Hepatotoxicity Mechanisms of Isoniazid: A Mini-Review

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Review Article

Received: 15 April 2015, Accepted: 17 April 2015 Published online in Wiley Online Library

(wileyonlinelibrary.com) DOI 10.1002/jat.3175

Hepatotoxicity mechanisms of isoniazid:


A mini-review
Hozeifa M. Hassana,f, Hong-li Guoa, Bashir A. Yousefa,g, Zhang Luyonga,b,d*
and Jiang Zhenzhoua,c,e*

ABSTRACT: Isoniazid (INH) is an antituberculosis drug associated with idiosyncratic liver injury in susceptible patients.
INH-induced hepatotoxicity remains a significant clinical problem, but the underlying mechanisms are still unclear, despite
the growing evidence that INH and/or its major metabolite, hydrazine, play an important role in hepatotoxicity. Copyright ©
2015 John Wiley & Sons, Ltd.

Keywords: isoniazid; hepatotoxicity mechanisms; CYP2E1

Introduction the purpose of this review is to understand the possible mecha-


nisms by which INH causes hepatotoxicity.
Tuberculosis (TB) remains a major global health problem, whose
effects have major impact in developing countries, despite the
availability of highly efficacious treatment for decades (Lyoumi Hepatotoxicity of Isoniazid
et al., 2013; Ramappa and Aithal, 2013; Singh et al., 2014). World
INH is a widely used first-line anti-TB drug that has been associated
Health Organization (WHO) declared TB a global public health
with idiosyncratic (host-dependent) drug-induced liver injury in
emergency in 1993 (Ramappa and Aithal, 2013). Recommended
susceptible patients. The incidence of INH-related hepatic adverse
standard treatment for adult respiratory TB is a regimen of isonia-
reactions is relatively high, as compared to that induced by other
zid (INH), rifampicin and pyrazinamide for 2 months, followed by 4
drugs (Lee et al., 2013). The use of INH is associated with elevated
months of INH and rifampicin. Ethambutol is usually added to this
serum levels of alanine aminotransferase in up to 20% of patients,
regimen and streptomycin is recommended by the WHO in
whereas overt hepatotoxicity can occur in up to 2% of patients
retreatment cases in most developing countries (Sharifzadeh
(Boelsterli and Lee, 2014; CDC, 2010; Metushi et al., 2011; Walubo
et al., 2005; Tostmann et al., 2008a). The most frequent adverse
et al., 1998). Although the pathology can vary, in most cases liver
effect of anti-TB treatment is hepatotoxicity (Obogwu et al., 2014;
biopsies from patients with severe hepatotoxicity are indistin-
Park et al., 2010; Sharifzadeh et al., 2005; Sharma, 2004; Shih
guishable from those from patients with viral hepatitis; the find-
et al., 2013; Tostmann et al., 2008a).
ings include necrosis, inflammation and, in some instances, an
Anti-TB drug-induced hepatotoxicity is a serious problem and
main cause of treatment interruption and change in treatment
regimen during the TB treatment course (Khalili et al., 2009; *Correspondence to: Zhang Luyong or Jiang Zhenzhou, Jiangsu Key Laboratory of
Tostmann et al., 2008b). Anti-TB drug-induced hepatotoxicity Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
causes substantial morbidity and mortality. Asymptomatic trans- E-mail: lyzhang@cpu.edu.cn; beaglejiang@cpu.edu.cn
aminase elevations are common during anti-TB treatment, but a
Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University,
hepatotoxicity can be fatal when not recognized early and when Nanjing, China
therapy is not interrupted in time. Anti-TB drugs such as INH, rifam-
b
picin and pyrazinamide have been found to be potentially hepato- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharma-
toxic (Park et al., 2010; Sharifzadeh et al., 2005). There has been a ceutical University, Nanjing, China
report of ethambutol-induced liver cholestatic jaundice, with un- c
Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharma-
clear circumstances. The risk of anti-TB drug-induced hepatotoxi- ceutical University), Ministry of Education, Nanjing, China
city has been found to increase by various factors such as high
d
alcohol intake, older age, pre-existing chronic liver disease, chronic Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China
Pharmaceutical University, Nanjing, China
viral infection, advanced TB, female sex, concomitant administra-
tion of hepatotoxic drugs, inappropriate use of drugs and e
State Key Laboratory of Natural Medicines, China Pharmaceutical University,
nutritional status (Hosford et al., 2015; Khalili et al., 2009; Sharma, Nanjing, China
2004; Uetrecht and Naisbitt, 2013).
f
INH is a first-line medication used in the treatment of TB. How- Department of Pharmacology, Faculty of Pharmacy, University of Gezira,
Wad-Medani, Sudan
ever, INH therapy is associated with serious hepatotoxicity and
potentially fatal liver injury (Georgieva et al., 2005; Metushi and g
Department of Pharmacology, Faculty of Pharmacy, University of Khartoum,
Uetrecht, 2014b; Sarich et al., 1995 Tafazoli et al., 2008). Therefore, Khartoum, Sudan

J. Appl. Toxicol. 2015 Copyright © 2015 John Wiley & Sons, Ltd.
H. M. Hassan et al.

infiltration of eosinophils (Bjornsson et al., 2007; Black et al., 1975; HepG2 cell line, could be mediated by the intrinsic pathway of
Maddrey and Boitnott, 1973). Histopathological changes in apoptosis.
INH-induced hepatotoxicity in humans range in severity from Mitochondrial dysfunction can encompass several changes such
focal and diffuse necrosis to multi-lobular, bridging and massive as decreased adenosine triphosphate production, mitochondrial
necrosis (Sarich et al., 1999). INH hepatotoxicity is slightly more reactive oxygen species (ROS) production and/or dissipation of
common in patients who are “slow acetylators” and have high the mitochondrial membrane potential. Either an exogenously im-
CYP2E1 levels, but this increased risk is much too small to be posed mitochondrial disease or a polymorphism could alter mito-
responsible for the idiosyncratic nature of INH hepatotoxicity chondrial function and render cells sensitive to a drug, resulting in
(Su et al., 2014). idiosyncratic toxicity (Waring and Anderson, 2005). Troglitazone
Overall, there are certain features common to INH-induced (Shishido et al., 2003), tolcapone (Haasio et al., 2002), diclofenac
hepatotoxicity, such as a delayed onset; however, there is also (Petrescu and Tarba, 1997), valproic acid (Keller et al., 1992) and
significant variability between patients. Specifically, in most INH (Schwab and Tuschl, 2003) are some of the drugs that cause
patients the injury resolves despite continued treatment, whereas mitochondrial dysfunction in hepatocytes in vitro.
in some patients it progresses even after the drug is stopped; in
most patients, re-challenge does not lead to immediate recurrence
Role of Oxidative Stress
of injury, but in some it does (Metushi et al., 2011).
One reason for the difficulty in identifying susceptibility factors Oxidative stress was proposed as one mechanism responsible for
in patients is the lack of a clear understanding of the molecular hepatic injury induced by INH (Rao et al., 2012; Shen et al., 2007).
mechanisms underlying INH-induced liver injury (Lee et al., 2013). The imbalance between the generation and the removal of free
Moreover, mechanistic studies are difficult to perform because it radicals after chronic INH treatment leads to pathological conse-
is important to obtain samples before the injury is severe so that quences, although multiple mechanisms are involved in INH
the events leading up to the injury can be studied, but it is impos- hepatotoxicity (Yue et al., 2009a). Meanwhile, clinical data showed
sible to predict which patient will develop serious toxicity. In addi- that an increased level of glutathione and a decreased level of
tion, it has not been possible to reproduce liver injury in animals malondialdehyde were measured in plasma of patient treated
with the same characteristics as the idiosyncratic injury that occurs with INH, suggesting that INH could result in oxidative stress
in patients (Metushi and Uetrecht, 2014a). (Chen et al., 2011). The phenomenon was glutathione-dependent
and antioxidants might have a protective role, which suggest the
important role of oxidative stress in INH hepatotoxicity (Enriquez-
Hepatotoxicity Mechanisms of Isoniazid Cortina et al., 2013). The observed hepatoprotective effect of
Although INH, among all drugs causing drug-induced liver injury, N-acetylcysteine (a sulfhydryl-containing compound that can re-
has a relatively high incidence of hepatocellular injury in suscepti- duce oxidized glutathione into reduced glutathione) in rats treated
ble patients, conventional models involving normal hepatocytes or with INH and rifampicin further supports this involvement. In
healthy laboratory animals have proven unsuccessful in reprodu- contrast, induced glutathione depletion does not affect in vitro
cing the clinical picture of liver injury, even when high doses of INH-induced toxicity, suggests that glutathione is not directly
INH were used. Unfortunately there are currently no animal models involved in INH-induced toxicity (Tostmann et al., 2008a).
available that recapitulate the clinical picture of INH hepatotoxicity CYP2E1 might be a central pathway in oxidative stress, produc-
that could elucidate the underlying mechanisms in detail, or it has tion of ROS and hepatotoxic injury, particularly in the presence of
not been possible to reproduce liver injury that has the same CYP2E1 inducers. Inductions of CYP2E1 in mice (Chowdhury
characteristics as the idiosyncratic injury that occurs in patients et al., 2006) and in rats (Zhai et al., 2008) with increased CYP2E1
(Lee et al., 2013; Metushi and Uetrecht, 2014b; Sarich et al., 1995). activity were found in animals dosed with INH.
Despite all these difficulties in clarifying the whole picture and INH is an effective inhibitor of antioxidant response elements
explaining the exact mechanisms of INH hepatotoxicity, many in- (ARE) activity. Even at concentrations far less than causes tangible
vestigations were conducted and aimed at identifying as well as cytotoxicity, INH readily suppresses the expression of many
exploring the potential mechanisms participating in INH-induced ARE-dependent antioxidant and phase II detoxification genes,
hepatotoxicity, which may allow clinicians to develop strategies including Nqo1, Ho1 and Gclc, under both basal and oxidative
to reduce the occurrence of hepatotoxicity and its adverse out- stressed conditions in 3T3-L1 cells and human hepatocyte HepG2
come. Those potential explanations were listed below. cells. These findings suggest that the toxic effects of INH, including
hepatotoxicity may be due partly to oxidative stress resulting
from inhibition of ARE-dependent antioxidant gene expression
Apoptosis and Mitochondrial Dysfunction in Isoniazid
(Chen et al., 2013).
Hepatotoxicity
It has been suggested that most drug-induced hepatotoxicity is
Lipid Peroxidation as a Potential Isoniazid Hepatotoxic
caused by mitochondrial damage (Boelsterli and Lee, 2014;
Mechanism
Pessayre et al., 2010). In vitro studies of a variety of animal cell lines
demonstrated that, INH toxicity results from the induction of Peroxidation of endogenous lipids has been shown to be a major
apoptosis with associated disruption of mitochondrial membrane factor in the cytotoxic action of INH (Mahmoud et al., 2014; Pal
potential and DNA strand breaks (Anon, 2008; Fatima et al., 2013; et al., 2008; Richards et al., 2004; Saad et al., 2010). INH-mediated
Lee and Boelsterli, 2014; Perwitasari et al., 2014; Shaw et al., 2010; oxidative damage is generally attributed to the formation of the
Singh et al., 2011). Recent reports suggest that INH induces oxida- highly ROS, which act as stimulators of lipid peroxidation and
tive stress, mitochondria dysfunction and apoptosis in HepG2 cells source for destruction and damage to the cell membrane. INH
(Bhadauria et al., 2007, 2010; Schwab and Tuschl, 2003). This gives and its metabolites act as stimulators of lipid peroxidation
an indication that INH-induced apoptosis, particularly in the resulting in cell death and hepatic necrosis (Caro and Cederbaum,

wileyonlinelibrary.com/journal/jat Copyright © 2015 John Wiley & Sons, Ltd. J. Appl. Toxicol. 2015
Hepatotoxicity mechanisms of isoniazid: A mini-review

2004; Yew and Leung, 2006; Yue and Peng, 2009). Peroxidation of and reactive nitrogen species in the rat hepatocytes after being me-
accumulated lipids leads to formation of toxic reactive aldehyde tabolized by enzymes, including CYP2E1 (Nanashima et al., 2012).
byproducts and downstream effects, such as impaired membrane INH is metabolized and cleared predominantly in the liver. The key
integrity, mitochondrial and sarcoplasmic reticulum dysfunction enzymes in the metabolic pathway, N-acetyltransferase 2 (NAT2)
and altered calcium homeostasis (Adhvaryu et al., 2007). and CYP2E1 determine the risk of hepatotoxicity (Ingawale et al.,
Moreover, INH through suppressing ARE activity, inhibits 2014; Li et al., 2013; Ohno et al., 2000; Saukkonen et al., 2006).
adipogenesis by interfering with the expression of C/EBPβ and As illustrated in Fig. 1, NAT2 is responsible for metabolism of INH
C/EBPδ during the early stage of adipogenesis, suggesting that to acetyl-INH, which in turn is hydrolyzed to acetyl hydrazine. The
INH treatment may impair the development and function of latter could be oxidized by CYP2E1 to form N-hydroxy acetyl
adipose tissues (Chen et al., 2013). hydrazine, which further dehydrates to yield acetyl diazine. Acetyl
Furthermore, alterations of various cellular defense mechanisms diazine may itself be the toxic metabolite or may break down to
consisting of enzymatic and non-enzymatic components have reactive acetyl onium ion, acetyl radical and ketene, which could
been reported to be involved in INH-induced hepatotoxicity bind covalently with hepatic macromolecules resulting in liver
(Saad et al., 2010). injury. The enzyme NAT2 is also responsible for further acetylation
of acetyl hydrazine to non-toxic diacetyl hydrazine (Ramappa and
Aithal, 2013; Singla et al., 2014).
Role of Isoniazid Metabolites in Hepatotoxicity
During the metabolism of INH, hydrazine was produced directly
INH-induced hepatotoxicity is considered idiosyncratic, i.e. reactive (from INH) or indirectly (from acetyl hydrazine). Previous evidence
toxic metabolites (hydrazine, mono acetyl hydrazine) rather than strongly suggest that hydrazine plays a crucial role in INH-induced
the parent drug are responsible for hepatotoxicity (Fatima et al., liver damage in rats (Issabeagloo et al., 2013; Mujahid et al., 2013;
2013; Sharma, 2011; Tafazoli et al., 2008). Hepatotoxicity of INH is Zhang et al., 2012). Hydrazine is implicated in inducing steatosis
considered due to metabolism of the nitrogen-containing group by altering the hepatic gene expression profile favoring produc-
in its chemical structure resulting in reactive metabolites that lead tion and intracellular transport of hepatic lipid over the removal
to hepatitis (Cheng et al., 2013). of fatty acid metabolites (Adhvaryu et al., 2007).
From an etiological perspective, the accumulation of these toxic There was a significant correlation between plasma hydrazine
metabolites, hydrazine and acetyl hydrazine, in hepatocytes contri- levels and INH hepatotoxicity; in contrast, plasma acetyl hydrazine
butes to anti-TB drug-induced hepatotoxicity. In addition, these or INH concentration did not correlate with INH-induced hepatic
hepatotoxic metabolites induced the excessive production of ROS necrosis or fatty changes. Elevation of hepatic CYP2E1 plays an

Figure 1. Pathways involved in the metabolism of isoniazid.

J. Appl. Toxicol. 2015 Copyright © 2015 John Wiley & Sons, Ltd. wileyonlinelibrary.com/journal/jat
H. M. Hassan et al.

essential role in the development of INH hepatotoxicity through CYP2E1 catalyzes the biotransformation of numerous drugs, in-
the generation of free radicals from hydrazine ( Jaswal et al., cluding INH. Importantly, CYP2E1-mediated biotransformation of
2013; Sarich et al., 1996; Yue et al., 2004, 2009b). INH can generate toxic reactive metabolites, thus playing a major
role in drug metabolism and the pathophysiology of drug-induced
liver injury (Aubert et al., 2011; Santos et al., 2013; Shayakhmetova
New Theories for Isoniazid Hepatotoxicity et al., 2015). Human genetic researches have revealed that
Beside the above-mentioned potential hypothesis regarding the cytochrome CYP2E1 is involved in INH-induced hepatotoxicity
explanation of INH hepatotoxic mechanisms, new theories were (Humayun et al., 2014).
discussed; among those theories was the interaction on the Although CYP2E1 was reported not be involved in INH-induced
immune system. hepatotoxicity; however, an INH metabolite derived from CYP2E1
Although it was previously believed that INH-induced liver injury appears to play a role in INH-induced hepatotoxicity through
was not immune mediated, there are now several lines of evidence enhancement of bile acid accumulation and mitochondria
that INH-induced liver injury is mediated by the adaptive immune β-oxidation. In that study, histological analysis revealed that INH
system ( James and Roberts, 2014; Metushi et al., 2012, 2014a; administration had no clear effect on normal liver histology in
Metushi and Uetrecht, 2014b). INH can bind to macrophages CYP2E1-null mice (have an absence of CYP2E1 protein expression),
in vitro, which suggests that the parent drug can form covalent while a slight cholestasis was observed in wild-type (WT) mice,
adducts and initiate an immune response by binding to an alde- although without significant difference of the histological score.
hyde group on macrophages and stimulating the production of INH significantly increased the serum cholesterol, serum triglyce-
interleukin-6 (Li and Uetrecht, 2009; Metushi et al., 2014a). It is pos- rides and hepatic bile acids in WT mice compared to no change
sible that some cases of INH-induced hepatotoxicity involve an im- observed in CYP2E1 null mice, which is in contrast to suppression
mune mechanism and others do not. However, this heterogeneity of the abundance of serum free fatty acids and BUN in WT mice
is common to idiosyncratic drug reactions, and the characteristics compared to no changes in CYP2E1-null mice (Cheng et al.,
of INH-induced hepatotoxicity are similar to many other types of 2013). As mentioned earlier, CYP2E1 also plays as a rate-limiting
idiosyncratic drug reactions for which there is good evidence for enzyme in the generation of oxidative stress and cytotoxic metab-
an immune mechanism. The fact that INH can induce a lupus-like olites of INH. Previous studies suggest that hepatic CYP2E1 plays
syndrome makes it clear that it can induce an immune response an essential role in INH-induced hepatotoxicity through the gener-
(Salazar-Paramo et al., 1992; Uetrecht, 2009; Umeki, 1988). ation of free radicals (Huang et al., 2003; Shen et al., 2006, 2008).
Furthermore, recent studies suggested that CYP3A4 might
contribute to INH metabolism, bioactivation and hepatotoxicity.
In a cell culture system containing CYP3A4 supersomes and INH,
Conclusion
cell viability was dramatically decreased and the toxicity was INH-induced hepatotoxicity manifests mainly as hepatocellular
reversed by a CYP3A inhibitor (Vignati et al., 2005). Similar results steatosis and necrosis, and it has been suggested that toxic INH
were noted in a cell line with low expression of superoxide dismu- metabolites bind covalently to cell macromolecules, which had
tase 2 and high expression of CYP3A4, in which INH cytotoxicity been shown in both animal and human case studies. The INH-
was significantly increased (Yoshikawa et al., 2009). In addition, proposed toxic metabolite is hydrazine and animal studies have
anti-CYP3A4 antibody was found in patients with TB who had shown that hydrazine causes steatosis, hepatocyte vacuolation
INH-associated hepatotoxicity (Metushi et al., 2014b). On the other and glutathione depletion. Both lipid vacuoles and mitochondrial
hand, another study did not find evidence for the involvement of swelling was found in hepatocytes. Reduced glutathione levels
CYP3A4 in INH bioactivation and hepatotoxicity in mice. CYP3A4 and reduced activity of glutathione-S-transferase, catalase and
has no effect on the systemic pharmacokinetics of INH in mice superoxide dismutase after INH and/or hydrazine administration
(Liu et al., 2014). However, the exact role of CYP3A in INH metabo- to rats indicates that oxidative stress is involved in INH-induced
lism, bioactivation and toxicity remains unknown. hepatotoxicity. Furthermore, novel experimental data suggest that
Moreover, the role of CYP2B6, as a treatment-response key pre- INH-induced activation of the adaptive and innate immune
dictor, in INH-induced hepatotoxicity was investigated. CYP2B6 system, disruption of endogenous metabolism and mitochondrial
gene polymorphism was found to have a role in INH toxicity dysfunction may be implicated in INH hepatotoxicity.
(Fernandes et al., 2014).
Bacterial lipopolysaccharides (LPS) also suggested playing a cru- Acknowledgments
cial role in the potentiation of INH hepatotoxicity. LPS influences
INH-induced toxicity by affecting its pharmacokinetics. LPS is able This work was partially supported by the National Natural Science
to suppress the protein translation and mRNA transcription of Foundation of China (No.81273604), also by Project Funded by
cytochrome P450 isozymes, including CYP2E1 (Hasegawa et al., the Priority Academic Program Development of Jiangsu Higher
1999; Su et al., 2014). Co-administration of INH and LPS caused Education Institutions (PAPD) and the 111 Project (111-2-07).
serious liver injury and mixed hepatotoxicity in rats, including he-
patocellular injury and cholestasis (Su et al., 2014), which augments
the role of inflammation in INH-induced hepatotoxicity.
Conflict of Interest
The authors did not report any conflict of interest.

CYP2E1 as a Cornerstone in Isoniazid


References
Hepatotoxicity
Anon. 2008. Isoniazid. Tuberc. 88: 112–116.
CYP2E1 is regarded as an important determinant of human suscep- Adhvaryu MR, Reddy N, Parabia MH. 2007. Effects of four Indian medicinal
tibility of industrial and environmental chemicals (Gonzalez, 2005). herbs on Isoniazid-Rifampicin- and Pyrazinamide-induced hepatic

wileyonlinelibrary.com/journal/jat Copyright © 2015 John Wiley & Sons, Ltd. J. Appl. Toxicol. 2015
Hepatotoxicity mechanisms of isoniazid: A mini-review

injury and immunosuppression in guinea pigs. World J. Gastroenterol. Issabeagloo E, Taghizadieh M, Asadi J. 2013. Sage leaves (Salvia miltiorrhiza)
13: 3199–3205. extract attenuates hepatic injury in Isoniazid induced hepatotoxicity in
Aubert J, Begriche K, Knockaert L, Robin MA, Fromenty B. 2011. Increased rats. Life Sci. J. 10: 495–500.
expression of cytochrome P450 2E1 in nonalcoholic fatty liver disease: James L, Roberts D. 2014. Isoniazid hepatotoxicity: progress in understand-
mechanisms and pathophysiological role. Clin. Res. Hepatol. ing the immunologic component. Hepatology 59: 746–748.
Gastroenterol. 35: 630–637. Jaswal A, Sinha N, Bhadauria M, Shrivastava S, Shukla S. 2013. Therapeutic
Bhadauria S, Singh G, Sinha N, Srivastava S. 2007. Isoniazid induces oxida- potential of thymoquinone against anti-tuberculosis drugs induced
tive stress, mitochondrial dysfunction and apoptosis in Hep G2 cells. liver damage. Environ. Toxicol. Pharmacol. 36: 779–786.
Cell. Mol. Biol. 53: 102–114. Keller BJ, Yamanaka H, Thurman RG. 1992. Inhibition of mitochondrial
Bhadauria S, Mishra R, Kanchan R, Tripathi C, Srivastava A, Tiwari A, Sharma respiration and oxygen-dependent hepatotoxicity by six structurally
S. 2010. Isoniazid-induced apoptosis in HepG2 cells: generation of dissimilar peroxisomal proliferating agents. Toxicology 71: 49–61.
oxidative stress and Bcl-2 down-regulation. Toxicol. Mech. Methods 20: Khalili H, Dashti KS, Rasoolinejad M, Rezaie L, Etminani M. 2009. Anti-
242–251. tuberculosis drugs related hepatotoxicity; incidence, risk factors, pattern
Bjornsson E, Kalaitzakis E, Olsson R. 2007. The impact of eosinophilia and of changes in liver enzymes and outcome. DARU 17: 163–167.
hepatic necrosis on prognosis in patients with drug-induced liver injury. Lee KK, Boelsterli UA. 2014. Bypassing the compromised mitochondrial
Aliment. Pharmacol. Ther. 25: 1411–1421. electron transport with methylene blue alleviates efavirenz/isoniazid-
Black M, Mitchell JR, Zimmerman HJ, Ishak KG, Epler GR. 1975. Isoniazid- induced oxidant stress and mitochondria-mediated cell death in mouse
associated hepatitis in 114 patients. Gastroenterology 69: 289–302. hepatocytes. Redox Biol. 2C: 599–609.
Boelsterli UA, Lee KK. 2014. Mechanisms of isoniazid-induced idiosyncratic Lee KK, Fujimoto K, Zhang C, Schwall CT, Alder NN, Pinkert CA, Krueger W,
liver injury: Emerging role of mitochondrial stress. J. Gastroenterol. Rasmussen T, Boelsterli UA. 2013. Isoniazid-induced cell death is precip-
Hepatol. 29: 678–687. itated by underlying mitochondrial complex I dysfunction in mouse
Caro AA, Cederbaum AI. 2004. Oxidative stress, toxicology, and pharmacol- hepatocytes. Free Radic. Biol. Med. 65: 584–594.
ogy of CYP2E1. Annu. Rev. Pharmacol. Toxicol. 44: 27–42. Li F, Lu J, Cheng J, Wang L, Matsubara T, Csanaky IL, Klaassen CD, Gonzalez
Centers for Disease Control and Prevention (CDC) 2010. Severe isoniazid- FJ, Ma X. 2013. Human PXR modulates hepatotoxicity associated with
associated liver injuries among persons being treated for latent tuber- rifampicin and isoniazid co-therapy. Nat. Med. 19: 418–420.
culosis infection - United States, 2004-2008. MMWR Morb. Mortal. Wkly Li J, Uetrecht JP. 2009. D-Penicillamine-induced autoimmunity: relationship
Rep. 59: 224–229. to macrophage activation. Chem. Res. Toxicol. 22: 1526–1533.
Chen X, Xu J, Zhang C, Yu T, Wang H, Zhao M, Duan ZH, Zhang Y, Xu JM, Liu K, Li F, Lu J, Gao Z, Klaassen CD, Ma X. 2014. Role of CYP3A in isoniazid
Xu DX. 2011. The protective effects of ursodeoxycholic acid on isoni- metabolism in vivo. Drug Metab. Pharmacokinet. 29: 219–222.
azid plus rifampicin induced liver injury in mice. Eur. J. Pharmacol. Lyoumi S, Lefebvre T, Karim Z, Gouya L, Puy H. 2013. PXR-ALAS1: A key
659: 53–60. regulatory pathway in liver toxicity induced by isoniazid-rifampicin an-
Chen Y, Xue P, Hou Y, Zhang H, Zheng H, Zhou T, Qu W, Teng W, Zhang Q, tituberculosis treatment. Clin. Res. Hepatol. Gastroenterol. 37: 439–441.
Andersen ME, Pi J. 2013. Isoniazid suppresses antioxidant response Maddrey WC, Boitnott JK. 1973. Isoniazid hepatitis. Ann. Intern. Med. 79:
element activities and impairs adipogenesis in mouse and human 1–12.
preadipocytes. Toxicol. Appl. Pharmacol. 273: 435–441. Mahmoud AM, Germoush MO, Soliman AS. 2014. Berberine attenuates
Cheng J, Krausz KW, Li F, Ma X, Gonzalez FJ. 2013. CYP2E1-dependent isoniazid-induced hepatotoxicity by modulating peroxisome proliferator-
elevation of serum cholesterol, triglycerides, and hepatic bile acids by activated receptor γ, oxidative stress and inflammation. Int. J. Pharmacol.
isoniazid. Toxicol. Appl. Pharmacol. 266: 245–253. 10: 451–460.
Chowdhury A, Santra A, Bhattacharjee K, Ghatak S, Saha DR, Dhali GK. 2006. Metushi IG, Uetrecht J. 2014a. Isoniazid-induced liver injury and immune
Mitochondrial oxidative stress and permeability transition in isoniazid response in mice. J. Immunotoxicol. 11: 383–392.
and rifampicin induced liver injury in mice. J. Hepatol. 45: 117–126. Metushi IG, Uetrecht J. 2014b. Lack of liver injury in Wistar rats treated with
Enriquez-Cortina C, Almonte-Becerril M, Clavijo-Cornejo D, Palestino- the combination of isoniazid and rifampicin. Mol. Cell. Biochem. 387: 9–17.
Dominguez M, Bello-Monroy O, Nuno N, Lopez A, Bucio L, Souza V, Metushi IG, Cai P, Zhu X, Nakagawa T, Uetrecht JP. 2011. A fresh look at the
Hernandez-Pando R, Munoz L, Gutierrez-Ruiz MC, Gomez-Quiroz LE. mechanism of isoniazid-induced hepatotoxicity. Clin. Pharmacol. Ther.
2013. Hepatocyte growth factor protects against isoniazid/rifampicin- 89: 911–914.
induced oxidative liver damage. Toxicol. Sci. 135: 26–36. Metushi IG, Nakagawa T, Uetrecht J. 2012. Direct oxidation and covalent
Humayun F, Tahir M, Lone KP, Munir B, Ahmad A, Latif W. 2014. Protective binding of isoniazid to rodent liver and human hepatic microsomes:
effect of ethanolic extract of propolis on isoniazid induced hepatotoxic- humans are more like mice than rats. Chem. Res. Toxicol. 25: 2567–2576.
ity in male albino mice. Biomedica 30: 85–91. Metushi IG, Sanders C, Lee WM, Uetrecht J. 2014a. Detection of anti-
Fatima R, Ashraf M, Ejaz S, Rasheed MA, Altaf I, Afzal M, Batool Z, Saleem U, isoniazid and anti-cytochrome P450 antibodies in patients with
Anwar K. 2013. In vitro toxic action potential of anti tuberculosis drugs isoniazid-induced liver failure. Hepatology 59: 1084–1093.
and their combinations. Environ. Toxicol. Pharmacol. 36: 501–513. Metushi IG, Sanders C, The Acute Liver Study G, Lee WM, Uetrecht J. 2014b.
Fernandes DC, Santos NP, Moraes MR, Braga AC, Silva CA, Ribeiro-Dos-Santos Detection of anti-isoniazid and anti–cytochrome P450 antibodies in
A, Santos S. 2014. Association of the CYP2B6 gene with anti-tuberculosis patients with isoniazid-induced liver failure. Hepatology 59: 1084–1093.
drug-induced hepatotoxicity in a Brazilian Amazon population. Int. J. Mujahid M, Siddiqui HH, Hussain A, Hussain MS. 2013. Hepatoprotective
Infect. Dis. 33C: 28–31. effects of Adenanthera pavonina (Linn.) against anti-tubercular drugs-
Georgieva N, Gadjeva V, Tolekova A, Dimitrov D. 2005. Hepatoprotective induced hepatotoxicity in rats. Pharmacognosy J. 5: 286–290.
effect of isonicotinoylhydrazone SH7 against chronic isoniazid toxicity. Nanashima K, Mawatari T, Tahara N, Higuchi N, Nakaura A, Inamine T, Kondo
Pharmazie 60: 138–141. S, Yanagihara K, Fukushima K, Suyama N, Kohno S, Tsukamoto K. 2012.
Gonzalez FJ. 2005. Role of cytochromes P450 in chemical toxicity and oxida- Genetic variants in antioxidant pathway: Risk factors for hepatotoxicity
tive stress: studies with CYP2E1. Mutat. Res. 569: 101–110. in tuberculosis patients. Tuberc. 92: 253–259.
Haasio K, Koponen A, Penttila KE, Nissinen E. 2002. Effects of entacapone Obogwu MB, Akindele AJ, Adeyemi OO. 2014. Hepatoprotective and in vivo
and tolcapone on mitochondrial membrane potential. Eur. J. Pharmacol. antioxidant activities of the hydroethanolic leaf extract of Mucuna
453: 21–26. pruriens (Fabaceae) in antitubercular drugs and alcohol models. Chin.
Hasegawa T, Takagi K, Kitaichi K. 1999. Effects of bacterial endotoxin on J Nat. Med. 12: 273–283.
drug pharmacokinetics. Nagoya J. Med. Sci. 62: 11–28. Ohno M, Yamaguchi I, Yamamoto I, Fukuda T, Yokota S, Maekura R, Ito M,
Hosford JD, von Fricken ME, Lauzardo M, Chang M, Dai Y, Lyon JA, Shuster J, Yamamoto Y, Ogura T, Maeda K, Komuta K, Igarashi T, Azuma J. 2000.
Fennelly KP. 2015. Hepatotoxicity from antituberculous therapy in the Slow N-acetyltransferase 2 genotype affects the incidence of isonia-
elderly: A systematic review. Tuberculosis (Edinb.) 95(2): 112–122. zid and rifampicin-induced hepatotoxicity. Int. J. Tuberc. Lung Dis. 4:
Huang YS, Chern HD, Su WJ, Wu JC, Chang SC, Chiang CH, Chang FY, Lee SD. 256–261.
2003. Cytochrome P450 2E1 genotype and the susceptibility to antitu- Pal R, Rana SV, Vaiphei K, Singh K. 2008. Isoniazid-rifampicin induced lipid
berculosis drug-induced hepatitis. Hepatology 37: 924–930. changes in rats. Clin. Chim. Acta 389: 55–60.
Ingawale DK, Mandlik SK, Naik SR. 2014. Models of hepatotoxicity and the Park WB, Kim W, Lee KL, Yim JJ, Kim M, Jung YJ, Kim NJ, Kim DH, Kim YJ,
underlying cellular, biochemical and immunological mechanism(s): a Yoon JH, Oh MD, Lee HS. 2010. Antituberculosis drug-induced liver
critical discussion. Environ. Toxicol. Pharmacol. 37: 118–133. injury in chronic hepatitis and cirrhosis. J. Infect. 61: 323–329.

J. Appl. Toxicol. 2015 Copyright © 2015 John Wiley & Sons, Ltd. wileyonlinelibrary.com/journal/jat
H. M. Hassan et al.

Perwitasari DA, Atthobari J, Wilffert B. 2014. Pharmacogenetics of Shishido S, Koga H, Harada M, Kumemura H, Hanada S, Taniguchi E,
isoniazid-induced hepatotoxicity. Drug Metab. Rev. 1–7. DOI: Kumashiro R, Ohira H, Sato Y, Namba M, Ueno T, Sata M. 2003. Hydro-
10.3109/03602532.2014.984070 gen peroxide overproduction in megamitochondria of troglitazone-
Pessayre D, Mansouri A, Berson A, Fromenty B. 2010. Mitochondrial treated human hepatocytes. Hepatology 37: 136–147.
involvement in drug-induced liver injury. Handbk. Exp. Pharmacol. Singh A, Bhat TK, Sharma OP. 2011. Clinical biochemistry of hepatotoxicity.
196: 311–365. J. Clinic. Toxicol. S4:001.
Petrescu I, Tarba C. 1997. Uncoupling effects of diclofenac and aspirin in the Singh C, Jodave L, Bhatt TD, Gill MS, Suresh S. 2014. Hepatoprotective agent
perfused liver and isolated hepatic mitochondria of rat. Biochim. tethered isoniazid for the treatment of drug-induced hepatotoxicity:
Biophys. Acta 15: 385–394. Synthesis, biochemical and histopathological evaluation. Toxicol. Rep.
Ramappa V, Aithal GP. 2013. Hepatotoxicity related to anti-tuberculosis 1: 885–893.
drugs: Mechanisms and management. J. Clin. Exp. Hepatol. 3: 37–49. Singh M, Sasi P, Rai G, Gupta V, Amarapurkar D, Wangikar P. 2011. Studies
Rao CV, Rawat AKS, Singh AP, Singh A, Verma N. 2012. Hepatoprotective on toxicity of antitubercular drugs namely isoniazid, rifampicin, and
potential of ethanolic extract of Ziziphus oenoplia (L.) Mill roots against pyrazinamide in an in vitro model of HepG2 cell line. Med. Chem. Res.
antitubercular drugs induced hepatotoxicity in experimental models. 20: 1611–1615.
Asian Pac. J. Trop. Med. 5: 283–288. Singla N, Gupta D, Birbian N, Singh J. 2014. Association of NAT2, GST and
Richards VE, Chau B, White MR, McQueen CA. 2004. Hepatic gene expres- CYP2E1 polymorphisms and anti-tuberculosis drug-induced hepatotox-
sion and lipid homeostasis in C57BL/6 mice exposed to hydrazine or icity. Tuberculosis (Edinb.) 94: 293–298.
acetylhydrazine. Toxicol. Sci. 82: 318–332. Su Y, Zhang Y, Chen M, Jiang Z, Sun L, Wang T, Zhang L. 2014. Lipopolysaccha-
Saad EI, El-Gowilly SM, Sherhaa MO, Bistawroos AE. 2010. Role of oxidative ride exposure augments isoniazide-induced liver injury. J. Appl. Toxicol.
stress and nitric oxide in the protective effects of alpha-lipoic acid and 34(12): 1436–1442.
aminoguanidine against isoniazid-rifampicin-induced hepatotoxicity in Tafazoli S, Mashregi M, O’Brien PJ. 2008. Role of hydrazine in isoniazid-
rats. Food Chem. Toxicol. 48: 1869–1875. induced hepatotoxicity in a hepatocyte inflammation model. Toxicol.
Salazar-Paramo M, Rubin RL, Garcia-De La Torre I. 1992. Systemic lupus Appl. Pharmacol. 229: 94–101.
erythematosus induced by isoniazid. Ann. Rheum. Dis. 51: 1085–1087. Tostmann A, Boeree MJ, Aarnoutse RE, de Lange WC, van der Ven AJ,
Santos NP, Callegari-Jacques SM, Ribeiro Dos Santos AK, Silva CA, Vallinoto Dekhuijzen R. 2008a. Antituberculosis drug-induced hepatotoxicity:
AC, Fernandes DC, de Carvalho DC, Santos SE, Hutz MH. 2013. N-acetyl concise up-to-date review. J. Gastroenterol. Hepatol. 23: 192–202.
transferase 2 and cytochrome P450 2E1 genes and isoniazid-induced Tostmann A, Boeree MJ, Peters WH, Roelofs HM, Aarnoutse RE, van der Ven
hepatotoxicity in Brazilian patients. Int. J. Tuberc. Lung Dis. 17: 499–504. AJ, Dekhuijzen PN. 2008b. Isoniazid and its toxic metabolite hydrazine in-
Sarich TC, Zhou T, Adams SP, Bain AI, Wall RA, Wright JM. 1995. A model of duce in vitro pyrazinamide toxicity. Int. J. Antimicrob. Agents 31: 577–580.
isoniazid-induced hepatotoxicity in rabbits. J. Pharmacol. Toxicol. Uetrecht J. 2009. Immunoallergic drug-induced liver injury in humans.
Methods 34: 109–116. Semin. Liver Dis. 29: 383–392.
Sarich TC, Youssefi M, Zhou T, Adams SP, Wall RA, Wright JM. 1996. Role of Uetrecht J, Naisbitt DJ. 2013. Idiosyncratic adverse drug reactions: current
hydrazine in the mechanism of isoniazid hepatotoxicity in rabbits. Arch. concepts. Pharmacol. Rev. 65: 779–808.
Toxicol. 70: 835–840. Umeki S. 1988. A case of isoniazid-induced systemic lupus erythematosus.
Sarich TC, Adams SP, Petricca G, Wright JM. 1999. Inhibition of isoniazid- Kekkaku 63: 713–717.
induced hepatotoxicity in rabbits by pretreatment with an amidase in- Vignati L, Turlizzi E, Monaci S, Grossi P, Kanter R, Monshouwer M. 2005. An
hibitor. J. Pharmacol. Exp. Ther. 289: 695–702. in vitro approach to detect metabolite toxicity due to CYP3A4-
Saukkonen JJ, Cohn DL, Jasmer RM, Schenker S, Jereb JA, Nolan CM, dependent bioactivation of xenobiotics. Toxicology 216: 154–167.
Peloquin CA, Gordin FM, Nunes D, Strader DB, Bernardo J, Walubo A, Smith P, Folb PI. 1998. The role of oxygen free radicals in
Venkataramanan R, Sterling TR. 2006. An Official ATS Statement: isoniazid-induced hepatotoxicity. Methods Find. Exp. Clin. Pharmacol.
Hepatotoxicity of antituberculosis therapy. Am. J. Respir. Crit. Care Med. 20: 649–655.
174: 935–952. Waring JF, Anderson MG. 2005. Idiosyncratic toxicity: mechanistic insights
Schwab CE, Tuschl H. 2003. In vitro studies on the toxicity of isoniazid in gained from analysis of prior compounds. Curr. Opin. Drug Discov. Dev.
different cell lines. Hum. Exp. Toxicol. 22: 607–615. 8: 59–65.
Sharifzadeh M, Rasoulinejad M, Valipour F, Nouraie M, Vaziri S. 2005. Evalu- Yew WW, Leung CC. 2006. Antituberculosis drugs and hepatotoxicity.
ation of patient-related factors associated with causality, preventability, Respirology 11: 699–707.
predictability and severity of hepatotoxicity during antituberculosis Yoshikawa Y, Hosomi H, Fukami T, Nakajima M, Yokoi T. 2009. Establishment
[correction of antituberclosis] treatment. Pharmacol. Res. 51: 353–358. of knockdown of superoxide dismutase 2 and expression of CYP3A4
Sharma SK. 2004. Antituberculosis drugs and hepatotoxicity. Infect. Genet. cell system to evaluate drug-induced cytotoxicity. Toxicol. In Vitro
Evol. 4: 167–170. 23: 1179–1187.
Shaw PJ, Ganey PE, Roth RA. 2010. Idiosyncratic drug-induced liver injury Yue J, Peng R. 2009. Does CYP2E1 play a major role in the aggravation of
and the role of inflammatory stress with an emphasis on an animal isoniazid toxicity by rifampicin in human hepatocytes? Br. J. Pharmacol.
model of trovafloxacin hepatotoxicity. Toxicol. Sci. 118: 7–18. 157: 331–333.
Shayakhmetova GM, Bondarenko LB, Voronina AK, Anisimova SI, Matvienko Yue J, Peng RX, Yang J, Kong R, Liu J. 2004. CYP2E1 mediated isoniazid-
AV, Kovalenko VM. 2015. Induction of CYP2E1 in testes of isoniazid- induced hepatotoxicity in rats. Acta Pharmacol. Sin. 25: 699–704.
treated rats as possible cause of testicular disorders. Toxicol. Lett. 234: Yue J, Dong G, He C, Chen J, Liu Y, Peng R. 2009a. Protective effects of
59–66. thiopronin against isoniazid-induced hepatotoxicity in rats. Toxicology
Shen C, Zhang H, Zhang G, Meng Q. 2006. Isoniazid-induced hepatotoxicity 264: 185–191.
in rat hepatocytes of gel entrapment culture. Toxicol. Lett. 167: 66–74. Yue J, Peng R, Chen J, Liu Y, Dong G. 2009b. Effects of rifampin on CYP2E1-
Shen C, Zhang G, Meng Q. 2007. An in vitro model for long-term hepatotox- dependent hepatotoxicity of isoniazid in rats. Pharmacol. Res. 59: 112–119.
icity testing utilizing rat hepatocytes entrapped in micro-hollow fiber Zhai Q, Lu S, Lin Y, Yang Q, Yu B. 2008. Oxidative stress potentiated by
reactor. Biochem. Eng. J. 34: 267–272. diallylsulfide, a selective CYP2E1 inhibitor, in isoniazid toxic effect on
Shen C, Meng Q, Zhang G, Hu W. 2008. Rifampicin exacerbates isoniazid- rat primary hepatocytes. Toxicol. Lett. 183: 95–98.
induced toxicity in human but not in rat hepatocytes in tissue-like Zhang Z-H, Tang J-H, Zhan Z-L, Zhang X-L, Wu H-H, Hou Y-N. 2012. Cellular
cultures. Br. J. Pharmacol. 153: 784–791. toxicity of isoniazid together with rifampicin and the metabolites of iso-
Shih TY, Pai CY, Yang P, Chang WL, Wang NC, Hu OY. 2013. A novel mech- niazid on QSG-7701 hepatocytes. Asian Pac. J. Trop. Med. 5: 306–309.
anism underlies the hepatotoxicity of pyrazinamide. Antimicrob. Agents
Chemother. 57: 1685–1690.

wileyonlinelibrary.com/journal/jat Copyright © 2015 John Wiley & Sons, Ltd. J. Appl. Toxicol. 2015

You might also like