Abstract
Liver injury resulting from exposure to drugs and environmental chemicals is a major health problem. Endoplasmic reticulum stress (ER stress) is considered to be an important factor in a wide range of diseases, such as cancer, neurological and cardiovascular disease, diabetes, and inflammatory diseases. The role of ER stress in drug-induced and environmental toxicant-induced liver toxicity has been underestimated in the past; emerging evidence indicates that ER stress makes a substantial contribution to the pathogenesis of drug-induced liver toxicity. In this review, we summarize current knowledge on drugs and environmental toxicants that trigger ER stress in liver and on the underlying molecular mechanisms. We also discuss experimental approaches for ER stress studies.
Keywords: Endoplasmic reticulum (ER) stress, liver toxicity, signaling pathways
INTRODUCTION
Drug-induced liver injury, one of the leading causes of drug failure and withdrawal from the market after approval, is a serious concern for public health as well as for drug development and approval [1–3]. Multiple factors and mechanisms are involved in drug-induced liver toxicity. For example, drug metabolism via liver metabolizing enzymes generates reactive electrophilic metabolites and free radicals. These toxic metabolites can covalently bind to proteins, lipids, and DNA and cause such adverse consequences as oxidative stress, glutathione depletion, lipid peroxidation, and cell death. Alteration of the immune response has often been described as an important mechanism of liver toxicity. Gunawan and Kaplowitz have summarized the mechanisms of drug-induced liver toxicity [4]. More recently, endoplasmic reticulum (ER) stress has begun to be recognized and is often associated with liver diseases including, but not restricted to, hepatitis, alcoholic liver disease, fatty liver disease, and drug-induced liver toxicity.
The ER is a critical organelle that plays a vital role in many cellular processes including protein synthesis, folding, assembly, trafficking, postmodulation, quality control of both secretory and membrane proteins, lipid synthesis, and regulation of intracellular calcium hemostasis. Disturbances of ER function by stimuli such as altered cellular redox status, oxidative stress, unbalanced calcium homeostasis, hypoxia, or energy deprivation lead to the accumulation of unfolded proteins in the ER, which is generally termed ER stress [5–9]. To re-establish ER function and restore homeostasis, a specific signaling pathway, the unfolded protein response (UPR), is activated as an adaptive mechanism. This adaptive mechanism is a transcriptional program to up-regulate the expression of genes involved in protein folding and quality control [9,10]. Excessive and prolonged ER stress causes apoptosis and necrotic cell death. Three key molecules that are present in the ER membrane—PERK (Protein Kinase R/PKR-like ER kinase), IRE1α (inositol-requiring enzyme 1α), and ATF6 (activation of transcription factor 6)—are ER stress sensors. When cells are under ER stress, glucose-regulated protein 78 (Grp78) dissociates from these three transmembrane ER signaling proteins, thereby leading to the activation of three major UPR branches (Figure 1). These three UPR transducers are activated by translocation of ATF6 to the Golgi and phosphorylation of PERK and IRE1α. (A) ATF6 is translocated to the Golgi and then cleaved by the Golgi resident proteases. The cleaved form of ATF6 is released to the cytosol and travels to the nucleus to regulate gene expression. (B) Phosphorylation of PERK leads to a reduction of the protein load on the ER by decreasing protein synthesis mediated by eIF2α phosphorylation. The expression of transcription factor 4 (ATF4), a member of the basic leucine-zipper (bZIP) family of transcription factors that regulate the promoters of numerous UPR genes, is induced. (C) Phosphorylation of IRE1α initiates an intron excision from the mRNA for a transcription factor, XBP1 (X-box binding protein 1), to produce a spliced form XBP1S. XBP1S binds to promoters of several genes, resulting in an increased expression of genes involved in restoring protein folding or degrading unfolded proteins [10,11]. These signaling pathways are referred to as pro-survival unfolded protein responses [9]. Excessive and prolonged ER stress stimulates death signals, which provoke apoptosis and necrotic cell death. For instance, pro-apoptotic protein C/EBP CCAAT/enhancer binding protein homologous protein (CHOP) is induced by ATF4 activation via the PERK-eIF2α pathway. ER resident caspase 12 (human form: caspase 4) is activated and becomes involved in ER-stress associated apoptosis [12].
Figure 1.
The unfolded protein response: Three UPR transducers are activated upon stimulation of ER. (A) ATF6 is translocated to the Golgi and then cleaved by resident Golgi proteases. The cleaved form of ATF6 is released to the cytosol and then travels to the nucleus to regulate gene expression; (B) Phosphorylation of PERK leads a reduction in the protein load on the ER by decreasing protein synthesis through eIF2α phosphorylation. The expression of transcription factor 4 (ATF4) is induced; ATF4 is a member of the bZIP family of transcription factors that regulate the promoters of numerous UPR genes; (C) Phosphorylation of IRE1α initiates an intron excision from the mRNA for the transcription factor, XBP1 (X-box binding protein 1), to produce a spliced form XBP1S. XBP1S binds to promoters of several genes, resulting in increased expression of genes involved in restoring protein folding or degrading unfolded proteins.
ER stress has been reported to be associated with a wide range of human diseases [13–15]; these include, but are not restricted to, cancer, neurological, kidney, lung, and cardiovascular disease, diabetes, and inflammatory diseases. A large number of ER stress review articles focusing on cancer [16–24]; Alzheimer disease [25–29]; Parkinson [30,31], kidney [32–34], lung [35–37], cardiovascular [38–42], and inflammatory diseases [43–47]; obesity [48–50]; gastrointestinal disease [51]; and diabetes [50,52–60] have recently been published. In addition, relationships between ER stress and environmental toxicants have also been summarized [61,62]. In this article we reviewed literature on drugs and some environmental toxicants in which the underlying mechanisms may be ER stress. We also summarize research approaches in ER stress studies.
ER STRESS AND DRUG-INDUCED LIVER TOXICITY
Acetaminophen
Acetaminophen overdose is the leading cause of drug-induced liver failure in the United State [1,63]. The liver toxicity of acetaminophen is known to be initiated by N-acetylbenzoquinoneimine, an active metabolite produced by the cytochrome P450 [64]. The toxicity of acetaminophen and its mechanisms have been extensively studied for decades because of its complexity, as it involves multiple signaling pathways and also resembles many aspects of drug-induced liver injury; however, the exact mode of cell death is not completely understood [65]. Recent studies have demonstrated a role of ER stress in acetaminophen-induced liver toxicity [65–68]. A sublethal dose of acetaminophen results in activation of ER stress markers include ATF6 and CHOP. In addition, caspase-12, an ER-resident caspase, is transiently activated [68]. It has been suggested that the mode of death for acetaminophen’s liver toxicity is caspase-independent apoptosis, initiated by activation of polymerase 1 (PARP-1) [67,68], which can be attenuated by the PARP-1 inhibitor BGP-15 (O-[3-piperidino-2-hydroxy-1-propyl]-nicotinic amidoxime). Interestingly, BGP-15 did not protect against the ER-stress related early events of redox stress, such as glutathione depletion, and BGP-15 did not counteract any ER stress signaling events. Thus, the protection against acetaminophen-induced apoptosis by BGP-15 is not likely to be due to inhibition of ER stress pathways. The authors suggested that caspase-independent apoptosis is initiated by the stressed ER and nucleus [67].
A recent study assessed the roles of ER stress in acetaminophen-induced liver toxicity using CHOP knock-out mice [65]. ER stress pathways were activated when toxic doses were given orally to the wild-type mice. While all three branches of UPR were activated, PERK-elF2-CHOP cascade seemed to be the most important signaling pathway. While wild-type mice developed extensive liver necrosis, CHOP knock-out mice were partially protected from liver toxicity, showed increased survival, and hepatocyte proliferation at the sites of necrosis, indicating that CHOP plays a critical role in acetaminophen’s toxicity and that up-regulation of CHOP compromises hepatocyte survival by reducing regeneration [65]. Unlike previous reports demonstrating that ER stress is an early event of apoptotic cell death and glutathione depletion [67,68], this study reported that ER stress occurred after a cascade of events such as glutathione depletion and oxidative stress [65]. The discrepancy is not clear; it could be due to different routes of administration (i.p. injection [67] vs. oral administration [65]). Further studies are warranted to explain the differences.
Anti-HIV Drugs
Liver toxicity represents one of the most common complications of immunodeficiency virus (HIV) treatments. Side effects have been reported to be associated with different classes of anti-HIV drugs, including nucleoside reverse transcriptase inhibitors, non-nucleoside analogue reverse transcriptase inhibitors, and HIV protease inhibitors [69].
Efavirenz
Efavirenz is a non-nucleoside analogue reverse transcriptase inhibitor. Although generally considered safe, some evidence indicates that efavirenz disrupts lipid metabolism [70] and induces liver fibrosis [69,71]. An in vitro study demonstrated that a battery of ER stress markers was induced when primary human hepatocytes and Hep3B cells were exposed to clinically relevant concentrations of efavirenz [72,73]. The ER stress responses included induction of CHOP and GRP78, phosphorylation of eIF2a, and the production of XBP1 isoform XBP1S. Morphological changes, such as dilation of ER membranes, were also observed. Because mitochondrial dysfunction is one the mechanisms responsible for efavirenz-induced liver damage [74,75], the interplay between ER stress and mitochondrial dysfunction was investigated in Rhoo cells, liver cells with nonfunctional mitochondria [76]. Efavirenz-treated Rhoo cells showed no appreciable increase in ER stress signals, such as the induction of CHOP and GRP78, compared to wild-type cells, demonstrating that mitochondria are involved in the efavirenz-induced ER stress response.
Lopinavir, ritonavir, saquinavir, nelfinavir, atazanavir, and amprenavir
HIV protease inhibitors
It has been documented that numerous HIV pro-tease inhibitors induced ER stress in both liver cells and in the livers of animals [77–79]. In an in vitro model, five HIV protease inhibitors (lopinavir, ritonavir, saquinavir, nelfinavir, and atazanavir) were studied for their ER stress-inducing abilities at clinically relevant concentrations [78]. The induction of CHOP, ATF4, ATF3, and some ER chaperones indicated that these HIV protease inhibitors caused ER stress in HepG2 cells, the ER stress response was attributed their proteasome inhibiting functions.
The effect of three HIV protease inhibitors, amprenavir, atazanavir, and ritonavir, on ER stress was studied and compared in primary rodent hepatocytes. Lipid metabolism was also investigated in the same study because lipid disturbances have been reported to be associated with the use of HIV protease inhibitors [79]. Both atazanavir- and ritonavir-induced ER stress and apoptosis, and increased sterol regulatory element-binding proteins (SREBP) levels, while amprenavir, at the same concentrations, had no effect.
Kou and colleagues investigated lopinavir- and ritonavir-induced ER stress and also examined the synergistic effect of alcohol-induced liver lipid accumulation and ER stress response in mice, primary human hepatocytes, and primary mouse hepatocytes [77]. Both lopinavir- and ritonavir-induced ER stress response in mice, but did not show a significant increase in serum alanine aminotransferase (ALT) levels. Co-administration of these HIV protease inhibitors with alcohol caused an increase in ALT and a synergistic increase in lipid accumulation and ER stress response. In addition, co-administration of alcohol and lopinavir or ritonavir reduced intracellular calcium levels and also potentiated cell death. These data suggest that HIV protease inhibitors potentiate alcohol-induced ER stress and worsen liver functions [77]. The underlying mechanisms that contribute to the liver toxicity of HIV protease inhibitors are not, at present, well understood. The studies described previously pinpointed that at therapeutic concentrations, HIV protease inhibitors induce ER stress response as major ER stress markers had been altered (activation of SREBPs, ATF4, CHOP, caspase-12, and splicing of XBP1), implying that ER stress maybe a new mechanism for the liver toxicities of HIV protease inhibitors.
Anti-type 2 Diabetes Drugs
Agonists of peroxisome proliferator-activated receptor γ (PPARγ) are oral drugs designed to treat type 2 (insulin-resistant) diabetes. Ciglitazone, the first PPARγ agonist to enter clinical trials, was not marketed because of liver toxicity. Troglitazone, the first PPARγ agonist approved by the US Food and Drug Administration (FDA), was withdrawn from the market after cases of severe liver failure and death were reported [80,81]. The mechanisms involved in the liver toxicity include mitochondrial dysfunction, apoptosis, disruption of calcium hemostasis, oxidative stress, and ER stress [82–89]. Studies using GN4 rat liver epithelial cells demonstrated that both troglitazone and ciglitazone activated the calcium-dependent p38 mitogen-activated protein kinase (MAPK) pathway and also induced ER stress [88,90]. ER stress induction by troglitazone and ciglitazone was indicated by the activation of the two key molecules (PERK and eIF2α) in the PERK branch [88]. A link between p38/MAPK activation and induction ER stress was also demonstrated. Treatment with KN-93, an inhibitor of calcium/calmodulin-dependent kinase II (CaMKII), which is an upstream regulator of p38, attenuated the activation of both MAPK (MKK3/6) and p38 and the phosphorylation of eIF2α. In addition, the activation of MAPK had shown to correlate with the phosphorylation of PERK and eIF2α. In contrast, the two other glitazones (rosiglitazone and pioglitazone), which have lower liver toxicities, failed to induce either MAPK or eIF2α, suggesting that induction of ER stress and the activation of MAPK signaling pathways may play critical roles in the effects of troglitazone or ciglitazone on cell growth and maturation [88].
Anti-depression Drugs
Sertraline is used for the treatment of depression, and also for the treatment of panic, obsessive-compulsive, and post-traumatic stress disorders. Although it is generally considered safe, acute liver failure due to sertraline use has been documented [91–99]. Sertraline was demonstrated to be toxic in primary rat hepatocytes and human HepG2 cells; mitochondrial dysfunction and apoptosis are the underlying mechanisms [100,101]. Besides mitochondrial dysfunction and apoptosis, ER stress was found to contribute to sertraline’s liver toxicity (SC and LG, unpublished data). The expression of typical ER stress markers, such as PERK, IRE1α, and CHOP, was significantly increased in sertraline-treated hepatic cells. The XPB1S isoform was also observed. The ER stress response was also measured quantitatively with two reporter assays, namely, Gaussia luciferase (Gluc) and secreted alkaline phosphatase (SEAP). Sertraline significantly decreased the activities of both Gaussia luciferase and SEAP. In addition, the interplay between the apoptosis and ER stress was studied. ER stress was an early response triggering apoptosis because ER stress occurred at earlier time points and lower concentrations compared to those for apoptosis. In addition, administration of a potent ER stress inhibitor, 4-phenylbutyrate (4-PBA) attenuated sertraline-caused apoptosis. Moreover, sertraline treatment increased the expression of tumor necrosis factor and the phosphorylation of JNK, ERK1/2, and p38, which are the major players in the MAPK signaling pathway; silencing MAP4K4 (the upstream kinase of JNK) attenuated both ER stress and apoptosis; and a JNK-specific inhibitor efficiently prevented the decreased activity of secreted Gluc, indicating that MAPK signaling pathway signaling is involved in both apoptosis and ER stress [SC and LG, unpublished data]. Taken together, these studies demonstrate that the mechanism of sertraline-induced liver toxicity is a complex with crosstalk between mitochondrial dysfunction, apoptosis, and ER stress, with the involvement the MAPK signaling pathway.
ER STRESS AND ALCOHOL-INDUCED LIVER TOXICITY
Excessive alcohol consumption leads to liver damage since alcohol is a direct hepatotoxin; it also potentiates other liver diseases, including chronic hepatitis and nonalcoholic fatty liver disease [102]. The mechanisms of alcohol-induced liver damage are complex and multifactorial. DNA adducts, protein adducts, glutathione depletion, lipid peroxidation, apoptosis, mitochondrial damage, inflammation, and cell-cycle arrest have been shown to be involved in alcohol-associated liver toxicity [102,103]. Alcohol-induced liver toxicity is metabolism-mediated; the reactive metabolite, acetaldehyde, created via alcohol dehydrogenase, aldehyde dehydrogenase, and CYP2E1, is a major contributor of the alcohol’s toxicity. ER stress has been reported to participate in alcohol-caused liver toxicity in both animal and cell models [104–107].
Alcoholic liver injury has been studied using intragastric alcohol-fed mice as a model system; this model reproduces the pathology features of early alcohol liver and demonstrated the involvement of ER stress [105]. Fatty liver, increased hepatic inflammation, apoptosis, and hyperhomocysteinemia were observed in these alcohol-fed animals. The expression of ER stress–related genes, including GRP78, GRP 94, CHOP/GADD153, and caspase-12, was found to be up-regulated, indicating that ER stress response may contribute to the pathologic features of alcoholic liver disease [105]. ER stress signals GRP78, caspase 12, and sterol regulatory element binding protein-1c (SREBP-1c) were activated in alcohol fed micropigs [107].
The role of CHOP in hepatocellular apoptosis and liver injury has been studied using CHOP knock-out mice [106]. Although no significant differences in pathological parameters and occurrence of ER stress were observed between wild-type and knock-out mice, apoptosis was present in wild-type mice but absent in CHOP knock-out mice. These results implied that ER stress may be responsible for apoptosis through CHOP and that CHOP up-regulation occurred later than apoptosis [106].
A recent study assessed the effect of alcohol on the integrated stress response (ISR) signaling pathway and defined the role of CYP2E1 in this response, using primary human hepatocytes and CYP2E1 over-expressing HepG2 cells [104]. ISR triggered by diverse stresses is a defense mechanism, which leads to suppression of global protein synthesis. ISR is one branch of ER stress response and ATF4 is a key component whose translation is turned on by the phosphorylation of eIF2a [6] (Figure 1). In alcohol-treated primary human hepatocytes and HepG2 cells over-expressing CYP2E1, ATF4, the master coordinator of ISR, was up-regulated. In CYP2E1 over-expressing cells, a set of ISR target genes, including HMOX-1, GCLC, AsnS, IGFBP-1, GADD34, CHOP, ATF3, and CHAC1, was induced. The antioxidant glutathione and CYP2E1 inhibition protected against both oxidative stress and ISR induced by alcohol. These results demonstrate that both CYP2E1 and ATF4-mediated ISR are detrimental cellular effects in alcohol-induced liver disease [104].
ER STRESS AND ENVIRONMENTAL TOXICANT-ASSOCIATED LIVER TOXICITY
Acrolein
Acrolein, a common environmental pollutant and a major component of cigarette smoke, is a highly reactive α,β-unsaturated aldehyde and is cytotoxic to many cells, including hepatocytes. The toxicities are considered to be due to oxidative stress via loss of glutathione and the formation of Michael-type addition adducts with proteins and DNA [108–111]. In hepatic cells, including both primary human hepatocytes and HepG2 hepatoma cells, acrolein caused apoptosis and necrosis, and triggered ER stress at pathophysiological concentrations [112]. Numerous typical ER stress markers, including eIF2α, ATF-3 and -4, and CHOP, were induced, presenting strong evidence of ER stress; however, the protective/adaptive components of ER stress, such as chaperone proteins GRP78 and GRP94 (assisting in protein refolding), were not changed. It appears that exposure to acrolein selectively impairs the ER protective mechanisms, thus leading cell death. The mechanisms behind the failure of the ER adaptive response in this case remain to be determined [112].
Arsenic
Humans are exposed to arsenic and inorganic arsenic compounds via different routes, such as drinking water, contaminated foods or soil, and dermal exposure. Such inorganic arsenic exposure is associated with adverse health effects, including skin lesions and increased risks of cancer of the lung, skin, liver, kidney, and bladder [113,114]. The mechanisms underlying arsenic-associated toxicity, such as DNA damage, formation of DNA and protein adducts, and production of reactive oxygen species (ROS), have been recognized [115–117]. Inorganic arsenicals are transformed by the liver into monomethylated and dimethylated arsenicals through methylation reactions and excreted into urine as pentavalent methylated forms. Although inorganic arsenic had been extensity studied, it still remains unclear which forms of inorganic arsenic are more toxic to particular organs; conclusions appear to also depend on the experimental approaches.
A study aimed to characterize and compare the different forms of inorganic arsenic-associated liver toxicity demonstrated that ER stress was induced by trivalent dimethylarsinous acid (DMAIII) but not the forms of trivalent ar-senite (iAsIII) and monomethylarsonous acid (MMAIII) [118]. The phosphorylation of PERK was induced in rat liver RLC-16 cells treated with DMAIII, but no significant PERK activation was detected when cells were treated with iAsIII or MMAIII. In the same study, it was reported that ER was a site of generation of ROS after DMAIII exposure, indicating that DMAIII targeted the ER for its toxicity. ER stress induction and ROS generation in the ER occurred simultaneously, as both were attenuated by the addition of cycloheximide, a protein biosynthesis inhibitor [118].
APPROACHES FOR DETECTING AND MONITORING ER STRESS
The study of ER stress is a growing research area and various experimental approaches have been applied. The most commonly used methods include immunohistochemistry and Western blots for protein detection, qPCR measurement of ER stress markers, PCR-based assays to detect the alternate splicing of XBP1 mRNA, and reporter assays for measuring the activity of ER stress-responding transcriptional factors such as XBP1 and ATF6 [119,120]. Recently, we developed two reporter assays in HepG2 cells, with secreted alkaline phosphatase (SEAP) and secreted Gaussia luciferase (Gluc) as reporters, and demonstrating the usefulness of these two in vitro assays [SC and LG, unpublished]. Genetically modified animal models include transgenic models and knock-out models of essential ER stress genes [65,121]. Descriptions of different methods follow.
Protein detection of ER stress markers: induction of various protein expression such as Grp78, PDI, CHOP, ATF4, ATF6, spliced XBP1, IRE1, phosphorylated-eIF2α, and phosphorylated-PERK have been reported in response to ER stress and these markers are very often to be chosen for ER stress identification. The detection of increased expression of these markers also can help to elucidate mechanisms of ER stress, that is, which branch (es) of UPR is altered (Figure 1). Both standard Western blots and immunohistochemistry have been applied.
Measurement of ER stress marker gene expression: a set of ER stress-related genes, including, but not restricted to, Atf4, Atf6, Bip (Grp78), Erp72, Chop, Perk, Ire1α, Gadd34, and Herp, can measured using PCR; real-time PCR is often applied nowadays because of its sensitivity and reliability compared to conventional PCR. ER stress pathway-focused PCR arrays can be used for screening and mechanism exploration (http://www. sabiosciences.com/rt_pcr_product/HTML/PAMM-089A.html). Gene expression analysis with microarray studies or next-generation sequencing can also be used for mechanism discovery.
-
Semiquantitative PCR and real-time quantitative PCR to measure the splicing of XBP1 mRNA: as depicted in Figure 1, in response to ER stress, Grp78 is released from the complex of Grp78/IRE1. IRE, an ER-located transmembrane RNase, is then activated by auto-phosphorylation of its ribonuclease domain. Activated IRE1 causes an unconventional splicing of the mRNA encoding XBP1, a transcription factor, excising a 26 nucleotide to form a spliced XBP1, referred to as XBP1S. XBP1S is more stable than XBP1, increasing its ability to act as a transcription factor that binds to the ER stress response element (ERSE) sequence of a variety of UPR target genes and leads to the transcription of many chaperone proteins [122–125]. The ratio of XBP1 to XBP1S correlates with the expression levels of other ER stress markers; thus, XBP1 mRNA splicing is often measured in ER stress studies [120,126,127]. The most commonly used method to measure XBP1 mRNA splicing is semiquantitative RT-PCR that can be visualized by gel electrophoresis. The detailed procedures, PCR conditions, and the primers used to detect both unspliced and spliced isoforms are given in Samali and colleagues [120].
Quantitative real-time RT-PCR has been suggested for measuring XBP1S mRNA instead of conventional RT-PCR [127]. The primers for real-time are designed to span the 26 base pair intron that is removed from the unspliced form of XBP1 mRNA; in this way, only spliced XBP1 (XBP1S) is amplified and measured. Using two well-known ER stress inducers, thapsigargin and tunicamycin, the correlation of the expression of CHOP and Bip to XBP1S was shown to be 0.962 and 0.884, respectively [127].
Reporter assays: transcriptional regulators ATF6, ATF4, and XBP1S are activated when ER is stressed (Figure 1). These transcription factors bind to ER stress-response elements (ERSE) or unfolded protein response elements (UPRE) on the promoters of their regulated downstream genes [128,129]. Based on the transcriptional activation of these regulators, reporter assays have been developed to detect ATF6 or XBP1S activation. A luciferase-based assay has been generally used, with the luciferase gene being under the control of promoters containing the ERSE or UPRE of the ATF6 or XBP1S gene or other ER stress-related genes [128,130]. Because blocking of or decreasing protein secretion is a distinct feature of ER stress, seeing a reduction of protein secretion indicates an ER stress response. Based on this feature, reporter assays for monitoring protein secretion have also been developed and used to study ER stress in various cells [119,131–135]. One of these, secreted alkaline phosphatase (SEAP) reporter, has been used in vitro for monitoring different biological processes [131–133]. Recently, the secreted Gaussia luciferase (Gluc) has gained popularity due to its high sensitivity [119,134,135]. Both reporter assays are capable of monitoring ER stress by measuring a decrease in Gluc or SEAP being excreted into the medium because ER stress blocks or decreases processing in the secretory pathway and trafficking [119]. Two stable cell lines were established recently in our laboratory, HepG2-Fluc-SEAP and HepG2-Fluc-Gluc to study ER stress associated liver toxicity (SC an LG, unpublished). Lentivirus vectors carrying the expression cassettes for Gluc or SEAP were used to generate stable cell lines (Figure 2); firefly luciferase (Fluc) was used as an internal control for normalizing the cell number. Both cell lines showed significantly decreased activity of SEAP or Gluc when treated with the well-known ER stress inducers brefeldin A or thapsigargin. The usefulness of these in vitro systems was demonstrated in a study on ER stress induced by sertraline, an antidepressant drug [101]. It is anticipated that such assays can be used in high-throughput settings to screen drugs for liver toxicity mediated by ER stress.
A fluorescence-based detection method has been applied to detect ER stress in liver cells [136]. Thioflavin T, a small molecule with fluorescence properties, exhibits enhanced fluorescence when it binds to protein aggregates. Thioflavin T fluorescence correlates directly with ER stress response and, thus, can be used to measure and quantify ER stress.
-
Genetically manipulated models: both cell and animal models with modified ER stress-related genes have been used in ER stress studies [65,121,137–144]. For animal models, both ER stress related gene knock-in and knock-out animals have been shown to be useful to study ER stress. For example, transgenic mice expressing the ER stress indicator XBP1S have been used to monitor physiological and pathological ER stress in vivo [121]. In this model, the stress indicator was constructed by fusing XBP1 and green fluorescent protein (GFP). When ER stress occurred, the spliced indicator mRNA is translated into an XBP1-GFP fusion protein and green fluorescence can be detected. Strong fluorescence in the kidney and pancreas was detected when a typical ER stress inducer, tunicamycin, was injected i.p. [121].
Transgenic animal models are not only used in monitoring ER stress but also to study mechanisms. For example, using IRE knock-out mice, it was shown that the inflammation occurred earlier than in IRE wild-type mice when exposed to dextran sodium sulfate to induce inflammatory bowel disease, indicating that perturbations in ER function participate in the development of colitis caused by environmental agents [144]. In a more recent study, CHOP knock-out mice were used in a mechanistic study of acetaminophen-induced liver toxicity [65]. Following administration of a toxic dose to wild-type mice, ER stress was activated; all three branches of UPR were activated, with PERK-elF2-CHOP being the most important pathway. While wild-type mice developed extensive necrosis, CHOP knock-out mice exhibited hepatocyte proliferation at the sites of necrosis and protected against acetaminophen-induced liver damage, indicating that CHOP plays a critical role in acetaminophen toxicity, and that up-regulation of CHOP compromises hepatocyte survival by reducing regeneration [65].
Figure 2.
Construction of plasmids used to establish Gluc or SEAP reporter gene assays for monitoring ER stress in HepG2 cells.
It should be stated that no single individual assay will be the most appropriate for all ER stress evaluations and mechanistic studies; thus, a set of assays should be considered and chosen to be used sequentially or in parallel, based on the requirements of a particular study. As an initial step to evaluate ER stress-mediated liver toxicity in our laboratory, we perform reporter assays using the established cell lines HepG2-Fluc-Gluc and/or HepG2-Fluc-SEAP and real-time PCR on a group of ER stress-related genes because of their sensitivity and ease of use.
CONCLUSIONS
Drug-induced liver injury is one of the leading causes of acute liver failure. Besides drugs, herbal dietary supplements and environmental pollutants are also reported to be associated with liver damage [145–149]. The causes of liver damage vary, including factors such as genetic makeup, personal lifestyle, environmental factors, and pre-existing diseases. Understanding of molecular mechanisms of liver damage is fundamental for preventing and curing liver injury caused by drugs and chemicals. Endoplasmic reticulum stress plays an important role in a range of diseases. The role of ER stress in drug-induced liver toxicity is beginning to be recognized, although not yet extensively studied and therefore the precise mechanisms remain elusive. Better knowledge of ER stress can help identify risk factors and decrease the incidence of drug-induced liver toxicity. It is anticipated that as more studies aimed at studying ER stress associated liver toxicity and the interplay between different organelles and pathways appear, the role of ER stress in liver toxicity will become widely recognized.
Acknowledgments
SC was supported by appointments to the Postgraduate Research Program at the National Center for Toxicological Research administered by the Oak Ridge Institute for Science Education through an interagency agreement between the US Department of Energy and the US FDA.
Footnotes
Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/lesc.
This article is not an official guidance or policy statement of the US FDA. No official support or endorsement by the US FDA is intended or should be inferred.
References
- 1.Lee WM. Drug-induced hepatotoxicity. N Engl J Med. 2003;349:474–485. doi: 10.1056/NEJMra021844. [DOI] [PubMed] [Google Scholar]
- 2.Kaplowitz N. Drug-induced liver disorders: implications for drug development and regulation. Drug Saf. 2001;24:483–490. doi: 10.2165/00002018-200124070-00001. [DOI] [PubMed] [Google Scholar]
- 3.Senior JR. Drug hepatotoxicity from a regulatory perspective. Clin Liver Dis. 2007;11:507–524. vi. doi: 10.1016/j.cld.2007.06.002. [DOI] [PubMed] [Google Scholar]
- 4.Gunawan BK, Kaplowitz N. Mechanisms of drug-induced liver disease. Clin Liver Dis. 2007;11:459–475. v. doi: 10.1016/j.cld.2007.06.001. [DOI] [PubMed] [Google Scholar]
- 5.Ellgaard L, Helenius A. Quality control in the endoplasmic reticulum. Nat Rev Mol Cell Biol. 2003;4:181–191. doi: 10.1038/nrm1052. [DOI] [PubMed] [Google Scholar]
- 6.Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–529. doi: 10.1038/nrm2199. [DOI] [PubMed] [Google Scholar]
- 7.Vembar SS, Brodsky JL. One step at a time: endoplasmic reticulum-associated degradation. Nat Rev Mol Cell Biol. 2008;9:944–957. doi: 10.1038/nrm2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011;334:1081–1086. doi: 10.1126/science.1209038. [DOI] [PubMed] [Google Scholar]
- 9.Wu J, Kaufman RJ. From acute ER stress to physiological roles of the Unfolded Protein Response. Cell Death Differ. 2006;13:374–384. doi: 10.1038/sj.cdd.4401840. [DOI] [PubMed] [Google Scholar]
- 10.Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest. 2005;115:2656–2664. doi: 10.1172/JCI26373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dara L, Ji C, Kaplowitz N. The contribution of endoplasmic reticulum stress to liver diseases. Hepatology. 2011;53:1752–1763. doi: 10.1002/hep.24279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hitomi J, Katayama T, Eguchi Y, Kudo T, Taniguchi M, Koyama Y, et al. Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Abeta-induced cell death. J Cell Biol. 2004;165:347–356. doi: 10.1083/jcb.200310015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Mekahli D, Bultynck G, Parys JB, De Smedt H, Missiaen L. Endoplasmic-reticulum calcium depletion and disease. Cold Spring Harb Perspect Biol. 2011:3. doi: 10.1101/cshperspect.a004317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang S, Kaufman RJ. The impact of the unfolded protein response on human disease. J Cell Biol. 2012;197:857–867. doi: 10.1083/jcb.201110131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yoshida H. ER stress and diseases. FEBS J. 2007;274:630–658. doi: 10.1111/j.1742-4658.2007.05639.x. [DOI] [PubMed] [Google Scholar]
- 16.Kepp O, Menger L, Vacchelli E, Locher C, Adjemian S, Yamazaki T, et al. Crosstalk between ER stress and immunogenic cell death. Cytokine Growth Factor Rev. 2013;24:311–318. doi: 10.1016/j.cytogfr.2013.05.001. [DOI] [PubMed] [Google Scholar]
- 17.Li Z. Glucose regulated protein 78: a critical link between tumor microenvironment and cancer hallmarks. Biochim Biophys Acta. 2012;1826:13–22. doi: 10.1016/j.bbcan.2012.02.001. [DOI] [PubMed] [Google Scholar]
- 18.Luo B, Lee AS. The critical roles of endoplasmic reticulum chaperones and unfolded protein response in tumorigenesis and anticancer therapies. Oncogene. 2013;32:805–818. doi: 10.1038/onc.2012.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Martinon F. Targeting endoplasmic reticulum signaling pathways in cancer. Acta Oncol. 2012;51:822–830. doi: 10.3109/0284186X.2012.689113. [DOI] [PubMed] [Google Scholar]
- 20.Schonthal AH. Pharmacological targeting of endoplasmic reticulum stress signaling in cancer. Biochem Pharmacol. 2013;85:653–666. doi: 10.1016/j.bcp.2012.09.012. [DOI] [PubMed] [Google Scholar]
- 21.Singleton DC, Harris AL. Targeting the ATF4 pathway in cancer therapy. Expert Opin Ther Targets. 2012;16:1189–1202. doi: 10.1517/14728222.2012.728207. [DOI] [PubMed] [Google Scholar]
- 22.Suh DH, Kim MK, Kim HS, Chung HH, Song YS. Unfolded protein response to autophagy as a promising druggable target for anticancer therapy. Ann N Y Acad Sci. 2012;1271:20–32. doi: 10.1111/j.1749-6632.2012.06739.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Healy SJ, Gorman AM, Mousavi-Shafaei P, Gupta S, Samali A. Targeting the endoplasmic reticulum-stress response as an anticancer strategy. Eur J Pharmacol. 2009;625:234–246. doi: 10.1016/j.ejphar.2009.06.064. [DOI] [PubMed] [Google Scholar]
- 24.Moenner M, Pluquet O, Bouchecareilh M, Chevet E. Integrated endoplasmic reticulum stress responses in cancer. Cancer Res. 2007;67:10631–10634. doi: 10.1158/0008-5472.CAN-07-1705. [DOI] [PubMed] [Google Scholar]
- 25.Roussel BD, Kruppa AJ, Miranda E, Crowther DC, Lomas DA, Marciniak SJ. Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol. 2013;12:105–118. doi: 10.1016/S1474-4422(12)70238-7. [DOI] [PubMed] [Google Scholar]
- 26.Cornejo VH, Hetz C. The unfolded protein response in Alzheimer’s disease. Semin Immunopathol. 2013;35:277–292. doi: 10.1007/s00281-013-0373-9. [DOI] [PubMed] [Google Scholar]
- 27.Ansari N, Khodagholi F. Molecular mechanism aspect of ER stress in Alzheimer’s disease: current approaches and future strategies. Curr Drug Targets. 2013;14:114–122. doi: 10.2174/138945013804806532. [DOI] [PubMed] [Google Scholar]
- 28.Viana RJ, Nunes AF, Rodrigues CM. Endoplasmic reticulum enrollment in Alzheimer’s disease. Mol Neurobiol. 2012;46:522–534. doi: 10.1007/s12035-012-8301-x. [DOI] [PubMed] [Google Scholar]
- 29.Kaneko M, Okuma Y, Nomura Y. Molecular approaches to the treatment, pro-phylaxis, and diagnosis of Alzheimer’s disease: possible involvement of HRD1, a novel molecule related to endoplasmic reticulum stress, in Alzheimer’s disease. J Pharmacol Sci. 2012;118:325–330. doi: 10.1254/jphs.11r11fm. [DOI] [PubMed] [Google Scholar]
- 30.Omura T, Kaneko M, Okuma Y, Matsubara K, Nomura Y. Endoplasmic reticulum stress and Parkinson’s disease: the role of HRD1 in averting apoptosis in neurodegenerative disease. Oxid Med Cell Longev. 2013;2013:239854. doi: 10.1155/2013/239854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mercado G, Valdes P, Hetz C. An ERcentric view of Parkinson’s disease. Trends Mol Med. 2013;19:165–175. doi: 10.1016/j.molmed.2012.12.005. [DOI] [PubMed] [Google Scholar]
- 32.Cybulsky AV. Endoplasmic reticulum stress in proteinuric kidney disease. Kidney Int. 2010;77:187–193. doi: 10.1038/ki.2009.389. [DOI] [PubMed] [Google Scholar]
- 33.Dickhout JG, Krepinsky JC. Endoplasmic reticulum stress and renal disease. Antioxid Redox Signal. 2009;11:2341–2352. doi: 10.1089/ars.2009.2705. [DOI] [PubMed] [Google Scholar]
- 34.Kitamura M. Endoplasmic reticulum stress and unfolded protein response in renal pathophysiology: Janus faces. Am J Physiol Renal Physiol. 2008;295:F323–334. doi: 10.1152/ajprenal.00050.2008. [DOI] [PubMed] [Google Scholar]
- 35.Wei J, Rahman S, Ayaub EA, Dickhout JG, Ask K. Protein misfolding and endoplasmic reticulum stress in chronic lung disease. Chest. 2013;143:1098–1105. doi: 10.1378/chest.12-2133. [DOI] [PubMed] [Google Scholar]
- 36.Osorio F, Lambrecht B, Janssens S. The UPR and lung disease. Semin Immunopathol. 2013;35:293–306. doi: 10.1007/s00281-013-0368-6. [DOI] [PubMed] [Google Scholar]
- 37.Ribeiro CM, O’Neal WK. Endoplasmic reticulum stress in chronic obstructive lung diseases. Curr Mol Med. 2012;12:872–882. doi: 10.2174/156652412801318791. [DOI] [PubMed] [Google Scholar]
- 38.Doroudgar S, Glembotski CC. New concepts of endoplasmic reticulum function in the heart: programmed to conserve. J Mol Cell Cardiol. 2013;55:85–91. doi: 10.1016/j.yjmcc.2012.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Groenendyk J, Agellon LB, Michalak M. Coping with endoplasmic reticulum stress in the cardiovascular system. Annu Rev Physiol. 2013;75:49–67. doi: 10.1146/annurev-physiol-030212-183707. [DOI] [PubMed] [Google Scholar]
- 40.Groenendyk J, Sreenivasaiah PK, Kimdo H, Agellon LB, Michalak M. Biology of endoplasmic reticulum stress in the heart. Circ Res. 2010;107:1185–1197. doi: 10.1161/CIRCRESAHA.110.227033. [DOI] [PubMed] [Google Scholar]
- 41.Wang WA, Groenendyk J, Michalak M. Calreticulin signaling in health and disease. Int J Biochem Cell Biol. 2012;44:842–846. doi: 10.1016/j.biocel.2012.02.009. [DOI] [PubMed] [Google Scholar]
- 42.Minamino T, Kitakaze M. ER stress in cardiovascular disease. J Mol Cell Cardiol. 2010;48:1105–1110. doi: 10.1016/j.yjmcc.2009.10.026. [DOI] [PubMed] [Google Scholar]
- 43.Claudio N, Dalet A, Gatti E, Pierre P. Mapping the crossroads of immune activation and cellular stress response pathways. EMBO J. 2013;32:1214–1224. doi: 10.1038/emboj.2013.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Adolph TE, Niederreiter L, Blumberg RS, Kaser A. Endoplasmic reticulum stress and inflammation. Dig Dis. 2012;30:341–346. doi: 10.1159/000338121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kolattukudy PE, Niu J. Inflammation, endoplasmic reticulum stress, autophagy, and the monocyte chemoattractant protein-1/CCR2 pathway. Circ Res. 2012;110:174–189. doi: 10.1161/CIRCRESAHA.111.243212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Niederreiter L, Kaser A. Endoplasmic reticulum stress and inflammatory bowel disease. Acta Gastroenterol Belg. 2011;74:330–333. [PubMed] [Google Scholar]
- 47.Kaser A, Flak MB, Tomczak MF, Blumberg RS. The unfolded protein response and its role in intestinal homeostasis and inflammation. Exp Cell Res. 2011;317:2772–2779. doi: 10.1016/j.yexcr.2011.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fu S, Watkins SM, Hotamisligil GS. The role of endoplasmic reticulum in hepatic lipid homeostasis and stress signaling. Cell Metab. 2012;15:623–634. doi: 10.1016/j.cmet.2012.03.007. [DOI] [PubMed] [Google Scholar]
- 49.Tripathi YB, Pandey V. Obesity and endoplasmic reticulum (ER) stresses. Front Immunol. 2012;3:240. doi: 10.3389/fimmu.2012.00240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cnop M, Foufelle F, Velloso LA. Endoplasmic reticulum stress, obesity and diabetes. Trends Mol Med. 2012;18:59–68. doi: 10.1016/j.molmed.2011.07.010. [DOI] [PubMed] [Google Scholar]
- 51.Kaser A, Adolph TE, Blumberg RS. The unfolded protein response and gastrointestinal disease. Semin Immunopathol. 2013;35:307–319. doi: 10.1007/s00281-013-0377-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Evans-Molina C, Hatanaka M, Mirmira RG. Lost in translation: endoplasmic reticulum stress and the decline of beta-cell health in diabetes mellitus. Diabetes Obes Metab. 2013;15(Suppl 3):159–169. doi: 10.1111/dom.12163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Su J, Zhou L, Kong X, Yang X, Xiang X, Zhang Y, Li X, Sun L. Endoplasmic reticulum is at the crossroads of autophagy, inflammation, and apoptosis signaling pathways and participates in the pathogenesis of diabetes mellitus. J Diabetes Res. 2013;2013:193461. doi: 10.1155/2013/193461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Flamment M, Hajduch E, Ferre P, Foufelle F. New insights into ER stress-induced insulin resistance. Trends Endocrinol Metab. 2012;23:381–390. doi: 10.1016/j.tem.2012.06.003. [DOI] [PubMed] [Google Scholar]
- 55.Kim MK, Kim HS, Lee IK, Park KG. Endoplasmic reticulum stress and insulin biosynthesis: a review. Exp Diabetes Res. 2012;2012:509437. doi: 10.1155/2012/509437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Basha B, Samuel SM, Triggle CR, Ding H. Endothelial dysfunction in diabetes mellitus: possible involvement of endoplasmic reticulum stress? Exp Diabetes Res. 2012;2012:481840. doi: 10.1155/2012/481840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Beriault DR, Werstuck GH. The role of glucosamine-induced ER stress in diabetic atherogenesis. Exp Diabetes Res. 2012;2012:187018. doi: 10.1155/2012/187018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhong J, Rao X, Xu JF, Yang P, Wang CY. The role of endoplasmic reticulum stress in autoimmune-mediated beta-cell destruction in type 1 diabetes. Exp Diabetes Res. 2012;2012:238980. doi: 10.1155/2012/238980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Back SH, Kaufman RJ. Endoplasmic reticulum stress and type 2 diabetes. Annu Rev Biochem. 2012;81:767–793. doi: 10.1146/annurev-biochem-072909-095555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cunard R, Sharma K. The endoplasmic reticulum stress response and diabetic kidney disease. Am J Physiol Renal Physiol. 2011;300:F1054–1061. doi: 10.1152/ajprenal.00021.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kitamura M. The unfolded protein response triggered by environmental factors. Semin Immunopathol. 2013;35:259–275. doi: 10.1007/s00281-013-0371-y. [DOI] [PubMed] [Google Scholar]
- 62.Blumental-Perry A. Unfolded protein response in chronic obstructive pulmonary disease: smoking, aging and disease: a SAD trifecta. Curr Mol Med. 2012;12:883–898. doi: 10.2174/156652412801318764. [DOI] [PubMed] [Google Scholar]
- 63.Larson AM, Polson J, Fontana RJ, Davern TJ, Lalani E, Hynan LS, et al. Acetaminophen-induced acute liver failure: results of a United States multicenter, prospective study. Hepatology. 2005;42:1364–1372. doi: 10.1002/hep.20948. [DOI] [PubMed] [Google Scholar]
- 64.James LP, Mayeux PR, Hinson JA. Acetaminophen-induced hepatotoxicity. Drug Metab Dispos. 2003;31:1499–1506. doi: 10.1124/dmd.31.12.1499. [DOI] [PubMed] [Google Scholar]
- 65.Uzi D, Barda L, Scaiewicz V, Mills M, Mueller T, Gonzalez-Rodriguez A, et al. CHOP is a critical regulator of acetaminophen-induced hepatotoxicity. J Hepatol. 2013;59:495–503. doi: 10.1016/j.jhep.2013.04.024. [DOI] [PubMed] [Google Scholar]
- 66.Wang X, Thomas B, Sachdeva R, Arterburn L, Frye L, Hatcher PG, Cornwell DG, Ma J. Mechanism of arylating quinone toxicity involving Michael adduct formation and induction of endoplasmic reticulum stress. Proc Natl Acad Sci U S A. 2006;103:3604–3609. doi: 10.1073/pnas.0510962103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Nagy G, Szarka A, Lotz G, Doczi J, Wunderlich L, Kiss A, et al. BGP-15 inhibits caspase-independent programmed cell death in acetaminophen-induced liver injury. Toxicol Appl Pharmacol. 2010;243:96–103. doi: 10.1016/j.taap.2009.11.017. [DOI] [PubMed] [Google Scholar]
- 68.Nagy G, Kardon T, Wunderlich L, Szarka A, Kiss A, Schaff Z, Banhegyi G, Mandl J. Acetaminophen induces ER dependent signaling in mouse liver. Arch Biochem Biophys. 2007;459:273–279. doi: 10.1016/j.abb.2006.11.021. [DOI] [PubMed] [Google Scholar]
- 69.Jones M, Nunez M. Liver toxicity of antiretroviral drugs. Semin Liver Dis. 2012;32:167–176. doi: 10.1055/s-0032-1316472. [DOI] [PubMed] [Google Scholar]
- 70.Tashima KT, Bausserman L, Alt EN, Aznar E, Flanigan TP. Lipid changes in patients initiating efavirenz- and indinavir-based antiretroviral regimens. HIV Clin Trials. 2003;4:29–36. doi: 10.1310/f2v7-3r46-vx6j-241r. [DOI] [PubMed] [Google Scholar]
- 71.Loko MA, Bani-Sadr F, Winnock M, Lacombe K, Carrieri P, Neau D, Morlat P, Serfaty L, Dabis F, Salmon D. Impact of HAART exposure and associated lipodys-trophy on advanced liver fibrosis in HIV/HCV-coinfected patients. J Viral Hepat. 2011;18:e307–314. doi: 10.1111/j.1365-2893.2010.01417.x. [DOI] [PubMed] [Google Scholar]
- 72.Apostolova N, Gomez-Sucerquia LJ, Alegre F, Funes HA, Victor VM, Barrachina MD, Blas-Garcia A, Esplugues JV. ER stress in human hepatic cells treated with Efavirenz: Mitochondria again. J Hepatol. 2013;59:780–789. doi: 10.1016/j.jhep.2013.06.005. [DOI] [PubMed] [Google Scholar]
- 73.Gomez-Sucerquia LJ, Blas-Garcia A, Marti-Cabrera M, Esplugues JV, Apostolova N. Profile of stress and toxicity gene expression in human hepatic cells treated with Efavirenz. Antiviral Res. 2012;94:232–241. doi: 10.1016/j.antiviral.2012.04.003. [DOI] [PubMed] [Google Scholar]
- 74.Apostolova N, Blas-Garcia A, Esplugues JV. Mitochondrial interference by anti-HIV drugs: mechanisms beyond Pol-gamma inhibition. Trends Pharmacol Sci. 2011;32:715–725. doi: 10.1016/j.tips.2011.07.007. [DOI] [PubMed] [Google Scholar]
- 75.Apostolova N, Gomez-Sucerquia LJ, Moran A, Alvarez A, Blas-Garcia A, Esplugues JV. Enhanced oxidative stress and increased mitochondrial mass during efavirenz-induced apoptosis in human hepatic cells. Br J Pharmacol. 2010;160:2069–2084. doi: 10.1111/j.1476-5381.2010.00866.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Apostolova N, Gomez-Sucerquia LJ, Gortat A, Blas-Garcia A, Esplugues JV. Compromising mitochondrial function with the antiretroviral drug efavirenz induces cell survival-promoting autophagy. Hepatology. 2011;54:1009–1019. doi: 10.1002/hep.24459. [DOI] [PubMed] [Google Scholar]
- 77.Kao E, Shinohara M, Feng M, Lau MY, Ji C. Human immunodeficiency virus pro-tease inhibitors modulate Ca2+ homeostasis and potentiate alcoholic stress and injury in mice and primary mouse and human hepatocytes. Hepatology. 2012;56:594–604. doi: 10.1002/hep.25702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Parker RA, Flint OP, Mulvey R, Elosua C, Wang F, Fenderson W, Wang S, Yang WP, Noor MA. Endoplasmic reticulum stress links dyslipidemia to inhibition of pro-teasome activity and glucose transport by HIV protease inhibitors. Mol Pharmacol. 2005;67:1909–1919. doi: 10.1124/mol.104.010165. [DOI] [PubMed] [Google Scholar]
- 79.Zhou H, Gurley EC, Jarujaron S, Ding H, Fang Y, Xu Z, Pandak WM, Jr, Hylemon PB. HIV protease inhibitors activate the unfolded protein response and disrupt lipid metabolism in primary hepatocytes. Am J Physiol Gastrointest Liver Physiol. 2006;291:G1071–1080. doi: 10.1152/ajpgi.00182.2006. [DOI] [PubMed] [Google Scholar]
- 80.Kohlroser J, Mathai J, Reichheld J, Banner BF, Bonkovsky HL. Hepatotoxicity due to troglitazone: report of two cases and review of adverse events reported to the United States Food and Drug Administration. Am J Gastroenterol. 2000;95:272–276. doi: 10.1111/j.1572-0241.2000.01707.x. [DOI] [PubMed] [Google Scholar]
- 81.Isley WL. Hepatotoxicity of thiazolidinediones. Expert Opin Drug Saf. 2003;2:581–586. doi: 10.1517/14740338.2.6.581. [DOI] [PubMed] [Google Scholar]
- 82.Kakuni M, Morita M, Matsuo K, Katoh Y, Nakajima M, Tateno C, Yokoi T. Chimeric mice with a humanized liver as an animal model of troglitazone-induced liver injury. Toxicol Lett. 2012;214:9–18. doi: 10.1016/j.toxlet.2012.08.001. [DOI] [PubMed] [Google Scholar]
- 83.Okuda T, Norioka M, Shitara Y, Horie T. Multiple mechanisms underlying troglitazone-induced mitochondrial permeability transition. Toxicol Appl Pharmacol. 2010;248:242–248. doi: 10.1016/j.taap.2010.08.007. [DOI] [PubMed] [Google Scholar]
- 84.Fujimoto K, Kumagai K, Ito K, Arakawa S, Ando Y, Oda S, Yamoto T, Manabe S. Sensitivity of liver injury in heterozygous Sod2 knockout mice treated with troglitazone or acetaminophen. Toxicol Pathol. 2009;37:193–200. doi: 10.1177/0192623308329282. [DOI] [PubMed] [Google Scholar]
- 85.Ong MM, Latchoumycandane C, Boelsterli UA. Troglitazone-induced hepatic necrosis in an animal model of silent genetic mitochondrial abnormalities. Toxicol Sci. 2007;97:205–213. doi: 10.1093/toxsci/kfl180. [DOI] [PubMed] [Google Scholar]
- 86.Nadanaciva S, Dykens JA, Bernal A, Capaldi RA, Will Y. Mitochondrial impairment by PPAR agonists and statins identified via immunocaptured OX-PHOS complex activities and respiration. Toxicol Appl Pharmacol. 2007;223:277–287. doi: 10.1016/j.taap.2007.06.003. [DOI] [PubMed] [Google Scholar]
- 87.Guo L, Zhang L, Sun Y, Muskhelishvili L, Blann E, Dial S, Shi L, Schroth G, Dragan YP. Differences in hepatotoxicity and gene expression profiles by anti-diabetic PPAR gamma agonists on rat primary hepatocytes and human HepG2 cells. Mol Divers. 2006;10:349–360. doi: 10.1007/s11030-006-9038-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Gardner OS, Shiau CW, Chen CS, Graves LM. Peroxisome proliferator-activated receptor gamma-independent activation of p38 MAPK by thiazolidinediones involves calcium/calmodulin-dependent protein kinase II and protein kinase R: correlation with endoplasmic reticulum stress. J Biol Chem. 2005;280:10109–10118. doi: 10.1074/jbc.M410445200. [DOI] [PubMed] [Google Scholar]
- 89.Bova MP, Tam D, McMahon G, Mattson MN. Troglitazone induces a rapid drop of mitochondrial membrane potential in liver HepG2 cells. Toxicol Lett. 2005;155:41–50. doi: 10.1016/j.toxlet.2004.08.009. [DOI] [PubMed] [Google Scholar]
- 90.Gardner OS, Dewar BJ, Earp HS, Samet JM, Graves LM. Dependence of per-oxisome proliferator-activated receptor ligand-induced mitogen-activated protein kinase signaling on epidermal growth factor receptor transactivation. J Biol Chem. 2003;278:46261–46269. doi: 10.1074/jbc.M307827200. [DOI] [PubMed] [Google Scholar]
- 91.Collados V, Hallal H, Andrade RJ. Sertraline hepatotoxicity: report of a case and review of the literature. Dig Dis Sci. 2010;55:1806–1807. doi: 10.1007/s10620-010-1192-7. [DOI] [PubMed] [Google Scholar]
- 92.Fartoux-Heymann L, Hezode C, Zafrani ES, Dhumeaux D, Mallat A. Acute fatal hepatitis related to sertraline. J Hepatol. 2001;35:683–684. doi: 10.1016/s0168-8278(01)00159-3. [DOI] [PubMed] [Google Scholar]
- 93.Hautekeete ML, Colle I, van Vlierberghe H, Elewaut A. Symptomatic liver injury probably related to sertraline. Gastroenterol Clin Biol. 1998;22:364–365. [PubMed] [Google Scholar]
- 94.Kim KY, Hwang W, Narendran R. Acute liver damage possibly related to sertra-line and venlafaxine ingestion. Ann Pharmacother. 1999;33:381–382. doi: 10.1345/aph.18155. [DOI] [PubMed] [Google Scholar]
- 95.Persky S, Reinus JF. Sertraline hepatotoxicity: a case report and review of the literature on selective serotonin reuptake inhibitor hepatotoxicity. Dig Dis Sci. 2003;48:939–944. doi: 10.1023/a:1023007831047. [DOI] [PubMed] [Google Scholar]
- 96.Tabak F, Gunduz F, Tahan V, Tabak O, Ozaras R. Sertraline hepatotoxicity: report of a case and review of the literature. Dig Dis Sci. 2009;54:1589–1591. doi: 10.1007/s10620-008-0524-3. [DOI] [PubMed] [Google Scholar]
- 97.Verrico MM, Nace DA, Towers AL. Fulminant chemical hepatitis possibly associated with donepezil and sertraline therapy. J Am Geriatr Soc. 2000;48:1659–1663. doi: 10.1111/j.1532-5415.2000.tb03879.x. [DOI] [PubMed] [Google Scholar]
- 98.Galan Navarro JL. Acute cholestatic hepatitis probably caused by sertraline. Rev Esp Enferm Dig. 2001;93:822. [PubMed] [Google Scholar]
- 99.Carvajal Garcia-Pando A, Garcia del Pozo J, Sanchez AS, Velasco MA, Rueda de Castro AM, Lucena MI. Hepatotoxicity associated with the new antidepressants. J Clin Psychiatry. 2002;63:135–137. doi: 10.4088/jcp.v63n0208. [DOI] [PubMed] [Google Scholar]
- 100.Li Y, Couch L, Higuchi M, Fang JL, Guo L. Mitochondrial dysfunction induced by sertraline, an antidepressant agent. Toxicol Sci. 2012;127:582–591. doi: 10.1093/toxsci/kfs100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chen S, Xuan J, Wan L, Lin H, Couch L, Mei N, Dobrovolsky VN, Guo L. Sertraline, an antidepressant, induces apoptosis in hepatic cells through the mitogen-activated protein kinase pathway. Toxicol Sci. doi: 10.1093/toxsci/kft254. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Orman ES, Odena G, Bataller R. Alcoholic liver disease: pathogenesis, management, and novel targets for therapy. J Gastroenterol Hepatol. 2013;28(Suppl 1):77–84. doi: 10.1111/jgh.12030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Mandrekar P, Szabo G. Signalling pathways in alcohol-induced liver inflammation. J Hepatol. 2009;50:1258–1266. doi: 10.1016/j.jhep.2009.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Magne L, Blanc E, Legrand B, Lucas D, Barouki R, Rouach H, Garlatti M. ATF4 and the integrated stress response are induced by ethanol and cytochrome P450 2E1 in human hepatocytes. J Hepatol. 2011;54:729–737. doi: 10.1016/j.jhep.2010.07.023. [DOI] [PubMed] [Google Scholar]
- 105.Ji C, Kaplowitz N. Betaine decreases hyperhomocysteinemia, endoplasmic reticulum stress, and liver injury in alcohol-fed mice. Gastroenterology. 2003;124:1488–1499. doi: 10.1016/s0016-5085(03)00276-2. [DOI] [PubMed] [Google Scholar]
- 106.Ji C, Mehrian-Shai R, Chan C, Hsu YH, Kaplowitz N. Role of CHOP in hepatic apoptosis in the murine model of intragastric ethanol feeding. Alcohol Clin Exp Res. 2005;29:1496–1503. doi: 10.1097/01.alc.0000174691.03751.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Esfandiari F, Villanueva JA, Wong DH, French SW, Halsted CH. Chronic ethanol feeding and folate deficiency activate hepatic endoplasmic reticulum stress pathway in micropigs. Am J Physiol Gastrointest Liver Physiol. 2005;289:G54–63. doi: 10.1152/ajpgi.00542.2004. [DOI] [PubMed] [Google Scholar]
- 108.Minko IG, Kozekov ID, Harris TM, Rizzo CJ, Lloyd RS, Stone MP. Chemistry and biology of DNA containing 1,N(2)-deoxyguanosine adducts of the alpha,beta-unsaturated aldehydes acrolein, crotonaldehyde, and 4-hydroxynonenal. Chem Res Toxicol. 2009;22:759–778. doi: 10.1021/tx9000489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.LoPachin RM, Gavin T. Molecular mechanism of acrylamide neurotoxicity: lessons learned from organic chemistry. Environ Health Perspect. 2012;120:1650–1657. doi: 10.1289/ehp.1205432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.LoPachin RM, Barber DS, Gavin T. Molecular mechanisms of the conjugated alpha,beta-unsaturated carbonyl derivatives: relevance to neurotoxicity and neurode-generative diseases. Toxicol Sci. 2008;104:235–249. doi: 10.1093/toxsci/kfm301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kehrer JP, Biswal SS. The molecular effects of acrolein. Toxicol Sci. 2000;57:6–15. doi: 10.1093/toxsci/57.1.6. [DOI] [PubMed] [Google Scholar]
- 112.Mohammad MK, Avila D, Zhang J, Barve S, Arteel G, McClain C, Joshi-Barve S. Acrolein cytotoxicity in hepatocytes involves endoplasmic reticulum stress, mitochondrial dysfunction and oxidative stress. Toxicol Appl Pharmacol. 2012;265:73–82. doi: 10.1016/j.taap.2012.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.ATSDR. Toxicological Profile for Arsenic. US Agency for Toxic Substances and Diseases Registry; 2005. [PubMed] [Google Scholar]
- 114.National Research Council. Arsenic in Drinking Water. National Academies Press (US); 2001. [Google Scholar]
- 115.Singh AP, Goel RK, Kaur T. Mechanisms pertaining to arsenic toxicity. Toxicol Int. 2011;18:87–93. doi: 10.4103/0971-6580.84258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Watanabe T, Hirano S. Metabolism of arsenic and its toxicological relevance. Arch Toxicol. 2013;87:969–979. doi: 10.1007/s00204-012-0904-5. [DOI] [PubMed] [Google Scholar]
- 117.Argos M, Ahsan H, Graziano JH. Arsenic and human health: epidemiologic progress and public health implications. Rev Environ Health. 2012;27:191–195. doi: 10.1515/reveh-2012-0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Naranmandura H, Xu S, Koike S, Pan LQ, Chen B, Wang YW, Rehman K, Wu B, Chen Z, Suzuki N. The endoplasmic reticulum is a target organelle for trivalent dimethylarsinic acid (DMAIII)-induced cytotoxicity. Toxicol Appl Pharmacol. 2012;260:241–249. doi: 10.1016/j.taap.2012.02.017. [DOI] [PubMed] [Google Scholar]
- 119.Badr CE, Hewett JW, Breakefield XO, Tannous BA. A highly sensitive assay for monitoring the secretory pathway and ER stress. PLoS One. 2007;2:e571. doi: 10.1371/journal.pone.0000571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Samali A, Fitzgerald U, Deegan S, Gupta S. Methods for monitoring endoplasmic reticulum stress and the unfolded protein response. Int J Cell Biol. 2010;2010:830307. doi: 10.1155/2010/830307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Iwawaki T, Akai R, Kohno K, Miura M. A transgenic mouse model for monitoring endoplasmic reticulum stress. Nat Med. 2004;10:98–102. doi: 10.1038/nm970. [DOI] [PubMed] [Google Scholar]
- 122.Cox JS, Shamu CE, Walter P. Transcriptional induction of genes encoding endoplasmic reticulum resident proteins requires a transmembrane protein kinase. Cell. 1993;73:1197–1206. doi: 10.1016/0092-8674(93)90648-a. [DOI] [PubMed] [Google Scholar]
- 123.Shamu CE, Walter P. Oligomerization and phosphorylation of the Ire1p kinase during intracellular signaling from the endoplasmic reticulum to the nucleus. EMBO J. 1996;15:3028–3039. [PMC free article] [PubMed] [Google Scholar]
- 124.Cox JS, Walter P. A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response. Cell. 1996;87:391–404. doi: 10.1016/s0092-8674(00)81360-4. [DOI] [PubMed] [Google Scholar]
- 125.Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 2002;415:92–96. doi: 10.1038/415092a. [DOI] [PubMed] [Google Scholar]
- 126.Hirota M, Kitagaki M, Itagaki H, Aiba S. Quantitative measurement of spliced XBP1 mRNA as an indicator of endoplasmic reticulum stress. J Toxicol Sci. 2006;31:149–156. doi: 10.2131/jts.31.149. [DOI] [PubMed] [Google Scholar]
- 127.van Schadewijk A, van’t Wout EF, Stolk J, Hiemstra PS. A quantitative method for detection of spliced X-box binding protein-1 (XBP1) mRNA as a measure of endoplasmic reticulum (ER) stress. Cell Stress Chaperones. 2012;17:275–279. doi: 10.1007/s12192-011-0306-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Kokame K, Kato H, Miyata T. Identification of ERSE-II, a new cis-acting element responsible for the ATF6-dependent mammalian unfolded protein response. J Biol Chem. 2001;276:9199–9205. doi: 10.1074/jbc.M010486200. [DOI] [PubMed] [Google Scholar]
- 129.Yamamoto K, Yoshida H, Kokame K, Kaufman RJ, Mori K. Differential contributions of ATF6 and XBP1 to the activation of endoplasmic reticulum stress-responsive cis-acting elements ERSE, UPRE and ERSE-II. J Biochem. 2004;136:343–350. doi: 10.1093/jb/mvh122. [DOI] [PubMed] [Google Scholar]
- 130.Wang Y, Shen J, Arenzana N, Tirasophon W, Kaufman RJ, Prywes R. Activation of ATF6 and an ATF6 DNA binding site by the endoplasmic reticulum stress response. J Biol Chem. 2000;275:27013–27020. doi: 10.1074/jbc.M003322200. [DOI] [PubMed] [Google Scholar]
- 131.Berger J, Hauber J, Hauber R, Geiger R, Cullen BR. Secreted placental alkaline phosphatase: a powerful new quantitative indicator of gene expression in eukaryotic cells. Gene. 1988;66:1–10. doi: 10.1016/0378-1119(88)90219-3. [DOI] [PubMed] [Google Scholar]
- 132.Cullen BR, Malim MH. Secreted placental alkaline phosphatase as a eukaryotic reporter gene. Methods Enzymol. 1992;216:362–368. doi: 10.1016/0076-6879(92)16033-g. [DOI] [PubMed] [Google Scholar]
- 133.Meng Y, Kasai A, Hiramatsu N, Hayakawa K, Takeda M, Shimizu F, Kawachi H, Yao J, Kitamura M. Real-time monitoring of mesangial cell-macrophage cross-talk using SEAP in vitro and ex vivo. Kidney Int. 2005;68:886–893. doi: 10.1111/j.1523-1755.2005.00471.x. [DOI] [PubMed] [Google Scholar]
- 134.Tannous BA. Gaussia luciferase reporter assay for monitoring biological processes in culture and in vivo. Nat Protoc. 2009;4:582–591. doi: 10.1038/nprot.2009.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wurdinger T, Badr C, Pike L, de Kleine R, Weissleder R, Breakefield XO, Tannous BA. A secreted luciferase for ex vivo monitoring of in vivo processes. Nat Methods. 2008;5:171–173. doi: 10.1038/nmeth.1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Beriault DR, Werstuck GH. Detection and quantification of endoplasmic reticulum stress in living cells using the fluorescent compound, Thioflavin T. Biochim Biophys Acta. 2013;1833:2293–2301. doi: 10.1016/j.bbamcr.2013.05.020. [DOI] [PubMed] [Google Scholar]
- 137.Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 2003;11:619–633. doi: 10.1016/s1097-2765(03)00105-9. [DOI] [PubMed] [Google Scholar]
- 138.Mehta SK, Cogan J, Reimold SC, De Lemos JA. Primary systemic amyloidosis presenting with advanced heart failure. Cardiol Rev. 2003;11:152–155. doi: 10.1097/01.crd.0000048198.06899.e0. [DOI] [PubMed] [Google Scholar]
- 139.Novoa I, Zhang Y, Zeng H, Jungreis R, Harding HP, Ron D. Stress-induced gene expression requires programmed recovery from translational repression. EMBO J. 2003;22:1180–1187. doi: 10.1093/emboj/cdg112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Reimold AM, Etkin A, Clauss I, Perkins A, Friend DS, Zhang J, et al. An essential role in liver development for transcription factor XBP-1. Genes Dev. 2000;14:152–157. [PMC free article] [PubMed] [Google Scholar]
- 141.Reimold AM, Iwakoshi NN, Manis J, Vallabhajosyula P, Szomolanyi-Tsuda E, Gravallese EM, Friend D, Grusby MJ, Alt F, Glimcher LH. Plasma cell differentiation requires the transcription factor XBP-1. Nature. 2001;412:300–307. doi: 10.1038/35085509. [DOI] [PubMed] [Google Scholar]
- 142.Wu J, Rutkowski DT, Dubois M, Swathirajan J, Saunders T, Wang J, Song B, Yau GD, Kaufman RJ. ATF6alpha optimizes long-term endoplasmic reticulum function to protect cells from chronic stress. Dev Cell. 2007;13:351–364. doi: 10.1016/j.devcel.2007.07.005. [DOI] [PubMed] [Google Scholar]
- 143.Harding HP, Zeng H, Zhang Y, Jungries R, Chung P, Plesken H, Sabatini DD, Ron D. Diabetes mellitus and exocrine pancreatic dysfunction in perk−/− mice reveals a role for translational control in secretory cell survival. Mol Cell. 2001;7:1153–1163. doi: 10.1016/s1097-2765(01)00264-7. [DOI] [PubMed] [Google Scholar]
- 144.Bertolotti A, Wang X, Novoa I, Jungreis R, Schlessinger K, Cho JH, West AB, Ron D. Increased sensitivity to dextran sodium sulfate colitis in IRE1beta-deficient mice. J Clin Invest. 2001;107:585–593. doi: 10.1172/JCI11476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Chen S, Wan L, Couch L, Lin H, Li Y, Dobrovolsky VN, Mei N, Guo L. Mechanism study of goldenseal-associated DNA damage. Toxicol Lett. 2013;221:64–72. doi: 10.1016/j.toxlet.2013.05.641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Guo L, Mei N, Xia Q, Chen T, Chan PC, Fu PP. Gene expression profiling as an initial approach for mechanistic studies of toxicity and tumorigenicity of herbal plants and herbal dietary supplements. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2010;28:60–87. doi: 10.1080/10590500903585416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Guo L, Li Q, Xia Q, Dial S, Chan PC, Fu P. Analysis of gene expression changes of drug metabolizing enzymes in the livers of F344 rats following oral treatment with kava extract. Food Chem Toxicol. 2009;47:433–442. doi: 10.1016/j.fct.2008.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Guo L, Shi Q, Fang JL, Mei N, Ali AA, Lewis SM, Leakey JE, Frankos VH. Review of usnic acid and Usnea barbata toxicity. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2008;26:317–338. doi: 10.1080/10590500802533392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Fu PP, Xia Q, Guo L, Yu H, Chan PC. Toxicity of kava kava. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev. 2008;26:89–112. doi: 10.1080/10590500801907407. [DOI] [PMC free article] [PubMed] [Google Scholar]


