Liver International ISSN 1478-3223

REVIEW ARTICLE

Acetaminophen-induced liver injury in obesity and nonalcoholic fatty liver disease Ana€ıs Michaut1, Caroline Moreau1,2, Marie-Anne Robin1 and Bernard Fromenty1 1 INSERM, U991, Universit e de Rennes 1, Rennes, France 2 Laboratoire de Biochimie et Toxicologie, CHU Pontchaillou, Rennes, France

Keywords acetaminophen – hepatotoxicity – obesity – fatty liver – steatosis – cytochrome P450 2E1

Correspondence Dr Bernard Fromenty, INSERM UMR991, Facult e de Pharmacie. 2, avenue du Professeur L eon Bernard, 35043 Rennes Cedex, France Tel: +33 2 23 23 30 44 Fax: +33 2 23 23 53 85 e-mail: [email protected] Received 9 January 2014 Revised 10 February 2014 Accepted 23 February 2014 DOI:10.1111/liv.12514

Abstract Although acetaminophen (APAP) is usually considered as a safe drug, this painkiller can lead to acute liver failure after overdoses. Moreover, there is evidence that the maximum recommended dosage can induce hepatic cytolysis in some individuals. Several predisposing factors appear to enhance the risk and severity of APAP-induced liver injury including chronic alcoholic liver disease and nonalcoholic fatty liver disease (NAFLD), which refers to a large spectrum of hepatic lesions linked to obesity. In contrast, obesity by itself does not seem to be associated with a higher risk of APAP-induced liver injury. Since 1987, seven studies dealt with APAP-induced hepatotoxicity in rodent models of NAFLD and five of them found that this liver disease was associated with higher APAP toxicity. Unfortunately, these studies did not unequivocally established the mechanism(s) whereby NAFLD could favour APAP hepatotoxicity, although some investigations suggested that pre-existent induction of hepatic cytochrome P450 2E1 (CYP2E1) could play a significant role by increasing the generation of N-acetyl-p-benzoquinone imine (NAPQI), the toxic metabolite of APAP. Moreover, pre-existent mitochondrial dysfunction associated with NAFLD could also be involved. In contrast, some investigations suggested that factors that could reduce the risk and severity of APAP hepatotoxicity in obesity and NAFLD include higher hepatic APAP glucuronidation, reduced CYP3A4 activity and increased volume of body distribution. Thus, the occurrence and the outcome of APAP-induced liver injury in an obese individual with NAFLD might depend on a delicate balance between metabolic factors that can be protective and others that favour large hepatic levels of NAPQI.

Acetaminophen (APAP), also known as paracetamol, is one of the most widely prescribed drugs for the management of pain and hyperthermia, even though its mode of action is still under investigation (1). The current maximum recommended dosage of APAP is 4 g/day in adults and 60–75 mg/kg/day in children. Although APAP is usually considered as a safe drug, APAP intoxication after overdosage can lead to massive hepatocellular necrosis and acute liver failure (2, 3). At the moment, N-acetylcysteine is the only approved drug to treat APAP-induced liver injury in patients after voluntary, or unintentional, APAP overdose (4). Importantly, there is accumulating evidence that the maximum recommended dose can induce mild-to-moderate hepatic cytolysis, even in healthy individuals (5, 6). It is also noteworthy that long-term APAP ingestion can induce chronic liver injury in some patients, such as granulomatous hepatitis and cirrhosis (7, 8). Different predisposing factors are known, or suspected, to significantly enhance the risk and severity of Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

APAP-induced hepatotoxicity. These factors include malnutrition and fasting, chronic alcohol abuse and alcoholic liver disease, and hepatitis C virus infection (9–11). Another factor could be comedication with drugs inducing cytochromes P450 (CYPs), such as isoniazid, rifampicin and phenobarbital (12, 13), or with drugs which may modulate liver injury and/or repair (14). The risk of APAP-induced liver injury could also be enhanced by some gene polymorphisms, in particular in genes encoding APAP metabolizing enzymes (15, 16). Lastly, a large retrospective study also reported that obesity-related nonalcoholic fatty liver disease (NAFLD) could be a risk factor for APAP-induced acute liver injury after APAP overdose (9). Nonalcoholic fatty liver disease refers to the large spectrum of liver lesions in obese individuals ranging from fatty liver to nonalcoholic steatohepatitis (NASH) and cirrhosis. In some countries such as the United States, it is estimated that the prevalence of NAFLD in the general population could be about 20% in adults

1

Michaut et al.

Acetaminophen hepatotoxicity in obesity

and 10% in children and adolescents (17, 18). Actually, a majority of overweight and obese people develop fatty liver, which is characterized by the presence of large vacuoles of lipids (mainly triacylglycerol) within the hepatocytes. During obesity, a major mechanism leading to hepatic accretion of lipids is insulin resistance, which favours the redistribution of fat from adipose tissue to the liver and higher de novo lipogenesis in hepatocytes (19, 20). Importantly, fatty liver can progress in the long term to NASH in 10–20% of patients. NASH is characterized not only by steatosis but also by necroinflammation (with ballooning degeneration of hepatocytes), some fibrosis and the presence of apoptotic hepatocytes. Key factors involved in the pathogenesis of NASH include oxidative stress, lipid peroxidation, mitochondrial dysfunction and overproduction of proinflammatory and profibrotic cytokines (20, 21). Impaired activity of the mitochondrial respiratory chain and induction of CYP2E1 are two major factors leading to reactive oxygen species (ROS) overproduction during NASH (20, 22). As previously mentioned, clinical investigations reported that NAFLD could favour APAP-induced acute liver failure after an overdose. Moreover, studies were performed in different animal models of obesity and NAFLD to find out whether these metabolic diseases could favour APAP-induced liver injury. To find out the clinical and experimental studies dealing with the topic of this review, we performed a PubMed search of literature published in the English language using the combination of at least two of the following queries: acetaminophen, obesity (or obese), liver, ‘fatty liver’, NASH, injury and ‘risk factors’. In the present article, the main findings of these clinical and experimental investigations will be presented and subsequently discussed. Prior to these sections, some major features of APAP metabolism and liver toxicity will be recalled. Main features of APAP metabolism and hepatotoxicity

This section will only summarize the current knowledge about APAP metabolism and mechanisms of hepatotoxicity as several comprehensive reviews were recently published on these topics (23–26). Briefly, APAP is mainly metabolized in the liver by phase II conjugating enzymes into the nontoxic glucuronide and sulphate conjugates, which respectively represent about 55% and 30–40% of the initial APAP dose (24, 27). In addition, a small amount of APAP is oxidized to the highly reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI) by several CYPs, especially by CYP2E1 and CYP3A4 (28, 29). There is strong evidence that CYP2E1 is the main enzyme generating NAPQI in mice (29, 30), but several investigations suggested that CYP3A4 could also be an important enzyme catalysing NAPQI generation in human (31–33). Importantly, obesity and related metabolic disorders are associated with significant alterations of APAP pharmacokinetics. In particular, increased

2

hepatic activity of CYP2E1 and higher APAP glucuronidation have consistently been reported in obesity and NAFLD (22, 34–36), although the exact mechanisms of these alterations are still poorly understood. In contrast, hepatic activity of CYP3A4 is reduced in obesity and NAFLD (36, 37). After its formation, NAPQI is rapidly detoxified by hepatic reduced glutathione (GSH) when APAP is taken at the recommended dosage. However, after APAP overdose, high levels of NAPQI can induce several detrimental effects within the liver. Indeed, hepatocellular GSH stocks are quickly depleted and once GSH is no longer available for NAPQI detoxication, this reactive metabolite binds to different macromolecules, including cytosolic and mitochondrial proteins (38, 39). These early cellular events are then followed by c-jun N-terminal kinase (JNK) activation, profound mitochondrial dysfunction and ATP depletion, overproduction of ROS and peroxynitrite and massive hepatocellular necrosis (23, 26, 40). Finally, this initial liver injury is followed by other key events such as inflammation, repair and regeneration that play a major role in the outcome of APAP-induced hepatotoxicity (23, 25, 41). Data from clinical investigations

As already mentioned in the introduction, a large retrospective study reported an increased risk of APAPinduced acute liver injury after APAP overdose in patients with NAFLD (9). In this study, patients with pre-existent NAFLD and hospitalized for APAP overdose had more than a seven-fold higher prevalence of acute liver injury as compared to those without NAFLD (9). In a subsequent report, Myers and Shaheen reanalysed the same database by taking into account potential differences in overdose circumstance, which could be a significant confounding factor (42). This reanalysis supported the previous findings of a higher risk of APAPinduced hepatotoxicity in patients with NAFLD (9), even though the magnitude of this risk was attenuated (i.e. 3.91 vs. 7.43). Importantly, both studies showed that the risk of severe acute liver injury after APAP overdose was increased about similarly with NAFLD and alcoholic liver disease (9, 42). In contrast, two studies reported that the susceptibility to APAP-induced acute liver injury was similar, or lower, in obese patients compared to nonobese individuals, but NAFLD was unfortunately not taken into consideration in these investigations (43, 44). Interestingly, in one of these studies, obese patients had significantly poorer outcomes after acute liver failure (43). Altogether, these clinical investigations suggest that NAFLD, rather than obesity by itself, could increase the risk of APAPinduced acute liver injury after APAP overdose. This important point will be discussed later on. However, it is noteworthy that the discordant results between the aforementioned studies on the association between NAFLD/obesity and APAP-induced acute liver injury could Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Michaut et al.

Acetaminophen hepatotoxicity in obesity

be caused by differences in the criteria adopted to define these conditions in patients. As previously mentioned, there is evidence that prescription of APAP at the recommended dosage can induce mild-to-moderate hepatic cytolysis, even in healthy individuals (5, 6). However, it is still unclear whether NAFLD could enhance the risk, or the severity, of liver injury after therapeutic dose of APAP, although this has been reported in some obese patients (45). Data from experimental studies

Different rodent models of obesity and NAFLD can be used experimentally, in particular in investigations dealing with the hepatotoxic effects of chemicals such as APAP (Table 1). Classically, obesity and related metabolic disorders can be induced by different types of high-calorie diets (including high-fat and high-sucrose/ fructose diets), by genetic defects altering different key physiological functions such as food intake and energy expenditure, or by the combination of nutritional and genetic approaches (46–50). Among the more commonly used murine models of genetic obesity, the

C57BL/6J-ob/ob (ob/ob) are characterized by a mutation in the leptin gene. Indeed, lack of the adipokine leptin leads to increased food consumption and lower energy expenditure, thus resulting in massive obesity, insulin resistance, dyslipidaemia and severe fatty liver (51, 52). C57BL/KsJ-db/db mice and Zucker fa/fa rats present a mutation in the gene encoding leptin receptor, which disrupts leptin signalling and induces obesity, insulin resistance and fatty liver (46, 52). The KK-Ay mice, which present a defect in the signalling of the melanocortin receptors, spontaneously develop obesity, fatty liver, type 2 diabetes, arterial hypertension and diabetic nephropathy (46, 52, 53). These genetic rodent models of obesity are associated with metabolic disorders that are more severe compared to the nutritional models, but usually with less variability between the animals. From 1987 to 2012, six different studies reported the effect of acute APAP intoxication in several animal models of obesity and NAFLD (54–59). The main data from these investigations are presented in Table 1. In addition, one study reported APAP hepatotoxicity in a murine model of methionine and choline deficient

Table 1. Studies reporting APAP-induced acute liver injury in different animal models of NAFLD References

Animal models of NAFLD

Presence of NAFLD

Dose of APAP

Response to APAP toxicity

CYP2E1 activity

Corcoran and Wong (54)

Male Sprague-Dawley rats fed with a high-fat diet for 24 weeks Obese male Zucker fa/fa rats Male C57Bl/6 mice fed a ‘Western-style’ diet for 16 weeks Male ob/ob mice

Not reported in this study*

710 mg/kg (i.p.)

Not reported in this study*

Not reported in this study† Yes

1300 mg (p.o.) 300 mg/kg (p.o.)

Not reported in this study‡

300 mg/kg (p.o.) 300 or 600 mg/kg (p.o.) 1 g/kg (p.o)

Higher toxicity after 48 h, compared to rats fed a standard diet Similar toxicity after 48 h, compared to lean rats Lower toxicity after 6 h, compared to mice fed a standard diet Lower toxicity after 6 h, compared to wild-type mice Higher toxicity after 6 h, compared to wild-type mice Higher toxicity after 24 and 48 h, compared to rats fed a standard diet Higher toxicity after 8 h, compared to wild-type mice Similar toxicity after 8 h, compared to wild-type mice Higher toxicity from 6 to 48 h after overdose, compared to mice fed a standard diet

Blouin et al. (55) Ito et al. (56)

Kon et al. (57)

Male KK-Ay mice

Yes

Kucera et al. (58)

Male Sprague-Dawley rats fed a high-fat diet for 6 weeks Female db/db mice

Yes

Yes

500 mg/kg (p.o.)

Female ob/ob mice

Yes

500 mg/kg (p.o.)

Male Swiss Webster mice fed a MCD diet for 1 month

Yes

360 mg/kg (i.p.)

Aubert et al. (59)

Donthamsetty et al. (60)

Not reported in this study† Not reported in this study* Not reported in this study‡ Not reported in this study§ Not reported in this study* Increased

Unchanged

Unchanged

*Different investigations in rats showed that long-term feeding with high-fat diets induce NAFLD (reviewed in 46 and 49) and increase the expression and/or activity of CYP2E1 (reviewed in 22). †Other studies showed that male obese and insulin resistant Zucker fa/fa rats present moderate fatty liver (106, 107), but reduced CYP2E1 activity (106, 108). ‡Other studies showed that male obese and diabetic ob/ob mice present major fatty liver (50, 104, 109), with unchanged (59) or reduced (67) CYP2E1 activity. §One study showed that male KK-Ay mice present unchanged hepatic mRNA expression of CYP2E1 (110).

Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

3

Acetaminophen hepatotoxicity in obesity

(MCD) diet-induced steatohepatitis (Table 1) (60). However, it must be pointed out that the MCD diet consistently leads to a dramatic loss of body weight (61– 63) and does not induce systemic insulin resistance (63, 64). It is also noteworthy that the doses of APAP greatly varied in these seven studies, with the highest doses used in the rat models (Table 1). This is because rats are much more resistant to APAP-induced liver injury compared to mice (65, 66). Among these different studies (Table 1), four of them showed that NAFLD was associated with higher liver injury after a single APAP overdose (57–60). Another study also showed higher APAP toxicity in rats fed a high-fat diet (54), although the presence of NAFLD can only be suspected in this study (Table 1). In contrast, the remaining investigations showed similar (55) or lower liver injury (56). Unfortunately, there is no definite explanation for these discrepancies, although some hypotheses can be put forward. For instance, investigations in male and female ob/ob and db/db mice showed that the hepatic CYP2E1 status could play a significant role (59). Indeed, higher APAP hepatotoxicity was observed in female db/db mice but not in female ob/ob mice and this was associated with a specific induction of CYP2E1 activity in the first group of animals. Interestingly, hepatic CYP2E1 activity was unchanged (59) or reduced (67) in male ob/ob mice and this could explain why Ito and collaborators reported lower acute APAP hepatotoxicity in this model (56). It is also noteworthy that plasma levels of APAP-glucuronide and APAP-sulphate were almost similar between female db/db mice and female ob/ob mice after 500 mg/kg of APAP (59). Hence, the observed difference of APAP-induced hepatotoxicity between these murine models cannot be attributed to APAP glucuronidation and sulphation. At the moment, it is still unclear why hepatic CYP2E1 is specifically induced in female but not in male db/db mice (59). Although there was an overall positive correlation between hepatic CYP2E1-mediated aniline hydroxylase activity and plasma glucose in all groups of untreated mice (i.e. wild-type, ob/ob and db/db of both sexes), plasma glucose levels were not different between male and female db/db mice (59). Thus, hepatic CYP2E1 induction in female db/db mice may not be explained by the severity of insulin resistance and type 2 diabetes in these animals, contrary to what has been suggested by some investigations (22). In addition, there was no difference in plasma b-hydroxybutyrate levels between female and male db/db mice (59). This suggests that ketone bodies may not be responsible for CYP2E1 induction in female db/db mice, although previous investigations found a significant correlation between plasma b-hydroxybutyrate levels and hepatic CYP2E1 activity (34, 68). Thus, further investigations will be needed to determine the exact mechanism of higher hepatic CYP2E1 activity in female db/db mice. Besides differences in APAP biotransformation to NAPQI, higher susceptibility to APAP-induced liver

4

Michaut et al.

injury might be explained by lipid accumulation and oxidative stress associated with NAFLD. Regarding lipid accretion, it is noteworthy that higher APAP hepatotoxicity in female db/db mice compared to female ob/ob mice was observed despite lower lipid accumulation in db/db liver (59). Importantly, the basal plasma levels of ALT and AST (i.e. before APAP intoxication) were comparable between female db/db and female ob/ob mice (59). Thus, increased susceptibility to APAP hepatotoxicity in female db/db mice cannot be attributed to the pre-existence of higher hepatic cytolysis. However, further studies will be required to find out whether the accumulation of some lipid species could be involved. Indeed, it is noteworthy that db/db hepatocytes present more microvesicular steatosis and less macrovacuolar steatosis compared to ob/ob hepatocytes (50, 59). Because the size of the droplets is greatly dependent on their lipid composition (69, 70), it is possible that the accumulation of some specific lipid species in female db/db hepatocytes could have played a role in the higher susceptibility to APAP toxicity. Nonalcoholic fatty liver disease is associated with oxidative stress, which can alter different cellular components such as nucleic acids and unsaturated fatty acids (20, 71, 72). Moreover, hepatic GSH reserves could be lowered in NAFLD, thus reducing the antioxidant capacity of the fatty hepatocytes (20, 73). Hence, the higher susceptibility to APAP hepatotoxicity observed in some rodent models of obesity might be related to basal oxidative stress and lower GSH levels. Yet, basal hepatic GSH levels were not significantly reduced in two rodent models of obesity showing higher vulnerability to APAP toxicity (58, 59). It is however noteworthy that total GSH levels were assessed in these studies and thus it will be important to determine whether mitochondrial GSH was specifically depleted. Indeed, some investigations suggested a key role of the mitochondrial pool of GSH in APAP-induced liver injury (74, 75). As previously mentioned, JNK activation plays a major role in APAP-induced hepatotoxicity (23, 26, 40). Interestingly, JNK activation could also play a significant role in NAFLD pathogenesis (76, 77). Only two studies examined the hepatic JNK status in obese animals intoxicated by APAP (57, 59). In the first study, JNK activation (as assessed by phospho-JNK) after APAP intoxication was significantly stronger in obese and diabetic KK-Ay mice compared to wild-type mice, and this was associated to higher APAP hepatotoxicity in the obese animals (57). In the second study, increased APAP hepatotoxicity in db/db mice was not associated to higher JNK phosphorylation (59). Unexpectedly, hepatic JNK was specifically activated in ob/ob mice although these animals did not present higher APAP liver injury (59). Thus, JNK activation might not be a key mechanism of APAP hepatotoxicity in some murine models of NAFLD. Further investigations will be needed to assess phospho-JNK not only in whole liver but also in isolated mitochondria (78). Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Michaut et al.

Acetaminophen hepatotoxicity in obesity

In addition to the investigations performed in rodent models of obesity, one study reported the effect of acute APAP intoxication in a murine model of MCD dietinduced steatohepatitis (Table 1) (60). In this study, higher APAP hepatotoxicity was observed despite the lack of pre-existent CYP2E1 induction and the presence of normal basal hepatic GSH levels (60), suggesting the involvement of other mechanism(s). Interestingly, the basal hepatic ATP levels were already reduced by 45% in mice fed the MCD diet (60), which is consistent with a significant impairment of the mitochondrial respiratory chain and oxidative phosphorylation in this experimental model (79, 80). Hence, severe pre-existent mitochondrial dysfunction might have played a role in the higher APAP hepatotoxicity observed in mice fed the MCD diet.

obesity and NAFLD (36, 37, 90, 91), and this may reduce the generation of NAPQI. In addition, obesity could lead to reduced APAP gastrointestinal absorption rate and higher APAP volume of distribution, thus favouring lower APAP plasma concentrations (92, 93). Thus, it appears that the occurrence of APAP-induced liver injury in an obese individual will depend on a delicate balance between metabolic factors that can be protective (higher APAP glucuronidation and volume of distribution, lower absorption rate and CYP3A4 activity) and others that can augment the hepatic production of NAPQI (CYP2E1 induction), or reduce the detoxication of this highly reactive metabolite (lower GSH stores) (Fig. 1).

General discussion

APAP-induced liver injury in obesity and NAFLD

Although the presence of NAFLD in obese individuals seems to increase the risk of APAP-induced acute liver injury (9, 42), these clinical studies did not address the mechanism whereby APAP could be more toxic in this metabolic situation. Nevertheless, increased hepatic CYP2E1 activity has frequently been reported in patients with NAFLD (22, 34, 81–83), as previously mentioned. However, it is still unclear whether CYP2E1 induction is specifically linked to this liver disease, or to other metabolic disturbances linked to obesity, such as insulin resistance and type 2 diabetes (22). In the experimental studies, fatty liver in obese rodents was in general associated with higher hepatotoxicity after APAP overdoses (Table 1)(57–59). In one of these studies, increased acute liver injury seemed to correlate with pre-existent hepatic CYP2E1 induction, but not with the degree of fat accretion (i.e. severity of fatty liver) (59). However, it is important to point out that greater hepatotoxicity in these different experimental studies was observed after the administration of APAP doses that were adapted to body weights. Hence, obese animals were treated with higher absolute doses of APAP. Contrasting with these studies, obesity was not associated with higher APAP hepatotoxicity in patients (for whom the presence of NAFLD was not reported, as previously pointed out) (43, 44). Experimentally, similar or lower APAP hepatotoxicity was observed in some rodent models of obesity (Table 1) (55, 56, 59). NAFLD was objectified only in some of these investigations, but for those where NAFLD was not determined it should be mentioned that previous studies demonstrated the presence of NAFLD in the same animal models (Table 1). Interestingly, obesity has been associated with higher APAP glucuronidation and clearance in different clinical and experimental investigations (35, 36, 59, 84– 86), as previously mentioned. In contrast, the influence of obesity and NAFLD on APAP sulphation is unclear since investigations in rodents and patients provided divergent results (59, 87–89). CYP3A activity is lower in Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Factors that could protect against liver injury Gastrointestinal absorption

Hepatic glucuronidation

Volume of distribution

Hepatic CYP3A4 activity

APAP plasma levels

Generation of NAPQI

Lower risk and severity of hepatic cytolysis

Factors that could favour liver injury Hepatic CYP2E1 activity Hepatic GSH stores

Generation of NAPQI NAPQI detoxication

Presence of NASH? Mito dysfunction?

Mito dysfunction Oxidative stress

Stronger risk and severity of hepatic cytolysis Fig. 1. Factors that could modulate the risk and severity of APAPinduced hepatotoxicity in obesity and NAFLD. (A) Factors that could protect against APAP-induced liver injury include reduced gastrointestinal absorption and hepatic CYP3A4 activity, as well as higher volume of distribution and hepatic glucuronidation. Overall, these factors can reduce plasma levels of APAP and the hepatic generation of NAPQI, a highly toxic APAP metabolite. (B) Factors that could favour APAP-induced liver injury include higher hepatic CYP2E1 activity, which leads to NAPQI overproduction and lower GSH stores in liver, which reduce its ability to detoxify NAPQI. Further investigations will be required to determine whether the presence of steatohepatitis (NASH) and/or the pre-existence of mitochondrial dysfunction could favour APAP-induced hepatotoxicity in obese individuals.

5

Michaut et al.

Acetaminophen hepatotoxicity in obesity

Remaining issues and future directions

Although there is some evidence that CYP2E1 induction could favour APAP-induced hepatotoxicity in the context of obesity and NAFLD, it is conceivable that other hepatic disturbances could play a role. For instance, it will be important to determine whether the severity of NAFLD could be a significant factor. Intuitively, one may expect that the presence of necroinflammation (i.e. NASH) could be associated with a higher risk, as compared to simple fatty liver. Likewise, the pre-existence of mitochondrial dysfunction linked to NASH (20, 94) could be of importance since this cellular event plays a key role in APAP-induced liver injury (23, 26, 40, 95). The relationship between NAFLD, reduced levels of hepatic GSH and APAP-induced liver injury will also require further investigations. Considering the low number of clinical studies published on APAP hepatotoxicity in obese patients, further investigations are clearly required to gain more information on this matter. More specifically, investigations will be needed to determine which obese individuals will be at risk for APAP-induced hepatotoxicity and those who will not. Indeed, at the current stage of knowledge, the health authorities cannot provide specific warnings for the whole obese population. Novel findings are not only needed on acute liver injury after APAP overdose but also regarding APAP hepatotoxicity occurring during chronic administration (7, 8). Thus, physicians could be encouraged to carry out a regular monitoring of liver function in obese patients treated with APAP, in particular in patients with pre-existing NAFLD. Large prospective studies will also be required to establish whether dysmetabolic disturbances other than NAFLD could favour APAP hepatotoxicity, such as insulin resistance and type 2 diabetes. Interestingly, diabetes has been associated with a higher risk of severe druginduced liver injury (96), although APAP cases were excluded in this study. It will also be important to determine the precise mechanism of CYP2E1 induction in obese patients with NAFLD. Previous clinical studies showed that hepatic CYP2E1 activity correlated with plasma ketone bodies and the degree of steatosis (34, 81, 82), suggesting that some lipid derivatives could be CYP2E1 inducers in humans. In keeping with this notion, current investigations in our laboratory indicate that CYP2E1 activity can significantly be induced in human hepatocytes by stearic acid, but not oleic acid. Moreover, our data show that stearate-induced CYP2E1 induction is reduced by insulin in a concentrationdependent manner. Interestingly, CYP2E1 activity was not correlated with its mRNA and protein expression suggesting post-translational regulation, as previously reported in other pathophysiological situations (97, 98). Thus, it appears that CYP2E1 activity in human liver could be controlled by some lipid derivatives and that lipid-induced CYP2E1 induction could be variable depending on insulin signalling. Identification of the

6

potential CYP2E1 endogenous inducers might help not only to decipher the pathophysiology of NAFLD (22) but also to understand why some obese individuals seem to present a higher risk of APAP-induced liver injury. It must finally be pointed out that besides APAP, other drugs such as halothane, methotrexate and some antiretroviral drugs are suspected to be more hepatotoxic in patients suffering from obesity and related metabolic disorders (99, 100). In contrast to APAP, virtually no experimental investigations have been performed with these drugs to understand why obesity can favour their hepatotoxicity. In future, it will be important to carry out experimental studies with the suspected drugs to determine the mechanism(s) of hepatotoxicity in the context of obesity and fatty liver, although one may expect that some mechanisms of hepatotoxicity could be common between certain drugs. In addition to in vitro studies using cellular models of nutritional fatty liver (101), in vivo investigations in obese rodents will be also required. Indeed, recent studies suggested that the effects of some xenobiotics on liver could be dependent on extra-hepatic tissues such as intestine and white adipose tissue (102–105). Hence, the mechanisms of drug-induced liver injury in obesity and NAFLD might be more complex than expected. Acknowledgements

Conflict of interest: The authors do not have any disclosures to report. References 1. Toussaint K, Yang XC, Zielinski MA, et al. What do we (not) know about how paracetamol (acetaminophen) works? J Clin Pharm Ther 2010; 35: 617–38. 2. Craig DG, Bates CM, Davidson JS, et al. Overdose pattern and outcome in paracetamol-induced acute severe hepatotoxicity. Br J Clin Pharmacol 2011; 71: 273–82. 3. Antoine DJ, Dear JW, Lewis PS, et al. Mechanistic biomarkers provide early and sensitive detection of acetaminophen-induced acute liver injury at first presentation to hospital. Hepatology 2013; 58: 777–87. 4. Ferner RE, Dear JW, Bateman DN. Management of paracetamol poisoning. BMJ 2011; 342: d2218. 5. Watkins PB, Kaplowitz N, Slattery JT, et al. Aminotransferase elevations in healthy adults receiving 4 grams of acetaminophen daily: a randomized controlled trial. JAMA 2006; 296: 87–93. 6. Winnike JH, Li Z, Wright FA, et al. Use of pharmacometabonomics for early prediction of acetaminopheninduced hepatotoxicity in humans. Clin Pharmacol Ther 2010; 88: 45–51. 7. Biour M, Ben Salem C, Chazouilleres O, et al. Druginduced liver injury; fourteenth updated edition of the bibliographic database of liver injuries and related drugs. Gastroenterol Clin Biol 2004; 28: 720–59. 8. Watelet J, Laurent V, Bressenot A, et al. Toxicity of chronic paracetamol ingestion. Aliment Pharmacol Ther 2007; 26: 1543–4.

Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Michaut et al.

9. 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: 1336–41. 10. Myers RP, Shaheen AA, Li B, et al. Impact of liver disease, alcohol abuse, and unintentional ingestions on the outcomes of acetaminophen overdose. Clin Gastroenterol Hepatol 2008; 6: 918–25. 11. Claridge LC, Eksteen B, Smith A, et al. Acute liver failure after administration of paracetamol at the maximum recommended daily dose in adults. BMJ 2010; 341: c6764. 12. Nolan CM, Sandblom RE, Thummel KE, et al. Hepatotoxicity associated with acetaminophen usage in patients receiving multiple drug therapy for tuberculosis. Chest 1994; 105: 408–11. 13. Lane JE, Belson MG, Brown DK, et al. Chronic acetaminophen toxicity: a case report and review of the literature. J Emerg Med 2002; 23: 253–6. 14. Suzuki A, Yuen N, Walsh J, et al. Co-medications that modulate liver injury and repair influence clinical outcome of acetaminophen-associated liver injury. Clin Gastroenterol Hepatol 2009; 7: 882–8. 15. Harrill AH, Watkins PB, Su S, et al. Mouse populationguided resequencing reveals that variants in CD44 contribute to acetaminophen-induced liver injury in humans. Genome Res 2009; 19: 1507–15. 16. Court MH, Peter I, Hazarika S, et al. Candidate gene polymorphisms in patients with acetaminophen-induced acute liver failure. Drug Metab Dispos 2014; 42: 28–32. 17. Lazo M, Hernaez R, Eberhardt MS, et al. Prevalence of nonalcoholic fatty liver disease in the United States: the Third National Health and Nutrition Examination Survey, 1988-1994. Am J Epidemiol 2013; 178: 38–45. 18. Berardis S, Sokal E. Pediatric non-alcoholic fatty liver disease: an increasing public health issue. Eur J Pediatr 2014; 173: 131–9. 19. Tamura S, Shimomura I. Contribution of adipose tissue and de novo lipogenesis to nonalcoholic fatty liver disease. J Clin Invest 2005; 115: 1139–42. 20. Begriche K, Massart J, Robin MA, et al. Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology 2013; 58: 1497–507. 21. Fabbrini E, Sullivan S, Klein S. Obesity and nonalcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology 2010; 51: 679–89. 22. Aubert J, Begriche K, Knockaert L, et al. Increased expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: mechanisms and pathophysiological role. Clin Res Hepatol Gastroenterol 2011; 35: 630–7. 23. Hinson JA, Roberts DW, James LP. Mechanisms of acetaminophen-induced liver necrosis. Handb Exp Pharmacol 2010; 196: 369–405. 24. Zhao L, Pickering G. Paracetamol metabolism and related genetic differences. Drug Metab Rev 2011; 43: 41–52. 25. Jaeschke H, Williams CD, Ramachandran A, et al. Acetaminophen hepatotoxicity and repair: the role of sterile inflammation and innate immunity. Liver Int 2012; 32: 8–20. 26. Jaeschke H, McGill MR, Ramachandran A. Oxidant stress, mitochondria, and cell death mechanisms in druginduced liver injury: lessons learned from acetaminophen hepatotoxicity. Drug Metab Rev 2012; 44: 88–106.

Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Acetaminophen hepatotoxicity in obesity

27. Prescott LF. Kinetics and metabolism of paracetamol and phenacetin. Br J Clin Pharmacol 1980; 10(Suppl 2): 291S– 8S. 28. Dahlin DC, Miwa GT, Lu AY, et al. N-acetyl-p-benzoquinone imine: a cytochrome P450-mediated oxidation product of acetaminophen. Proc Natl Acad Sci USA 1984; 81: 1327–31. 29. Gonzalez FJ. The 2006 Bernard B. Brodie Award Lecture. Cyp2E1. Drug Metab Dispos 2007; 35: 1–8. 30. Lee SS, Buters JT, Pineau T, et al. Role of CYP2E1 in the hepatotoxicity of acetaminophen. J Biol Chem 1996; 271: 12063–7. 31. Thummel KE, Lee CA, Kunze KL, et al. Oxidation of acetaminophen to N-acetyl-p-aminobenzoquinone imine by human CYP3A4. Biochem Pharmacol 1993; 45: 1563– 9. 32. Yoshitomi S, Ikemoto K, Takahashi J, et al. Establishment of the transformants expressing human cytochrome P450 subtypes in HepG2, and their applications on drug metabolism and toxicology. Toxicol In Vitro 2001; 15: 245–56. 33. Laine JE, Auriola S, Pasanen M, et al. Acetaminophen bioactivation by human cytochrome P450 enzymes and animal microsomes. Xenobiotica 2009; 39: 11–21. 34. Chalasani N, Gorski JC, Asghar MS, et al. Hepatic cytochrome P450 2E1 activity in nondiabetic patients with nonalcoholic steatohepatitis. Hepatology 2003; 37: 544– 50. 35. Abernethy DR, Greenblatt DJ, Divoll M, et al. Enhanced glucuronide conjugation of drugs in obesity: studies of lorazepam, oxazepam, and acetaminophen. J Lab Clin Med 1983; 101: 873–80. 36. Brill MJ, Diepstraten J, van Rongen A, et al. Impact of obesity on drug metabolism and elimination in adults and children. Clin Pharmacokinet 2012; 51: 277–304. 37. Patoine D, Levac X, Pilote S, et al. Decreased CYP3A expression and activity in guinea pig models of dietinduced metabolic syndrome: is fatty liver infiltration involved? Drug Metab Dispos 2013; 41: 952–7. 38. Qiu Y, Benet LZ, Burlingame AL. Identification of the hepatic protein targets of reactive metabolites of acetaminophen in vivo in mice using two-dimensional gel electrophoresis and mass spectrometry. J Biol Chem 1998; 273: 17940–53. 39. Muldrew KL, James LP, Coop L, et al. Determination of acetaminophen-protein adducts in mouse liver and serum and human serum after hepatotoxic doses of acetaminophen using high-performance liquid chromatography with electrochemical detection. Drug Metab Dispos 2002; 30: 446–51. 40. Han D, Shinohara M, Ybanez MD, et al. Signal transduction pathways involved in drug-induced liver injury. Handb Exp Pharmacol 2010; 196: 267–310. 41. Uzi D, Barda L, Scaiewicz V, et al. CHOP is a critical regulator of acetaminophen-induced hepatotoxicity. J Hepatol 2013; 59: 495–503. 42. Myers RP, Shaheen AA. Hepatitis C alcohol abuse, and unintentional overdoses are risk factors for acetaminophen-related hepatotoxicity. Hepatology 2009; 49: 1399– 400. 43. Rutherford A, Davern T, Hay JE, et al. Influence of high body mass index on outcome in acute liver failure. Clin Gastroenterol Hepatol 2006; 4: 1544–9.

7

Michaut et al.

Acetaminophen hepatotoxicity in obesity

44. 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 2013; doi: 10.1097/01.mjt.0000434043. 62372.00 [Epub ahead of print]. 45. 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. 46. Koteish A, Diehl AM. Animal models of steatosis. Semin Liver Dis 2001; 21: 89–104. 47. Begriche K, Letteron P, Abbey-Toby A, et al. Partial leptin deficiency favors diet-induced obesity and related metabolic disorders in mice. Am J Physiol Endocrinol Metab 2008; 294: E939–51. 48. Begriche K, Massart J, Abbey-Toby A, et al. b-Aminoisobutyric acid prevents diet-induced obesity in mice with partial leptin deficiency. Obesity 2008; 16: 2053–67. 49. Hebbard L, George J. Animal models of nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2011; 8: 35–44. 50. Trak-Smayra V, Paradis V, Massart J, et al. Pathology of the liver in obese and diabetic ob/ob and db/db mice fed a standard or high-calorie diet. Int J Exp Pathol 2011; 92: 413–21. 51. Lindstr€ om P. The physiology of obese-hyperglycemic mice [ob/ob mice]. ScientificWorldJournal 2007; 7: 666–85. 52. Fellmann L, Nascimento AR, Tibiricßa E, et al. Murine models for pharmacological studies of the metabolic syndrome. Pharmacol Ther 2013; 137: 331–40. 53. Okazaki M, Saito Y, Udaka Y, et al. Diabetic nephropathy in KK and KK-Ay mice. Exp Anim 2002; 51: 191–6. 54. Corcoran GB, Wong BK. Obesity as a risk factor in druginduced organ injury: increased liver and kidney damage by acetaminophen in the obese overfed rat. J Pharmacol Exp Ther 1987; 241: 921–7. 55. Blouin RA, Dickson P, McNamara PJ, et al. Phenobarbital induction and acetaminophen hepatotoxicity: resistance in the obese Zucker rodent. J Pharmacol Exp Ther 1987; 243: 565–70. 56. Ito Y, Abril ER, Bethea NW, et al. Dietary steatotic liver attenuates acetaminophen hepatotoxicity in mice. Microcirculation 2006; 13: 19–27. 57. Kon K, Ikejima K, Okumura K, et al. Diabetic KK-A(y) mice are highly susceptible to oxidative hepatocellular damage induced by acetaminophen. Am J Physiol Gastrointest Liver Physiol 2010; 299: G329–37. 58. Kucera O, Rousar T, Stankova P, et al. Susceptibility of rat non-alcoholic fatty liver to the acute toxic effect of acetaminophen. J Gastroenterol Hepatol 2012; 27: 323–30. 59. Aubert J, Begriche K, Delannoy M, et al. Differences in early acetaminophen hepatotoxicity between obese ob/ob and db/db mice. J Pharmacol Exp Ther 2012; 342: 676–87. 60. Donthamsetty S, Bhave VS, Mitra MS, et al. Nonalcoholic steatohepatitic (NASH) mice are protected from higher hepatotoxicity of acetaminophen upon induction of PPARalpha with clofibrate. Toxicol Appl Pharmacol 2008; 230: 327–37. 61. Leclercq IA, Farrell GC, Field J, et al. CYP2E1 and CYP4A as microsomal catalysts of lipid peroxides in murine nonalcoholic steatohepatitis. J Clin Invest 2000; 105: 1067–75. 62. Rizki G, Arnaboldi L, Gabrielli B, et al. Mice fed a lipogenic methionine-choline-deficient diet develop

8

63.

64. 65.

66.

67.

68. 69. 70. 71. 72.

73. 74. 75. 76. 77.

78.

79.

80.

hypermetabolism coincident with hepatic suppression of SCD-1. J Lipid Res 2006; 47: 2280–90. Leclercq IA, Lebrun VA, St€arkel P, et al. Intrahepatic insulin resistance in a murine model of steatohepatitis: effect of PPARgamma agonist pioglitazone. Lab Invest 2007; 87: 56–65. Rinella ME, Green RM. The methionine-choline deficient dietary model of steatohepatitis does not exhibit insulin resistance. J Hepatol 2004; 40: 47–51. Tee LB, Davies DS, Seddon CE, et al. Species differences in the hepatotoxicity of paracetamol are due to differences in the rate of conversion to its cytotoxic metabolite. Biochem Pharmacol 1987; 36: 1041–52. Jemnitz K, Veres Z, Monostory K, et al. Interspecies differences in acetaminophen sensitivity of human, rat, and mouse primary hepatocytes. Toxicol In Vitro 2008; 22: 961–7. Enriquez A, Leclercq I, Farrell GC, et al. Altered expression of hepatic CYP2E1 and CYP4A in obese, diabetic ob/ ob mice, and fa/fa Zucker rats. Biochem Biophys Res Commun 1999; 255: 300–6. Bellward GD, Chang T, Rodrigues B, et al. Hepatic cytochrome P-450j induction in the spontaneously diabetic BB rat. Mol Pharmacol 1988; 33: 140–3. Guo Y, Walther TC, Rao M, et al. Functional genomic screen reveals genes involved in lipid-droplet formation and utilization. Nature 2008; 453: 657–61. Shi X, Li J, Zou X, et al. Regulation of lipid droplet size and phospholipid composition by stearoyl-CoA desaturase. J Lipid Res 2013; 54: 2504–14. Seki S, Kitada T, Sakaguchi H. Clinicopathological significance of oxidative cellular damage in non-alcoholic fatty liver diseases. Hepatol Res 2005; 33: 132–4. Bell LN, Temm CJ, Saxena R, et al. Bariatric surgeryinduced weight loss reduces hepatic lipid peroxidation levels and affects hepatic cytochrome P450 protein content. Ann Surg 2010; 251: 1041–8. Videla LA, Rodrigo R, Orellana M, et al. Oxidative stressrelated parameters in the liver of non-alcoholic fatty liver disease patients. Clin Sci 2004; 106: 261–8. Zhao P, Kalhorn TF, Slattery JT. Selective mitochondrial glutathione depletion by ethanol enhances acetaminophen toxicity in rat liver. Hepatology 2002; 36: 326–35. Fernandez-Checa JC, Kaplowitz N. Hepatic mitochondrial glutathione: transport and role in disease and toxicity. Toxicol Appl Pharmacol 2005; 204: 263–73. Singh R, Wang Y, Xiang Y, et al. Differential effects of JNK1 and JNK2 inhibition on murine steatohepatitis and insulin resistance. Hepatology 2009; 49: 87–96. Abdelmegeed MA, Banerjee A, Yoo SH, et al. Critical role of cytochrome P450 2E1 (CYP2E1) in the development of high fat-induced non-alcoholic steatohepatitis. J Hepatol 2012; 57: 860–6. Hanawa N, Shinohara M, Saberi B, et al. Role of JNK translocation to mitochondria leading to inhibition of mitochondria bioenergetics in acetaminophen-induced liver injury. J Biol Chem 2008; 283: 13565–77. Serviddio G, Bellanti F, Tamborra R, et al. Alterations of hepatic ATP homeostasis and respiratory chain during development of non-alcoholic steatohepatitis in a rodent model. Eur J Clin Invest 2008; 38: 245–52. Serviddio G, Bellanti F, Giudetti AM, et al. A silybinphospholipid complex prevents mitochondrial dysfunc-

Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Michaut et al.

81.

82.

83.

84.

85.

86.

87. 88.

89.

90. 91.

92. 93. 94. 95.

tion in a rodent model of nonalcoholic steatohepatitis. J Pharmacol Exp Ther 2010; 332: 922–32. Emery MG, Fisher JM, Chien JY, et al. CYP2E1 activity before and after weight loss in morbidly obese subjects with nonalcoholic fatty liver disease. Hepatology 2003; 38: 428–35. Chtioui H, Semela D, Ledermann M, et al. Expression and activity of the cytochrome P450 2E1 in patients with nonalcoholic steatosis and steatohepatitis. Liver Int 2007; 27: 764–71. Varela NM, Qui~ nones LA, Orellana M, et al. Study of cytochrome P450 2E1 and its allele variants in liver injury of nondiabetic, nonalcoholic steatohepatitis obese women. Biol Res 2008; 41: 81–92. Barshop NJ, Capparelli EV, Sirlin CB, et al. Acetaminophen pharmacokinetics in children with nonalcoholic fatty liver disease. J Pediatr Gastroenterol Nutr 2011; 52: 198–202. Xu J, Kulkarni SR, Li L, et al. UDP-glucuronosyltransferase expression in mouse liver is increased in obesity and fasting-induced steatosis. Drug Metab Dispos 2012; 40: 259–66. Gicquel T, Aubert J, Lepage S, et al. Quantitative analysis of acetaminophen and its primary metabolites in small plasma volumes by liquid chromatography-tandem mass spectrometry. J Anal Toxicol 2013; 37: 110–6. Corcoran GB, Wong BK, Shum L, et al. Acetaminophen sulfation deficit in obese rats overfed an energy-dense cafeteria diet. Endocr Res 1987; 13: 101–21. Chaudhary IP, Tuntaterdtum S, McNamara PJ, et al. Effect of genetic obesity and phenobarbital treatment on the hepatic conjugation pathways. J Pharmacol Exp Ther 1993; 265: 1333–8. Hardwick RN, Ferreira DW, More VR, et al. Altered UDP-glucuronosyltransferase and sulfotransferase expression and function during progressive stages of human nonalcoholic fatty liver disease. Drug Metab Dispos 2013; 41: 554–61. Yoshinari K, Takagi S, Yoshimasa T, et al. Hepatic CYP3A expression is attenuated in obese mice fed a highfat diet. Pharm Res 2006; 23: 1188–200. Kolwankar D, Vuppalanchi R, Ethell B, et al. Association between nonalcoholic hepatic steatosis and hepatic cytochrome P-450 3A activity. Clin Gastroenterol Hepatol 2007; 5: 388–93. Lee WH, Kramer WG, Granville GE. The effect of obesity on acetaminophen pharmacokinetics in man. J Clin Pharmacol 1981; 21: 284–7. Abernethy DR, Divoll M, Greenblatt DJ, et al. Obesity, sex, and acetaminophen disposition. Clin Pharmacol Ther 1982; 31: 783–90. Grattagliano I, de Bari O, Bernardo TC, et al. Role of mitochondria in nonalcoholic fatty liver disease: from origin to propagation. Clin Biochem 2012; 45: 610–8. Knockaert L, Descatoire V, Vadrot N, et al. Mitochondrial CYP2E1 is sufficient to mediate oxidative stress and

Liver International (2014) © 2014 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Acetaminophen hepatotoxicity in obesity

96.

97.

98.

99. 100. 101.

102.

103.

104.

105. 106. 107.

108.

109.

110.

cytotoxicity induced by ethanol and acetaminophen. Toxicol In Vitro 2011; 25: 475–84. Chalasani N, Fontana RJ, Bonkovsky HL, et al. Causes, clinical features, and outcomes from a prospective study of drug-induced liver injury in the United States. Gastroenterology 2008; 135: 1924–34. Oesch-Bartlomowicz B, Padma PR, Becker R, et al. Differential modulation of CYP2E1 activity by cAMP-dependent protein kinase upon Ser129 replacement. Exp Cell Res 1998; 242: 294–302. Wauthier V, Schenten V, Verbeeck RK, et al. Ageing is associated with increased expression but decreased activity of CYP2E1 in male Wistar rats. Life Sci 2006; 79: 1913–20. Fromenty B. Drug-induced liver injury in obesity. J Hepatol 2013; 58: 824–6. Pais R, Rusu E, Ratziu V. The impact of obesity and metabolic syndrome on chronic hepatitis B and druginduced liver disease. Clin Liver Dis 2014; 18: 165–78. Antherieu S, Rogue A, Fromenty B, et al. Induction of vesicular steatosis by amiodarone and tetracycline is associated with up-regulation of lipogenic genes in HepaRG cells. Hepatology 2011; 53: 1895–905. Song Z, Zhou Z, Deaciuc I, et al. Inhibition of adiponectin production by homocysteine: a potential mechanism for alcoholic liver disease. Hepatology 2008; 47: 867–79. Fromenty B, Vadrot N, Massart J, et al. Chronic ethanol consumption lessens the gain of body weight, liver triglycerides, and diabetes in obese ob/ob mice. J Pharmacol Exp Ther 2009; 331: 23–34. Massart J, Robin MA, Noury F, et al. 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: 1361–74. Tan X, Sun X, Li Q, et al. Leptin deficiency contributes to the pathogenesis of alcoholic fatty liver disease in mice. Am J Pathol 2012; 181: 1279–86. Carmiel-Haggai M, Cederbaum AI, Nieto N. Binge ethanol exposure increases liver injury in obese rats. Gastroenterology 2003; 125: 1818–33. Cipriani S, Mencarelli A, Palladino G, et al. FXR activation reverses insulin resistance and lipid abnormalities and protects against liver steatosis in Zucker (fa/fa) obese rats. J Lipid Res 2010; 51: 771–84. Zaluzny L, Farrell GC, Murray M. Effect of genetic obesity and experimental diabetes on hepatic microsomal mixed function oxidase activities. J Gastroenterol Hepatol 1990; 5: 256–63. Robin MA, Demeilliers C, Sutton A, et al. Alcohol increases tumor necrosis factor-a and decreases nuclear factor-B to activate hepatic apoptosis in genetically obese mice. Hepatology 2005; 42: 1280–90. Okumura K, Ikejima K, Kon K, et al. Exacerbation of dietary steatohepatitis and fibrosis in obese, diabetic KKA(y) mice. Hepatol Res 2006; 36: 217–28.

9

Acetaminophen-induced liver injury in obesity and nonalcoholic fatty liver disease.

Although acetaminophen (APAP) is usually considered as a safe drug, this painkiller can lead to acute liver failure after overdoses. Moreover, there i...
221KB Sizes 0 Downloads 3 Views