JD, Feinglos MN, Surwit RS. Caffeine increases ambulatory glucose and postprandial responses in coffee drinkers with type 2 diabetes. Diabetes Care. 2008;31:221–222. doi: 10.2337/dc07-1112. [DOI] [PubMed] [Google Scholar]
  • [174].Lane JD, Lane AJ, Surwit RS, Kuhn CM, Feinglos MN. Pilot study of caffeine abstinence for control of chronic glucose in type 2 diabetes. J Caffeine Res. 2012;2:45–47. doi: 10.1089/jcr.2012.0003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Wei P, Zhang J, Egan-Hafley M, Liang S, Moore DD. The nuclear receptor CAR mediates specific xenobiotic induction of drug metabolism. Nature. 2000;407:920–923. doi: 10.1038/35038112. [DOI] [PubMed] [Google Scholar]
  • [176].Elcombe CR, Peffer RC, Wolf DC, Bailey J, Bars R, Bell D, Cattley RC, Ferguson SS, Geter D, Goetz A, Goodman JI, Hester S, Jacobs A, Omiecinski CJ, Schoeny R, Xie W, Lake BG. Mode of action and human relevance analysis for nuclear receptor-mediated liver toxicity: A case study with phenobarbital as a model constitutive androstane receptor (CAR) activator. Crit Rev Toxicol. 2014;44:64–82. doi: 10.3109/10408444.2013.835786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Morel G, Cossec B, Lambert AM, Binet S. Evaluation of rat hepatic 2E1 activity in function of age, sex and inducers: choice of an experimental model capable of testing the hepatotoxicity of low molecular weight compounds. Toxicol Lett. 1999;106:171–180. doi: 10.1016/s0378-4274(99)00058-2. [DOI] [PubMed] [Google Scholar]
  • [178].Madan A, Graham RA, Carroll KM, Mudra DR, Burton LA, Krueger LA, Downey AD, Czerwinski M, Forster J, Ribadeneira MD, Gan LS, LeCluyse EL, Zech K, Robertson P, Jr, Koch P, Antonian L, Wagner G, Yu L, Parkinson A. Effects of prototypical microsomal enzyme inducers on cytochrome P450 expression in cultured human hepatocytes. Drug Metab Dispos. 2003;31:421–431. doi: 10.1124/dmd.31.4.421. [DOI] [PubMed] [Google Scholar]
  • [179].Zhao X, Zhang JJ, Wang X, Bu XY, Lou YQ, Zhang GL. Effect of berberine on hepatocyte proliferation, inducible nitric oxide synthase expression, cytochrome P450 2E1 and 1A2 activities in diethylnitrosamine- and phenobarbital-treated rats. Biomed Pharmacother. 2008;62:567–572. doi: 10.1016/j.biopha.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • [180].Zannikos PN, Bandyopadhyay AM, Robertson LW, Blouin RA. Effect of nutritional obesity on the induction of CYP2B enzymes following phenobarbital treatment. Drug Metab Dispos. 1993;21:782–787. [PubMed] [Google Scholar]
  • [181].Zong H, Armoni M, Harel C, Karnieli E, Pessin JE. Cyto-chrome P-450 CYP2E1 knockout mice are protected against high-fat diet-induced obesity and insulin resistance. Am J Physiol Endocrinol Metab. 2012;302:E532–E539. doi: 10.1152/ajpendo.00258.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Abdelmegeed MA, Banerjee A, Yoo SH, Jang S, Gonzalez FJ, Song BJ. Critical role of cytochrome P450 2E1 (CYP2E1) in the development of high fat-induced non-alcoholic steatohepatitis. J Hepatol. 2012;57:860–866. doi: 10.1016/j.jhep.2012.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Chen YY, Zhang CL, Zhao XL, Xie KQ, Zeng T. Inhibition of cytochrome P4502E1 by chlormethiazole attenuated acute ethanol-induced fatty liver. Chem Biol Interact. 2014;222:18–26. doi: 10.1016/j.cbi.2014.08.009. [DOI] [PubMed] [Google Scholar]
  • [184].Abdelmegeed MA, Choi Y, Ha SK, Song BJ. Cytochrome P450-2E1 promotes aging-related hepatic steatosis, apoptosis and fibrosis through increased nitroxidative stress. Free Radic Biol Med. 2016;91:188–202. doi: 10.1016/j.freeradbiomed.2015.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Begriche K, Igoudjil A, Pessayre D, Fromenty B. Mitochondrial dysfunction in NASH: causes, consequences and possible means to prevent it. Mitochondrion. 2006;6:1–28. doi: 10.1016/j.mito.2005.10.004. [DOI] [PubMed] [Google Scholar]
  • [186].Soccio RE, Chen ER, Lazar MA. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab. 2014;20:573–591. doi: 10.1016/j.cmet.2014.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Kohlroser J, Mathai J, Reichheld J, Banner BF, Bonkovsky HL. Hepatotoxicity due to troglitazone: report of two cases and review of adverse events reported to the United States Food and Drug Administration. Am J Gastroenterol. 2000;95:272–276. doi: 10.1111/j.1572-0241.2000.01707.x. [DOI] [PubMed] [Google Scholar]
  • [188].Ikeda T. Drug-induced idiosyncratic hepatotoxicity: prevention strategy developed after the troglitazone case. Drug Metab Pharmacokinet. 2011;26:60–70. doi: 10.2133/dmpk.dmpk-10-rv-090. [DOI] [PubMed] [Google Scholar]
  • [189].Yokoi T. Troglitazone. Handb Exp Pharmacol. 2010;196:419–435. doi: 10.1007/978-3-642-00663-0_14. [DOI] [PubMed] [Google Scholar]
  • [190].Hu D, Wu CQ, Li ZJ, Liu Y, Fan X, Wang QJ, Ding RG. Characterizing the mechanism of thiazolidinedione-induced hepatotoxicity: An in vitro model in mitochondria. Toxicol Appl Pharmacol. 2015;284:134–141. doi: 10.1016/j.taap.2015.02.018. [DOI] [PubMed] [Google Scholar]
  • [191].Al-Salman J, Arjomand H, Kemp DG, Mittal M. Hepatocellular injury in a patient receiving rosiglitazone. A case report. Ann Intern Med. 2000;132:121–124. doi: 10.7326/0003-4819-132-2-200001180-00006. [DOI] [PubMed] [Google Scholar]
  • [192].Forman LM, Simmons DA, Diamond RH. Hepatic failure in a patient taking rosiglitazone. Ann Intern Med. 2000;132:118–121. doi: 10.7326/0003-4819-132-2-200001180-00005. [DOI] [PubMed] [Google Scholar]
  • [193].Nadanaciva S, Dykens JA, Bernal A, Capaldi RA, Will Y. Mitochondrial impairment by PPAR agonists and statins identified via immunocaptured OXPHOS complex activities and respiration. Toxicol Appl Pharmacol. 2007;223:277–287. doi: 10.1016/j.taap.2007.06.003. [DOI] [PubMed] [Google Scholar]
  • [194].Chang E, Park CY, Park SW. Role of thiazolidinediones, insulin sensitizers, in non-alcoholic fatty liver disease. J Diabetes Investig. 2013;4:517–524. doi: 10.1111/jdi.12107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Ratziu V, Charlotte F, Bernhardt C, Giral P, Halbron M, Lenaour G, Hartmann-Heurtier A, Bruckert E, Poynard T. LIDO Study Group. Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: results of the fatty liver improvement by rosiglitazone therapy (FLIRT 2) extension trial. ology. 2010;51:445–453. doi: 10.1002/hep.23270. [DOI] [PubMed] [Google Scholar]
  • [196].Ratziu V. Pharmacological agents for NASH. Nat Rev Gastroenterol Hepatol. 2013;10:676–685. doi: 10.1038/nrgastro.2013.193. [DOI] [PubMed] [Google Scholar]
  • [197].Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, Oliver D, Bacon BR. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-γ ligand rosiglitazone. Hepatology. 2003;38:1008–1017. doi: 10.1053/jhep.2003.50420. [DOI] [PubMed] [Google Scholar]
  • [198].Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, Sponseller CA, Hampton K, Bacon BR. Interim results of a pilot study demonstrating the early effects of the PPAR-γ ligand rosiglitazone on insulin sensitivity, aminotransferases, hepatic steatosis and body weight in patients with non-alcoholic steato-hepatitis. J Hepatol. 2003;38:434–440. doi: 10.1016/s0168-8278(03)00027-8. [DOI] [PubMed] [Google Scholar]
  • [199].Lemoine M, Serfaty L, Cervera P, Capeau J, Ratziu V. Hepatic molecular effects of rosiglitazone in human non-alcoholic steatohepatitis suggest long-term pro-inflammatory damage. Hepatol Res. 2014;44:1241–1247. doi: 10.1111/hepr.12244. [DOI] [PubMed] [Google Scholar]
  • [200].Watkins SM, Reifsnyder PR, Pan HJ, German JB, Leiter EH. Lipid metabolome-wide effects of the PPARγ agonist rosiglitazone. J Lipid Res. 2002;43:1809–1817. doi: 10.1194/jlr.m200169-jlr200. [DOI] [PubMed] [Google Scholar]
  • [201].Pan HJ, Reifsnyder P, Vance DE, Xiao Q, Leiter EH. Pharmacogenetic analysis of rosiglitazone-induced hepatosteatosis in new mouse models of type 2 diabetes. Diabetes. 2005;54:1854–1862. doi: 10.2337/diabetes.54.6.1854. [DOI] [PubMed] [Google Scholar]
  • [202].Garcia-Ruiz I, Rodríguez-Juan C, Díaz-Sanjuán T, Martínez MA, Muñoz-Yagüe T, Solís-Herruzo JA. Effects of rosiglitazone on the liver histology and mitochondrial function in ob/ob mice. Hepatology. 2007;46:414–423. doi: 10.1002/hep.21687. [DOI] [PubMed] [Google Scholar]
  • [203].Zhou M, Xu A, Lam KS, Tam PK, Che CM, Chan L, Lee IK, Wu D, Wang Y. Rosiglitazone promotes fatty acyl CoA accumulation and excessive glycogen storage in livers of mice without adiponectin. J Hepatol. 2010;53:1108–1116. doi: 10.1016/j.jhep.2010.05.034. [DOI] [PubMed] [Google Scholar]
  • [204].Rull A, Geeraert B, Aragonès G, Beltrán-Debón R, Rodríguez-Gallego E, García-Heredia A, Pedro-Botet J, Joven J, Holvoet P, Camps J. Rosiglitazone and fenofibrate exacerbate liver steatosis in a mouse model of obesity and hyperlipidemia. A transcriptomic and metabolomic study. J Proteome Res. 2014;13:1731–1743. doi: 10.1021/pr401230s. [DOI] [PubMed] [Google Scholar]
  • [205].Gao M, Ma Y, Alsaggar M, Liu D. Dual outcomes of rosiglitazone treatment on fatty liver. AAPS J. 2016;18:1023–1031. doi: 10.1208/s12248-016-9919-9. [DOI] [PubMed] [Google Scholar]
  • [206].Yang YJ, Kim KM, An JH, Lee DB, Shim JH, Lim YS, Lee HC, Lee YS, Ahn JH, Jung KH, Kim SB. Clinical significance of fatty liver disease induced by tamoxifen and toremifene in breast cancer patients. Breast. 2016;28:67–72. doi: 10.1016/j.breast.2016.04.017. [DOI] [PubMed] [Google Scholar]
  • [207].Ogawa Y, Murata Y, Nishioka A, Inomata T, Yoshida S. Tamoxifen-induced fatty liver in patients with breast cancer. Lancet. 1998;351:725. doi: 10.1016/S0140-6736(05)78493-2. [DOI] [PubMed] [Google Scholar]
  • [208].Nishino M, Hayakawa K, Nakamura Y, Morimoto T, Mukaihara S. Effects of tamoxifen on hepatic fat content and the development of hepatic steatosis in patients with breast cancer: high frequency of involvement and rapid reversal after completion of tamoxifen therapy. AJR Am J Roentgenol. 2003;180:129–134. doi: 10.2214/ajr.180.1.1800129. [DOI] [PubMed] [Google Scholar]
  • [209].Pratt DS, Knox TA, Erban J. Tamoxifen-induced steatohepatitis. Ann Intern Med. 1995;123:236. doi: 10.7326/0003-4819-123-3-199508010-00018. [DOI] [PubMed] [Google Scholar]
  • [210].Oien KA, Moffat D, Curry GW, Dickson J, Habeshaw T, Mills PR, MacSween RN. Cirrhosis with steatohepatitis after adjuvant tamoxifen. Lancet. 1999;353:36–37. doi: 10.1016/S0140-6736(05)74872-8. [DOI] [PubMed] [Google Scholar]
  • [211].Saphner T, Triest-Robertson S, Li H, Holzman P. The association of nonalcoholic steatohepatitis and tamoxifen in patients with breast cancer. Cancer. 2009;115:3189–3195. doi: 10.1002/cncr.24374. [DOI] [PubMed] [Google Scholar]
  • [212].Elefsiniotis IS, Pantazis KD, Ilias A, Pallis L, Mariolis A, Glynou I, Kada H, Moulakakis A. Tamoxifen induced hepatotoxicity in breast cancer patients with pre-existing liver steatosis: the role of glucose intolerance. Eur J Gastroenterol Hepatol. 2004;16:593–598. doi: 10.1097/00042737-200406000-00013. [DOI] [PubMed] [Google Scholar]
  • [213].Bruno S, Maisonneuve P, Castellana P, Rotmensz N, Rossi S, Maggioni M, Persico M, Colombo A, Monasterolo F, Casadei-Giunchi D, Desiderio F, Stroffolini T, Sacchini V, Decensi A, Veronesi U. Incidence and risk factors for non-alcoholic steatohepatitis: prospective study of 5408 women enrolled in Italian tamoxifen chemoprevention trial. BMJ. 2005;330:932. doi: 10.1136/bmj.38391.663287.E0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Pinto HC, Baptista A, Camilo ME, de Costa EB, Valente A, de Moura MC. Tamoxifen-associated steatohepatitis--report of three cases. J Hepatol. 1995;23:95–97. doi: 10.1016/0168-8278(95)80316-5. [DOI] [PubMed] [Google Scholar]
  • [215].Takamura T, Shimizu A, Komura T, Ando H, Zen Y, Minato H, Matsushita E, Kaneko S. Selective estrogen receptor modulator raloxifene-associated aggravation of nonalcoholic steatohepatitis. Intern Med. 2007;46:579–581. doi: 10.2169/internalmedicine.46.6374. [DOI] [PubMed] [Google Scholar]
  • [216].Larosche I, Lettéron P, Fromenty B, Vadrot N, Abbey-Toby A, Feldmann G, Pessayre D, Mansouri A. Tamoxifen inhibits topoisomerases, depletes mitochondrial DNA, and triggers steatosis in mouse liver. J Pharmacol Exp Ther. 2007;321:526–535. doi: 10.1124/jpet.106.114546. [DOI] [PubMed] [Google Scholar]
  • [217].Tuquet C, Dupont J, Mesneau A, Roussaux J. Effects of tamoxifen on the electron transport chain of isolated rat liver mitochondria. Cell Biol Toxicol. 2000;16:207–219. doi: 10.1023/a:1007695308257. [DOI] [PubMed] [Google Scholar]
  • [218].Cardoso CM, Custódio JB, Almeida LM, Moreno AJ. Mechanisms of the deleterious effects of tamoxifen on mitochondrial respiration rate and phosphorylation efficiency. Toxicol Appl Pharmacol. 2001;176:145–152. doi: 10.1006/taap.2001.9265. [DOI] [PubMed] [Google Scholar]
  • [219].Parvez S, Tabassum H, Rehman H, Banerjee BD, Athar M, Raisuddin S. Catechin prevents tamoxifen-induced oxidative stress and biochemical perturbations in mice. Toxicology. 2006;225:109–118. doi: 10.1016/j.tox.2006.05.009. [DOI] [PubMed] [Google Scholar]
  • [220].Parvez S, Tabassum H, Banerjee BD, Raisuddin S. Taurine prevents tamoxifen-induced mitochondrial oxidative damage in mice. Basic Clin Pharmacol Toxicol. 2008;102:382–387. doi: 10.1111/j.1742-7843.2008.00208.x. [DOI] [PubMed] [Google Scholar]
  • [221].Leone A, Nie A, Brandon Parker J, Sawant S, Piechta LA, Kelley MF, Mark Kao L, Jim Proctor S, Verheyen G, Johnson MD, Lord PG, McMillian MK. Oxidative stress/reactive metabolite gene expression signature in rat liver detects idiosyncratic hepatotoxicants. Toxicol Appl Pharmacol. 2014;275:189–197. doi: 10.1016/j.taap.2014.01.017. [DOI] [PubMed] [Google Scholar]
  • [222].Kakimoto PA, Kowaltowski AJ. Effects of high fat diets on rodent liver bioenergetics and oxidative imbalance. Redox Biol. 2016;8:216–225. doi: 10.1016/j.redox.2016.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Farrell GC. Drug-induced hepatic injury. J Gastroenterol Hepatol. 1997;12:S242–S250. doi: 10.1111/j.1440-1746.1997.tb00507.x. [DOI] [PubMed] [Google Scholar]
  • [224].Fréneaux E, Labbe G, Letteron P, The Le Dinh, Degott C, Genève J, Larrey D, Pessayre D. Inhibition of the mitochondrial oxidation of fatty acids by tetracycline in mice and in man: possible role in microvesicular steatosis induced by this antibiotic. Hepatology. 1988;8:1056–1062. doi: 10.1002/hep.1840080513. [DOI] [PubMed] [Google Scholar]
  • [225].Amacher DE, Martin BA. Tetracycline-induced steatosis in primary canine hepatocyte cultures. Fundam Appl Toxicol. 1997;40:256–263. doi: 10.1006/faat.1997.2389. [DOI] [PubMed] [Google Scholar]
  • [226].Yu HY, Wang BL, Zhao J, Yao XM, Gu Y, Li Y. Protective effect of bicyclol on tetracycline-induced fatty liver in mice. Toxicology. 2009;261:112–118. doi: 10.1016/j.tox.2009.04.058. [DOI] [PubMed] [Google Scholar]
  • [227].Deng Z, Yan S, Hu H, Duan Z, Yin L, Liao S, Sun Y, Yin D, Li G. Proteomic profile of carbonylated proteins in rat liver: discovering possible mechanisms for tetracycline-induced steatosis. Proteomics. 2015;15:148–59. doi: 10.1002/pmic.201400115. [DOI] [PubMed] [Google Scholar]
  • [228].Zhang L, Ging NC, Komoda T, Hanada T, Suzuki T, Watanabe K. Antibiotic susceptibility of mammalian mitochondrial translation. FEBS Lett. 2005;579:6423–6427. doi: 10.1016/j.febslet.2005.09.103. [DOI] [PubMed] [Google Scholar]
  • [229].McKee EE, Ferguson M, Bentley AT, Marks TA. Inhibition of mammalian mitochondrial protein synthesis by oxazolidinones. Antimicrob Agents Chemother. 2006;50:2042–2049. doi: 10.1128/AAC.01411-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Moullan N, Mouchiroud L, Wang X, Ryu D, Williams EG, Mottis A, Jovaisaite V, Frochaux MV, Quiros PM, Deplancke B, Houtkooper RH, Auwerx J. Tetracyclines disturb mitochondrial function across eukaryotic models: a call for caution in biomedical research. Cell Rep. 2017 doi: 10.1016/j.celrep.2015.02.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Ito M, Suzuki J, Sasaki M, Watanabe K, Tsujioka S, Takahashi Y, Gomori A, Hirose H, Ishihara A, Iwaasa H, Kanatani A. Development of nonalcoholic steatohepatitis model through combination of high-fat diet and tetracycline with morbid obesity in mice. Hepatol Res. 2006;34:92–98. doi: 10.1016/j.hepres.2005.12.001. [DOI] [PubMed] [Google Scholar]
  • [232].Labbe G, Fromenty B, Freneaux E, Morzelle V, Letteron P, Berson A, Pessayre D. Effects of various tetracycline derivatives on in vitro and in vivo beta-oxidation of fatty acids, egress of triglycerides from the liver, accumulation of hepatic triglycerides, and mortality in mice. Biochem Pharmacol. 1991;41:638–641. doi: 10.1016/0006-2952(91)90640-q. [DOI] [PubMed] [Google Scholar]
  • [233].Cuenca-Lopez MD, Karachitos A, Massarotto L, Oliveira PJ, Aguirre N, Galindo MF, Kmita H, Jordán J. Minocycline exerts uncoupling and inhibiting effects on mitochondrial respiration through adenine nucleotide translocase inhibition. Pharmacol Res. 2012;65:120–128. doi: 10.1016/j.phrs.2011.08.007. [DOI] [PubMed] [Google Scholar]
  • [234].Lettéron P, Sutton A, Mansouri A, Fromenty B, Pessayre D. Inhibition of microsomal triglyceride transfer protein: another mechanism for drug-induced steatosis in mice. Hepatology. 2003;38:133–140. doi: 10.1053/jhep.2003.50309. [DOI] [PubMed] [Google Scholar]
  • [235].Anthérieu S, Rogue A, Fromenty B, Guillouzo A, Robin MA. Induction of vesicular steatosis by amiodarone and tetracycline is associated with up-regulation of lipogenic genes in HepaRG cells. Hepatology. 2011;53:1895–1905. doi: 10.1002/hep.24290. [DOI] [PubMed] [Google Scholar]
  • [236].Schoepfer AM, Engel A, Fattinger K, Marbet UA, Criblez D, Reichen J, Zimmermann A, Oneta CM. Herbal does not mean innocuous: ten cases of severe hepatotoxicity associated with dietary supplements from Herbalife products. J Hepatol. 2007;47:521–526. doi: 10.1016/j.jhep.2007.06.014. [DOI] [PubMed] [Google Scholar]
  • [237].Chalasani N, Fontana RJ, Bonkovsky HL, Watkins PB, Davern T, Serrano J, Yang H, Rochon J. Causes, clinical features, and outcomes from a prospective study of drug-induced liver injury in the United States. Gastroenterology. 2008;135:1924–1934. doi: 10.1053/j.gastro.2008.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Seeff LB, Bonkovsky HL, Navarro VJ, Wang G. Herbal products and the liver: a review of adverse effects and mechanisms. Gastroenterology. 2015;148:517–532. doi: 10.1053/j.gastro.2014.12.004. [DOI] [PubMed] [Google Scholar]
  • [239].Sorrentino P, Tarantino G, Conca P, Perrella A, Terracciano ML, Vecchione R, Gargiulo G, Gennarelli N, Lobello R. Silent non-alcoholic fatty liver disease-a clinical-histological study. J Hepatol. 2004;41:751–757. doi: 10.1016/j.jhep.2004.07.010. [DOI] [PubMed] [Google Scholar]
  • [240].Marchesini G, Moscatiello S, Di Domizio S, Forlani G. Obesity-associated liver disease. J Clin Endocrinol Metab. 2008;93:S74–S80. doi: 10.1210/jc.2008-1399. [DOI] [PubMed] [Google Scholar]
  • [241].Palekar NA, Naus R, Larson SP, Ward J, Harrison SA. Clinical model for distinguishing nonalcoholic steatohepatitis from simple steatosis in patients with nonalcoholic fatty liver disease. Liver Int. 2006;26:151–156. doi: 10.1111/j.1478-3231.2005.01209.x. [DOI] [PubMed] [Google Scholar]
  • [242].Fierbinteanu-Braticevici C, Dina I, Petrisor A, Tribus L, Negreanu L, Carstoiu C. Noninvasive investigations for non alcoholic fatty liver disease and liver fibrosis. World J Gastroenterol. 2010;16:4784–4791. doi: 10.3748/wjg.v16.i38.4784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Sgro C, Clinard F, Ouazir K, Chanay H, Allard C, Guilleminet C, Lenoir C, Lemoine A, Hillon P. Incidence of drug-induced hepatic injuries: a French population-based study. Hepatology. 2002;36:451–455. doi: 10.1053/jhep.2002.34857. [DOI] [PubMed] [Google Scholar]
  • [244].Donato MT, Jiménez N, Serralta A, Mir J, Castell JV, Gómez-Lechón MJ. Effects of steatosis on drug-metabolizing capability of primary human hepatocytes. Toxicol In Vitro. 2007;21:271–276. doi: 10.1016/j.tiv.2006.07.008. [DOI] [PubMed] [Google Scholar]
  • [245].Tirosh O, Ilan E, Anavi S, Ramadori G, Madar Z. Nutritional lipid-induced oxidative stress leads to mitochondrial dysfunction followed by necrotic death in FaO hepatocytes. Nutrition. 2009;25:200–208. doi: 10.1016/j.nut.2008.07.023. [DOI] [PubMed] [Google Scholar]
  • [246].Janorkar AV, King KR, Megeed Z, Yarmush ML. Development of an in vitro cell culture model of hepatic steatosis using hepatocyte-derived reporter cells. Biotechnol Bioeng. 2009;102:1466–1474. doi: 10.1002/bit.22191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Luo Y, Rana P, Will Y. Palmitate increases the susceptibility of cells to drug-induced toxicity: an in vitro method to identify drugs with potential contraindications in patients with metabolic disease. Toxicol Sci. 2012;129:346–362. doi: 10.1093/toxsci/kfs208. [DOI] [PubMed] [Google Scholar]
  • [248].Kim MS, Lee KT, Iseli TJ, Hoy AJ, George J, Grewal T, Roufogalis BD. Compound K modulates fatty acid-induced lipid droplet formation and expression of proteins involved in lipid metabolism in hepatocytes. Liver Int. 2013;33:1583–1593. doi: 10.1111/liv.12287. [DOI] [PubMed] [Google Scholar]
  • [249].Chang JJ, Hsu MJ, Huang HP, Chung DJ, Chang YC, Wang CJ. Mulberry anthocyanins inhibit oleic acid induced lipid accumulation by reduction of lipogenesis and promotion of hepatic lipid clearance. J Agric Food Chem. 2013;61:6069–6076. doi: 10.1021/jf401171k. [DOI] [PubMed] [Google Scholar]
  • [250].Park MJ, Kim DI, Choi JH, Heo YR, Park SH. New role of irisin in hepatocytes: The protective effect of hepatic steatosis in vitro. Cell Signal. 2015;27:1831–1839. doi: 10.1016/j.cellsig.2015.04.010. [DOI] [PubMed] [Google Scholar]
  • [251].Garcia-Ruiz I, Solís-Muñoz P, Fernández-Moreira D, Muñoz-Yagüe T, Solís-Herruzo JA. In vitro treatment of HepG2 cells with saturated fatty acids reproduces mitochondrial dysfunction found in nonalcoholic steatohepatitis. Dis Model Mech. 2015;8:183–191. doi: 10.1242/dmm.018234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Siculella L, Tocci R, Rochira A, Testini M, Gnoni A, Damiano F. Lipid accumulation stimulates the cap-independent translation of SREBP-1a mRNA by promoting hnRNP A1 binding to its 5’-UTR in a cellular model of hepatic steatosis. Biochim Biophys Acta. 2016;1861:471–481. doi: 10.1016/j.bbalip.2016.02.003. [DOI] [PubMed] [Google Scholar]
  • [253].Porceddu M, Buron N, Roussel C, Labbe G, Fromenty B, Borgne-Sanchez A. Prediction of liver injury induced by chemicals in human with a multiparametric assay on isolated mouse liver mitochondria. Toxicol Sci. 2012;129:332–345. doi: 10.1093/toxsci/kfs197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [254].Finsterer J, Segall L. Drugs interfering with mitochondrial disorders. Drug Chem Toxicol. 2010;33:138–151. doi: 10.3109/01480540903207076. [DOI] [PubMed] [Google Scholar]
  • [255].Hynes J, Nadanaciva S, Swiss R, Carey C, Kirwan S, Will Y. A high-throughput dual parameter assay for assessing drug- induced mitochondrial dysfunction provides additional predictivity over two established mitochondrial toxicity assays. Toxicol In Vitro. 2013;27:560–569. doi: 10.1016/j.tiv.2012.11.002. [DOI] [PubMed] [Google Scholar]
  • [256].Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology. 2010;52:774–788. doi: 10.1002/hep.23719. [DOI] [PubMed] [Google Scholar]
  • [257].Duwaerts CC, Maher JJ. Mechanisms of liver injury in non-alcoholic steatohepatitis. Curr Hepatol Rep. 2014;13:119–129. doi: 10.1007/s11901-014-0224-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Lavallard VJ, Gual P. Autophagy and non-alcoholic fatty liver disease. Biomed Res Int. 2014;2014:120179. doi: 10.1155/2014/120179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Anstee QM, Day CP. The genetics of nonalcoholic fatty liver disease: spotlight on PNPLA3 and TM6SF2. Semin Liver Dis. 2015;35:270–290. doi: 10.1055/s-0035-1562947. [DOI] [PubMed] [Google Scholar]
  • [260].Trépo E, Romeo S, Zucman-Rossi J, Nahon P. PNPLA3 gene in liver diseases. J Hepatol. 2016;65:399–412. doi: 10.1016/j.jhep.2016.03.011. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Clinical and Translational Research are provided here courtesy of Whioce Publishing Pte. Ltd.

RESOURCES