Skip to main content
Oxidative Medicine and Cellular Longevity logoLink to Oxidative Medicine and Cellular Longevity
. 2016 Dec 22;2016:3453926. doi: 10.1155/2016/3453926

Naturally Occurring Nrf2 Activators: Potential in Treatment of Liver Injury

Ravirajsinh N Jadeja 1, Kapil K Upadhyay 2, Ranjitsinh V Devkar 2, Sandeep Khurana 1,*
PMCID: PMC5215260  PMID: 28101296

Abstract

Oxidative stress plays a major role in acute and chronic liver injury. In hepatocytes, oxidative stress frequently triggers antioxidant response by activating nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor, which upregulates various cytoprotective genes. Thus, Nrf2 is considered a potential therapeutic target to halt liver injury. Several studies indicate that activation of Nrf2 signaling pathway ameliorates liver injury. The hepatoprotective potential of naturally occurring compounds has been investigated in various models of liver injuries. In this review, we comprehensively appraise various phytochemicals that have been assessed for their potential to halt acute and chronic liver injury by enhancing the activation of Nrf2 and have the potential for use in humans.

1. Introduction

Liver has an extraordinary capacity to detoxify compounds that have potential to induce liver injury. As a consequence, the liver is also vulnerable to injury. Although liver injury is a major cause of morbidity and mortality, medical therapies to prevent hepatocyte loss or protect hepatocytes are limited. One example is N-acetylcysteine (NAC), which is used for the treatment of acetaminophen- (APAP-) induced liver injury to promote recovery and reduce the need for liver transplantation. Hence, it is imperative to identify better medical therapies that are hepatoprotective and have minimal side effects.

Naturally occurring compounds have long been used as potential hepatoprotective agents. Drafts such as Ayurveda and traditional Japanese, Chinese, and Kampo (traditional Chinese medicine but adapted to the Japanese culture) medicine recommended the use of formulations of specific plants and fruits in the treatment of liver diseases [14]. In recent years, technological advances led to the isolation of active phytochemicals, which are now available as potential therapeutic agents [5, 6].

Oxidative stress is a major factor in the mechanism underlying liver diseases. It contributes to the initiation as well as progression of liver injury [7]. Factors such as alcohol, drugs, heavy metals, and high-fat diet are now identified as inducers of hepatic oxidative stress [8]. In liver injury, hepatocytes, the key parenchymal cells, suffer oxidative stress the most. In response to oxidative stress, Kupffer cells produce a variety of cytokines which contribute to hepatocyte apoptosis [9]. Oxidative stress also induces proliferation of stellate cells and collagen synthesis, thus promoting fibrosis and cirrhosis. In response to overwhelming oxidative stress, there is a significant use of antioxidant proteins, along with an increase in lipid peroxidation. However, to maintain redox homeostasis, hepatocytes have a sophisticated antioxidant system comprising antioxidant proteins, enzymes, and transcription factors to combat oxidative stress. Hence, regulation of hepatic oxidative stress can play a critical role in the treatment of various liver diseases.

Nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor of the cap'n'collar basic leucine zipper family [10], is a key regulator of oxidative stress in numerous cell types including hepatocytes [1116]. Nrf2 is primarily regulated by Kelch-like ECH-associated protein 1 (Keap1), a substrate adaptor for a cul3-containing E3 ubiquitin ligase [17]. In the absence of oxidative stress, Nrf2 is located in the cytoplasm where it interacts with Keap1 and is rapidly degraded by the ubiquitin-proteasome pathway [18, 19]. However, under oxidative stress, phosphorylation of Nrf2 leads to its dissociation from Keap1 and subsequent translocation to the nucleus [14, 15, 19]. Herein, it binds to antioxidant response element (ARE) sequences and, in partnership with other nuclear proteins, enhances the transcription of ARE-responsive genes such as hemeoxygenase-1 (HO-1), NAD(P)H:quinone oxidoreductase 1 (NQO1), glutathione-S-transferases (GST), glutamate-cysteine ligase modifier subunit (GCLM), glutathione peroxidase (GPX), and glutamate-cysteine ligase catalytic subunit (GCLC) to mount strong antioxidant and cytoprotective responses [20, 21].

Numerous studies have shown that natural products regulate oxidative stress in the liver by modulating Nrf2-ARE pathway to render hepatoprotective effect. This review discusses the importance of Nrf2-ARE in regulating liver injury and the role of natural product based activators (phytochemicals) of Nrf2-ARE pathway in treating liver injury (Table 1).

Table 1.

Various Nrf2 activator phytochemicals and their role in liver injury.

Phytochemicals Effective doses Experimental procedure (injury model) Outcomes References
Acetaminophen toxicity
Ginsenoside Rg3 3 μg/mL Rat hepatocytes treated with 200 μM NAPQI Repletion of GSH content and enhanced expression of Mrp expression [39]
Oleanolic acid 90 mg/kg Mice injected with 330 μmol/kg APAP into the right femoral vein Enhanced antioxidant response to reduce hepatocyte necrosis [37]
Salvianolic acid B 25 and 50 mg/kg Mice treated with single dose of 300 mg/kg APAP (i.g.) Antioxidant response and phase II enzyme induction via activation of PI3K/Akt and PKC signaling to reduce liver injury [35]
Sauchinone 30 mg/kg Mice treated with single dose of 500 mg/kg APAP (i.p.) Induction of antioxidant genes to reduce hepatocyte necrosis [34]
Oleanolic acid 5 mg/kg Mice treated with single dose of 300 mg/kg APAP (i.p.) Reduction of ROS generation, GSH depletion, and lipid peroxidation coupled with upregulation of antioxidant genes [36]
Withaferin A 40 mg/kg Mice treated with single dose of 250 mg/kg APAP (i.p.) Reduced hepatocyte injury by reducing GSH depletion [38]

Inflammatory injury
Ellagic acid 5, 10, and 20 mg/kg Mice treated with single dose of 800 mg/kg Gal + 50 μg/kg LPS (i.p.) Reduced LPS/GalN-induced NF-κB activation and increased antioxidant genes [43]
Linalool 10, 20, and 40 mg/kg Mice treated with single dose of 800 mg/kg Gal + 50 μg/kg LPS (i.p.) Reduced LPS/GalN-induced NF-κB activation and induction of cytoprotective genes [44]
Mangiferin 5, 10, and 20 mg/kg Mice treated with single dose of 800 mg/kg Gal + 50 μg/kg LPS (i.p.) Reduced liver injury by activating antioxidant pathway and inhibiting NLRP3 inflammasome activation [45]
Oroxylin A 15, 30, and 60 mg/kg Mice treated with single dose of 800 mg/kg Gal + 50 μg/kg LPS (i.p.) Decreased liver injury by activating antioxidant genes and inhibiting TLR4 signaling-mediated inflammation [46]

Chemical toxicity
Tungtungmadic acid 5 and 20 μM Hepa1c1c7 cells treated with 250 μM t-BHP HO-1 induction via the PI3K/Akt signaling pathway to reduce hepatocyte death [47]
Antcin C 20 μM in cells, 100 mg/kg in mice HepG2 cells treated with 10 mM AAPH, mice treated with single dose of 80 mg/kg AAPH (i.p.) Induction of antioxidant response via increase of JNK1/2 and PI3K/Akt activities [48]
Butein and phloretin 25 μM in vitro, 30 mg/kg in vivo Mouse hepatocytes treated with 0.5 mM t-BHP, rats treated with single dose of 1 mL/kg CCl4 (i.p.) Upregulation of HO-1 and GCLC expression through ERK2 pathway [49]
Carthamus red 10 and 20 mg/kg Mice treated with two doses of 2 mL/kg CCl4-olive oil mixture (1 : 1) Upregulation of Nrf2, GSTα, and NQO1 expressions associated with decreased hepatocyte injury and ALT levels [50]
Curcumin 200 mg/kg Mice treated with single dose of 20 mg/kg DEN (i.p.) Nrf2-mediated HO-1 induction and amelioration of hepatocyte injury [51]
Diallyl disulfide 50 and 100 mg/kg Rats treated with single dose of 2 mL/kg CCl4 (i.g.) Induction of antioxidant and detoxifying enzyme activities and suppressing of inflammatory cytokines production by reducing NF-κB activation [52, 53]
Ginsenoside Rg1 20 and 40 mg/kg Rats treated with 2 mL/kg of 50% CCl4 (s.c.) twice a week for 8 weeks Reduced liver fibrosis by augmented antioxidant systems [54]
Glycyrrhetinic acid 25 and 50 mg/kg Mice treated with 6.4 g/kg CCl4 (s.c.) for 30 days Enhanced antioxidant genes expression to reduce hepatocyte injury [55]
Hesperidin 40 and 80 μM LO-2 cells treated with 150 μM t-BHP ERK-mediated nuclear translocation of Nrf2 to induce HO-1 gene expression and antioxidant response [56]
Isoorientin 5 μg/mL HepG2 cells treated with 200 μM t-BHP Upregulation of antioxidant enzyme expression through PI3K/Akt pathway [57]
Naringenin 50 mg/kg Rats treated with 2 mL/kg CCl4-olive oil mixture (1 : 1) on days 2 and 5 (i.p.) Increase in Nrf2 and HO-1 expression to reduce liver injury [58]
Oxyresveratrol 10 μM for in vitro study, 10 and 30 mg/kg for in vivo study 200 μM t-BHP treatment to HepG2 cells, ice treated with single dose of 0.5 mL/kg CCl4 (i.p.) ERK phosphorylation-mediated induction of antioxidant pathway to protect hepatocytes against oxidative stress, mitochondrial damage, and resultant cell death [59]
Puerarin 100 μM 500 μM t-BHP treatment to Hepa1c1c7 and HepG2 cells Augmentation of cellular antioxidant defenses through Nrf2-dependent HO-1 induction via PI3K pathway [60]
Resveratrol 50 and 75 μM Primary rat hepatocytes treated with 500 μM t-BHP Reduced hepatocyte death by improving antioxidant status [61]
Schisandrin B 15 μM AML12 cells treated with 20 μM menadione for 1 h Induction of ERK/Nrf2 signaling to enhance glutathione-mediated antioxidant response to protect hepatocytes against menadione-induced apoptosis [62]

Metal toxicity
Curcumin 200 mg/kg Exposure of mice to NaAsO2 (100 mg/L) in drinking water Induction of antioxidant genes and enhanced methylation and elimination of arsenic [63]
Lutein 40 mg/kg Mice treated with 4 mg/kg As2O3 (i.g.) Reduced liver injury by induction of antioxidant response [64]
S-Allylcysteine 100 mg/kg Mice treated with single dose of 17 mg/kg K2Cr2O7 (s.c.) Induction of antioxidant response to reduce liver injury [65]

Alcohol toxicity
Lucidone 1, 5, and 10 μg/mL HepG2 cells treated with 100 mM ethanol Induction of HO-1 via Nrf2 signaling pathway to enhance antioxidant response [66]
Quercetin 100 μM Primary human hepatocytes treated with 100 mM ethanol ERK- and p38-mediated Nrf2 nuclear translocation and subsequent induction of HO-1 activity [67, 68]
Quercetin 50 μM LO-2 cells treated with 100 mM ethanol Preventing hepatotoxicity by inducing p62 expression and induction of antioxidant response [69]
Sulforaphane 50 mg/kg Mice treated with 3 g/kg ethanol (30%) for 5 days (i.g.) Decreased hepatocyte lipid accumulation and injury without altering CYP2E1 expression [70]

Nonalcoholic steatohepatitis
Baicalein 10 mg/kg Rats fed with MCD diet for 8 weeks Reduction in inflammation and oxidative hepatocyte injury [71]
Curcumin 50 mg/kg Rats fed with HFD for 6 weeks Reduced hepatocyte lipid accumulation and improved insulin resistance and anti-inflammatory and antioxidant effects [72]
Gastrodin 10, 20, and 50 mg/kg HL-7702 cells treated with 0.6 mM of OA for 24 h, mice fed with HFD for 10 weeks AMPK-mediated induction of Nrf2 pathway to enhance expression of antioxidant enzymes [73]
Lycopene 15 mg/kg Mice fed with HFD for the next 6 weeks following a single dose of 30 mg/kg DEN injection Reduction in hepatocyte injury by induction of antioxidant pathway along with a decrease in CY2E1 expression [74]

Cholestatic liver injury
Oleanolic acid 20 mg/kg Mice treated with 125 mg/kg LCA (i.p.) Upregulation of Mrp2, Mrp3, and Mrp4 to reduce cholestatic liver injury [75]
Oleanolic acid 20 mg/kg Bile duct ligation in mice Induction of Mrps and FXR antagonism to reduce cholestatic liver injury [76]
Paeoniflorin 200 mg/kg Rats treated with 50 mg/kg ANIT for 4 days (i.g.) Enhanced GSH synthesis by activating Nrf2 through PI3K/Akt-dependent pathway [8]
Sulforaphane 50 mg/kg Mice treated with 3 g/kg ethanol (30%) for 5 days (i.g.) Decreased hepatocyte lipid accumulation and injury without altering CYP2E1 expression [70]
Sulforaphane 25 mg/kg Bile duct ligation in mice Antifibrotic response by inhibition of TGF-β/Smad signaling pathway [77]

AAPH: 2,2′-azobis(2-amidinopropane) dihydrochloride; MCD: methionine and choline deficient; CCl4: carbon tetrachloride; DEN: dimethylnitrosamine; HFD: high-fat diet; Gal: galactosamine; LPS: lipopolysaccharide; OA: oleic acid; NAPQI: N-acetylbenzoquinoneimine; i.p.: intraperitoneal; s.c.: subcutaneous; i.g.: intragastric; t-BHP: tert-butyl hydroperoxide; APAP: acetaminophen; LCA: lithocholic acid; ANIT: alpha-naphthylisothiocyanate; Nrf2: nuclear factor (erythroid-derived 2)-like 2; JNK1/2: c-Jun N-terminal kinases 1/2; PI3K/AKT: phosphoinositide 3-kinase/protein kinase B; HO-1: heme oxygenase-1; GCLC: glutamate-cysteine ligase catalytic subunit; ALT: alanine transaminase; GST: glutathione S-transferase; NQO1: NAD(P)H quinone dehydrogenase 1; AMPK: 5′ AMP-activated protein kinase; GSH: glutathione; CYP2E1: cytochrome P450 2E1; NLRP3: NLR family pyrin domain containing 3; Mrp: multidrug resistance-associated protein; TLR4: Toll-like receptor 4; Keap1: Kelch-like ECH-associated protein 1.

2. Nrf2 Signaling in Acetaminophen-Induced Hepatotoxicity

APAP is one of the most widely used over-the-counter analgesics. APAP is safe when taken at therapeutic doses but causes severe liver injury when ingested in higher-than-recommended doses. Acute liver failure due to APAP overdose is associated with high mortality [22]. In the United States, the incidence of APAP overdose is over 100,000 cases each year [23]. When ingested in therapeutic doses, APAP is mainly metabolized by sulfation and glucuronidation, leaving only a small fraction to be metabolized by cytochrome p4502E1 (CYP2E1) [24]. However, upon overdose, glucuronidation and sulfation pathways get saturated leading to APAP's metabolism by CYP2E1, resulting in generation of N-acetyl-p-benzoquinone imine (NAPQI), a toxic intermediate metabolite capable of inducing oxidative stress. NAPQI depletes hepatic glutathione (GSH) content and binds to cellular proteins, with subsequent activation of c-Jun N-terminal kinase (JNK). JNK activation results in overproduction of reactive oxygen species (ROS) and reactive nitrogen species (RNS) resulting in mitochondrial dysfunction and hepatocyte death [2527]. Within 60 minutes of administration of APAP in mice, there is an increase in Nrf2 nuclear translocation in hepatocytes with concomitant increase in the expression of several Nrf2 target genes [28, 29]. Studies using Nrf2-knockout mice showed that even lower doses of APAP could induce mortality due to suppressed GSH synthesis pathway [30]. Contrary to this, hepatocyte-specific deletion of Keap1 activates Nrf2 and confers resistance against APAP toxicity [31]. Interestingly, in Nrf2-knockout mice, the elimination of APAP metabolites is also decreased as a result of reduced expression of multidrug resistance proteins (MRPs) [32]. And, in Keap1-knockout mice, MRP expression is increased, which enhanced efflux of APAP metabolites [29]. These observations indicate that, in APAP-induced hepatotoxicity, Nrf2 modulates injury not only by regulating antioxidant response but also by modulating APAP elimination.

Protein tyrosine phosphorylation is pivotal in cell survival. In this context, the balance between tyrosine kinases and phosphatases determines cell fate. One of the protein tyrosine phosphatases, protein tyrosine phosphatase 1B (PTP1B), is widely expressed and inactivates many tyrosine kinase family members by dephosphorylation. During APAP toxicity in mice, PTP1B expression is significantly increased. PTP1B deficient mouse hepatocytes are protected against APAP-induced GSH depletion and oxidative stress by prolonged Nrf2 nuclear accumulation [33].

Various classes of phytochemicals have been shown to activate Nrf2 pathway and reduce APAP toxicity (Table 1). Sauchinone, a polyphenol, reduces the impact of APAP overdose by enhancing Nrf2 phosphorylation via protein kinase C-δ (PKCδ) and decreasing interaction amongst Nrf2 and Keap1 [34]. Salvianolic acid B reduces APAP-induced liver injury via phosphoinositide-3-kinase- (PI3K-) and PKC-mediated Nrf2 activation, resulting in enhanced HO-1 and GCLC expressions [35]. Sulforaphane (an isothiocyanate found in cruciferous vegetables, namely, cauliflower, broccoli, kale, cole crops, cabbage, collards, brussels sprouts, and mustard) and oleanolic acid (a triterpenoid found in olives) both reduce APAP-induced oxidative stress and liver injury via Nrf2 and related antioxidant gene activation [36, 37]. A recent study from our laboratory evaluated the hepatoprotective potential of withaferin A, an active ingredient of Withania somnifera. We observed that withaferin A treatment 1 h after APAP intoxication activates Nrf2 responsive genes such as NQO1 and GCLC to reduce oxidative stress and subsequent liver injury [38]. An in vitro study, using rat hepatocytes, reported that ginsenoside Rg3 (a ginseng saponin) upregulates antioxidant genes (GCLC, GCLM) and basolateral MRPs via Nrf2 activation [39].

Currently, NAC is the only FDA-approved antidote for APAP hepatotoxicity and is most effective within 8–10 h after APAP overdose. Various studies that utilized phytochemicals to reduce APAP toxicity provide a strong proof of concept that naturally existing activators of Nrf2 can be used to reduce APAP-induced hepatotoxicity. However, these studies have to be interpreted carefully. Many studies have used a preventive experimental strategy, where the phytochemical was administered before inducing APAP overdose, or simultaneously, both unlikely clinical scenarios; patients seek care after APAP overdose. Moreover, such strategies suggest that phytochemicals may interfere with APAP metabolism by suppressing CYP2E1 activity and preventing generation of NAPQI, thus making the interpretation of these studies difficult [40, 41]. In our recent work, we employed an experimental strategy that was clinically relevant, that is, administering WA 1 h after APAP overdose, which provides sufficient time for APAP's metabolism into NAPQI and generation of protein adducts to adequately mimic clinical APAP hepatotoxicity in humans [38, 42]. Therefore, additional studies that not only employ clinically relevant strategies but also use a combination of Nrf2 activators and NAC to combat APAP toxicity are warranted.

3. Acute Inflammatory Liver Injury and Nrf2

Inflammation-mediated repeated liver injury may progress to chronic fibrosis and cirrhosis. Studies in various models of liver injury have shown that Nrf2 plays a role in inflammation-induced liver injury. Lack of an active Nrf2 signaling pathway results in severe inflammation-induced oxidative stress. Concanavalin A (ConA) treatment to mice activates and recruits T-lymphocytes and causes severe inflammation and hepatocyte apoptosis [78, 79]. Following intravenous injection of ConA, Nrf2-KO mice develop increased liver injury compared to WT mice. Hepatocyte-specific conditional Keap1-null mice and WT mice pretreated with three daily doses of CDDO-Im, a Nrf2 activator, had significantly reduced ConA-induced injury compared to Nrf2-KO mice. Similarly, in response to LPS/galactosamine treatment, Nrf2-null mice had increased liver injury compared to WT mice, and, predictably, hepatocyte-specific Keap1-KO mice were protected [80]. These results highlight the importance of Nrf2-mediated regulation of cytokine-dependent hepatocyte apoptosis [81]. In another study, investigators used a short interfering RNA (siRNA) against Keap1 to assess the role of Nrf2 in liver injury; mice were injected with Keap1- or luciferase (control)-siRNA-containing liposomes via the tail vein and, after 48 hours, with ConA. Silencing of hepatic Keap1 attenuated ConA-induced inflammation-associated liver damage [82].

Mangiferin and oroxylin A, the naturally occurring flavonoids, reduce LPS/galactosamine-induced liver injury in mice by reducing oxidative stress and inflammation. Mangiferin enhanced Nrf2/HO-1 signaling and inhibited NLR family, pyrin domain containing 3 (NLRP3) inflammasome [45]. Oroxylin A activated Nrf2 signaling and inhibited Toll-like receptor 4 (TLR4) signaling [46]. Linalool, a major volatile component of essential oils in several aromatic plant species, and ellagic acid (EA), a naturally occurring plant phenol found in certain fruits, nuts, and vegetables, have been shown to possess anti-inflammatory and antioxidant properties. Both of these phytochemicals reduced LPS/galactosamine-induced hepatocyte apoptosis by activating hepatocyte Nrf2-mediated antioxidant response and inflammation by suppressing NF-kB pathway in the liver [43, 44]. These phytochemicals, which reduce both oxidative stress and inflammation, are potential agents for the treatment of fulminant hepatitis in humans.

4. Role of Nrf2 in Chemical-Induced Liver Injury

Carbon tetrachloride (CCl4) is a potent hepatotoxin known to cause centrilobular hepatic necrosis in rodent models of liver fibrosis. Acute administration of CCl4 at higher doses causes severe hepatocyte necrosis, while chronic administration at lower doses is used to induce hepatic fibrosis [40]. However, this model is often criticized because it affects the central zone of the hepatic acinus and some nonmetabolized intermediates of CCl4 can induce lung and kidney injury [83]. In hepatocytes, cytochrome P450 metabolizes CCl4 to trichloromethyl free radicals (CCl3 and/or CCl3OO), which subsequently leads to generation of ROS (O2 , H2O2, and OH) [84]. Chemical hepatotoxins such as bromobenzene, CCl4, and furosemide trigger hepatic Nrf2 nuclear translocation and Nrf2-regulated gene expression [85]. In Nrf2-deficient mice, repair of liver injury after a single treatment with CCl4 was severely delayed [86]. Similarly, after 1-bromopropane exposure, compared to WT mice, Nrf2-knockout mice had increased liver injury with reduced antioxidant response [87].

Lee and colleagues studied the efficacy of diallyl disulfide (DADS), a secondary component derived from garlic, in amelioration of CCl4-induced acute liver injury. Pretreatment with DADS (50 and 100 mg/kg/day) significantly activated Nrf2 expression and antioxidant and phase II enzymes to reduce liver injury [52, 53]. Oxyresveratrol (OXY), an antioxidant present in mulberry fruits and twigs, ameliorated tert-butyl hydroperoxide- (t-BHP-) and CCl4-induced hepatotoxicity by reducing oxidative stress possibly via extracellular signal-regulated kinase- (ERK-) mediated Nrf2 activation in hepatocytes [59]. Other flavonoids such as carthamus red and naringenin also activate Nrf2 signaling to reduce CCl4 injury in rats [50, 58]. Glycyrrhetinic acid, a triterpenoid, and ginsenoside Rg1, a phytoestrogen, ameliorated CCl4-induced liver fibrosis via nuclear Nrf2 translocation and upregulation of antioxidant enzymes to reduce oxidative stress [54, 55].

Several other compounds such as 2,2′-azobis(2-amidinopropane) dihydrochloride (AAPH), menadione (MEN), and carboxymethyllysine (CML) induce free radical-induced damage, and each of them is a potent inducer of hepatotoxicity. Phytocompounds such as antcin C [48], dimerumic acid [88], and schisandrin B [62] have been shown to ameliorate AAPH-, CML-, and MEN-induced liver injury, respectively. Dimethylnitrosamine (DMN) is a semivolatile organic chemical produced as a byproduct of industrial processes and is also known to cause oxidative liver injury. In DMN-administered rats, oral administration of curcumin enhanced nuclear translocation and ARE binding of Nrf2, producing robust antioxidant response thereby reducing hepatotoxicity [51].

In laboratories, t-BHP is a commonly used inducer of oxidative stress in vitro [53, 89, 90]. In hepatocytes, t-BHP is metabolized by CYP450 into free radical intermediates such as t-butoxyl and t-methyl radicals [91], which induce lipid peroxidation and glutathione (GSH) depletion resulting in organelle damage and cell death [92]. Hence, t-BHP-induced hepatotoxicity model is widely used for evaluating merits of Nrf2-ARE signaling in hepatocytes. Various plant flavonoids possess excellent antioxidant potential and confer hepatoprotection. Flavonoids such as hesperidin, butein, and phloretin reduce t-BHP toxicity in hepatocytes via ERK phosphorylation [49, 56], whereas another member of the same class, isoorientin, induces Nrf2 activation via PI3K/Akt pathway [57]. Additionally, phenolics such as 3-caffeoyl, 4-dihydrocaffeoyl quinic acid, and resveratrol ameliorate t-BHP-induced hepatocyte oxidative stress via Nrf2 induction [47, 61]. Another phytoestrogen, puerarin, reduces t-BHP-induced oxidative stress in Hepa1c1c7 cells via PI3K/Akt pathway [60]. These findings underscore the importance of phytochemicals-mediated regulation of Nrf2 activity in diverse models of hepatocyte toxicity, but many of these results need to be validated in animal models of liver injury.

5. Metal Toxicity and Nrf2

Heavy metals, such as cadmium (Cd), chromium (Cr), copper (Cu), lead (Pb), and mercury (Hg), pose health concerns via exposure through water, food, or environmental waste [93]. In experimental models, these heavy metals cause severe oxidative stress and hepatotoxicity. Since these metals induce ROS, it is conceivable that they might induce Nrf2 activation in hepatocytes. A study using Nrf2-KO, Keap1-KO, and hepatocyte-specific Keap1-knockout mice showed that Cd caused extensive liver damage in Nrf2-null mice, whereas Keap1-KO and Keap1-HKO mice were resistant to injury [94].

Arsenic (a metalloid) is a known pollutant for drinking water and has provoked public health concern worldwide. Arsenic modulates Nrf2 signaling in vivo and in vitro [46, 95, 96]. Lutein (a carotenoid) and curcumin (a diarylheptanoid) have been shown to reduce arsenic toxicity via Nrf2 activation and subsequent antioxidant gene expression [63, 64]. Additionally, in mice, curcumin promoted methylation of arsenic and accelerated its excretion [63].

Several studies demonstrated that Cr(VI) induces hepatotoxicity by increasing ROS generation [97, 98]. In Hepa1c1c7 cells, Cr(VI) induces ROS production and in turn triggers activation of Nrf2 pathway [99]. Garlic and its derivative S-allylcysteine (SAC) reduced Cr(VI)-induced hepatotoxicity [65].

In this industrialization era, metal toxicity is a growing concern, and Nrf2 presents a potential target that can be explored to discover novel hepatoprotective agents. These studies suggest that the use of phytochemicals may be helpful in negating the hepatotoxic effects of heavy metals.

6. Nrf2 and Alcoholic Liver Injury

Chronic alcohol consumption is known to result in liver injury-associated deaths [100]. Alcohol abuse leads to increased production of ROS, depletion of hepatocytes antioxidant levels, and enhanced oxidative stress [101, 102]. However, it also leads to an increase in Nrf2 mRNA and protein in the livers [103]. Gong and Cederbaum speculated that the increase in Nrf2 expression was dependent on ethanol-mediated induction of CYP2E1. Compared to WT mice, ethanol-administered Nrf2-KO mice had depletion of total and mitochondrial GSH in their livers resulting in increased liver failure and mortality, while ethanol-fed Keap1-HKO mice were protected [104]. Keap1 knockdown in mice, which induces Nrf2 activation, also blunted ethanol-mediated increase in serum triglycerides and hepatic free fatty acids [105].

Quercetin (a plant flavonoid), a known antioxidant and free radical scavenger, reduces ethanol toxicity in hepatocytes which is Nrf2-mediated. Quercetin upregulates HO-1 via the MAPK/Nrf2 pathway to confer hepatoprotection [67, 68]. A recent study showed that quercetin interacts with Keap1 and blocks its binding to Nrf2 [69]. Sulforaphane (an isothiocyanate) is a known activator of Nrf2 pathway. Zhou et al. showed that sulforaphane induces Nrf2 activation to reduce lipid accumulation and oxidative stress in hepatocytes, an effect independent of CYP2E1 [70]. Lucidone (a naturally occurring cyclopentenedione in Lindera sp.) reduces ethanol-induced oxidative stress in HepG2 cells [66] by inducing Nrf2-mediated antioxidant response via profound upregulation of HO-1.

Despite its known detrimental effects, alcohol remains a major cause of liver injury. These observations from preclinical studies indicate that targeting Nrf2 has merits in treating alcoholic liver injury. These natural Nrf2 activators need to be studied in clinical settings to determine the impact on alcohol abuse-mediated liver injury burden.

7. Nrf2 and Nonalcoholic Steatohepatitis

Nonalcoholic fatty liver disease (NAFLD) is becoming the most common cause of chronic liver disease [106, 107]. In NAFLD, multiple mechanisms operate simultaneously which results in hepatocyte apoptosis, inflammation, and fibrosis, that is, nonalcoholic steatohepatitis (NASH) [107, 108]. In NASH, excessive lipid accumulation and subsequent generation of ROS from impaired mitochondrial respiratory chain result in GSH depletion [109, 110]. It is interesting to note that two research groups showed in mice that high-fat diet (HFD) can increase or decrease hepatic Nrf2 [111]. However, upon HFD feeding, Nrf2-null mice suffered from severe liver injury compared to wild-type mice [112]. Livers of methionine-choline deficient diet- (MCD-) fed Nrf2-knockout mice had higher levels of oxidative stress, iron accumulation, fibrosis, and inflammation than wild-type mice [113115]. It is suggested that Nrf2 activation protects against NAFLD and NASH also via controlling inflammation [115]. The overall merits of Nrf2-mediated prevention of oxidative damage and progression of NAFLD imply its potential use in patients suffering from NASH.

Studies have shown the beneficial role of phytochemicals in treating NASH in preclinical and clinical settings [116118]. Lycopene (a carotenoid derived from tomato) is shown to reduce HFD-induced steatohepatitis in rats [119]. Wang et al. showed that lycopene-induced inhibition of NASH and hepatocarcinogenesis in rats is partially due to induction of Nrf2 and HO-1 genes [74]. Gastrodin (GSTD, a natural compound isolated from Gastrodia elata BI, a traditional Chinese herbal medicine) ameliorates oxidative stress and proinflammatory response in cellular and animal models of NAFLD. GSTD-induced AMPK activation phosphorylates Nrf2, which in turn enhances its nuclear translocation and expression of antioxidant genes (HO-1 and SOD1) [73]. In rats, baicalein (a flavonoid) has been shown to ameliorate MCD-diet-induced NASH by activating multiple pathways including Nrf2-ARE pathway [71]. Similar results have been reported in studies with curcumin where Nrf2 activation ameliorated hepatotoxicity and reduced NASH [72]. Scientific data available till date indicate that the role of natural compounds in Nrf2 activation and alleviation of NASH in experimental models warrants additional human studies. Owing to the multifactorial nature of NASH, a combination of Nrf2 activator and lipid-lowering drugs may translate into an effective therapeutic strategy.

8. Nrf2 and Cholestatic Liver Injury

Cholestasis, a reduction in bile flow, results in a dramatic increase in both liver and serum bile acid concentrations leading to acute liver toxicity, proliferation of bile ducts, and eventually cirrhosis [120]. Bile duct ligated- (BDL-) Nrf2-deficient mice showed reduced elimination of bile acids and higher intrahepatic accumulation of toxic bile acids leading to GSH depletion and injury [121, 122]. BDL-Keap1-KO mice had sustained activation of Nrf2 and increased expression of MRP efflux transporters, detoxifying enzymes, and antioxidant genes [121, 123]. Interestingly, following BDL, Nrf2-KO mice developed into a cholestatic phenotype but liver injury was not different from WT mice undergoing BDL, suggesting that Nrf2 plays a role in the regulation of bile acid homeostasis in the liver [121]. Nrf2-KO mice treated with lithocholic acid (LCA, a toxic bile acid) compared to wild-type mice had severe multifocal liver necrosis and increased inflammation and serum ALT levels [124].

Ursodeoxycholic acid (UDCA), a secondary bile acid, is reported to be hepatoprotective [125, 126]. UDCA treatment of WT mice significantly increased nuclear Nrf2 expression and that of hepatic Mrp2, Mrp3, and Mrp4 [127]. In patients with biliary cirrhosis, UDCA treatment enhanced hepatic Nrf2 expression with an upregulated hepatic thioredoxin and thioredoxin reductase 1 expression [128]. These studies indicate a role of Nrf2 and its activation as a viable target in treating cholestatic liver injury.

Oleanolic acid (OA), a natural triterpenoid, has been shown to reduce LCA- and BDL-induced cholestatic injury in mice [75, 76]. Protection of both of these models is thought to be due to Nrf2-mediated upregulation of Mrps. However, high doses of OA induce cholestatic liver injury in mice by downregulation of hepatic transporters and disruption of bile acid uptake and metabolism [129]. Sulforaphane also reduces cholestasis injury in mice by hepatocyte Nrf2 activation [77]. Alpha-naphthylisothiocyanate (ANIT) is used to induce hepatotoxicity in animal models, which mimics drug-induced cholestatic hepatic injury in humans [130]. Chen et al. demonstrated that paeoniflorin (a monoterpene glycoside) ameliorates ANIT-induced cholestasis in rats by activating Nrf2 via PI3K/Akt pathway [8]. Since Nrf2 regulates both antioxidant genes and multidrug resistance-associated proteins, it has great implication in treating cholestasis.

9. In Vitro Nrf2-ARE Pathway Activation by Phytochemicals

Various hepatocyte cell lines and primary hepatocytes have been used to demonstrate mechanisms of phytochemicals-induced Nrf2 activation. A handful of phytochemicals have been reported to activate Nrf2 signaling in hepatocytes and related cell lines even in the absence of exogenous oxidative stress stimuli. For example, compared to vehicle, phytochemicals such as isothiocyanate (6-methylthiohexyl isothiocyanate), capsaicinoids (capsaicin), terpenoids (carnosol and maslinic acid), polyphenols (eckol, quercetin, and phloretamide), chalconoids (xanthohumol), and flavonoids (epicatechin) induce Nrf2 activation in hepatocytes [131139]. Although these studies comprehensively demonstrate the ability of phytochemicals to induce Nrf2 activation in hepatocytes, one of the merits of these compounds under conditions of oxidative stress, both in vitro and in vivo, is the prevailing lacunae, which leaves many questions unanswered.

10. Cytotoxicity of Phytochemicals

Consumption of herbal mixtures or bioactive molecules can lead to adverse events as well as hepatotoxicity [140]. Phytochemicals found naturally in many plants, fruits, and vegetables are generally believed to be devoid of major side effects [140]. However, safety studies are lacking and many phytochemicals that are marketed as food supplements have already been shown to have adverse effects. Polyphenols and flavonoids, which are popular for their beneficial effects, have been reported to induce adverse events. For example, consumption of polyphenols inhibits nonheme iron absorption and may lead to iron depletion [141]. At higher doses, flavonoids have been reported to act as mutagens, prooxidants, and inhibitors of key enzymes involved in hormone metabolism [142]. Oleanolic acid, which is a known inducer of Nrf2 pathway, has been shown to cause cholestasis at higher doses [129, 143].

Medical literature is littered with various reports of herb-induced hepatotoxicity [144, 145]. For example, black cohosh used for menopausal symptoms can lead to liver failure. We refer the reader to recent articles [146149]. Safety studies for most phytochemicals discussed here are lacking. The FDA is not authorized to review dietary supplement products for safety and effectiveness before they are marketed (http://www.fda.gov). Hence, there is a big gap in the knowledge regarding the safety profile of phytochemicals and toxicity studies will be needed before they are considered safe for human usage.

11. Concluding Remarks

Almost all modes of liver injury are associated with increased oxidative stress and an overwhelmed antioxidant defense system. Since Nrf2 activation is associated with the enhancement of endogenous antioxidant system, it can be an ideal therapeutic target for reducing oxidative stress. Nrf2-ARE pathway boosts hepatocyte antioxidant defense system but may not be able to reduce pathogenesis of liver injury. Hence, a synchronal use of natural Nrf2 activators in combination with other pharmacological agents can be a potential multipronged approach to reduce liver injury. The search for treatments for liver injury is ongoing and potential candidates include whole herbal extracts as well as purified phytochemicals. The literature on preclinical use of phytochemicals to treat various modes of liver injury is ever-growing. However, very few phytochemicals have been tested in clinical settings. Some of the key limitations in this regard are (1) poor choice of experimental models and their clinical relevance, (2) lack of a specific identified mechanism of action, and (3) lack of comprehensive time point data for better interpretation of preclinical findings. These limitations have resulted in weakened significance of these compounds as hepatoprotective agents. Therefore, further studies are needed to explore the full potential of these compounds.

Acknowledgments

This work was supported by SERB EMR/2015/002001 (Ranjitsinh V. Devkar) and the Medical College of Georgia (Sandeep Khurana).

Competing Interests

The authors declare that there are no competing interests regarding the publication of this paper.

References

  • 1.Girish C., Pradhan S. C. Indian herbal medicines in the treatment of liver diseases: problems and promises. Fundamental and Clinical Pharmacology. 2012;26(2):180–189. doi: 10.1111/j.1472-8206.2011.01011.x. [DOI] [PubMed] [Google Scholar]
  • 2.Dhiman R. K., Chawla Y. K. Herbal medicines for liver diseases. Digestive Diseases and Sciences. 2005;50(10):1807–1812. doi: 10.1007/s10620-005-2942-9. [DOI] [PubMed] [Google Scholar]
  • 3.Kuruvilla A. Herbal formulations as pharmacotherapeutic agents. Indian Journal of Experimental Biology. 2002;40(1):7–11. [PubMed] [Google Scholar]
  • 4.Lu K.-H., Liu C.-T., Raghu R., Sheen L.-Y. Therapeutic potential of chinese herbal medicines in alcoholic liver disease. Journal of Traditional and Complementary Medicine. 2012;2(2):115–122. doi: 10.1016/S2225-4110(16)30084-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Farghali H., Canová N. K., Zakhari S. Hepatoprotective properties of extensively studied medicinal plant active constituents: possible common mechanisms. Pharmaceutical Biology. 2014;53(6):781–791. doi: 10.3109/13880209.2014.950387. [DOI] [PubMed] [Google Scholar]
  • 6.Ghosh N., Ghosh R., Mandal V., Mandal S. C. Recent advances in herbal medicine for treatment of liver diseases. Pharmaceutical Biology. 2011;49(9):970–988. doi: 10.3109/13880209.2011.558515. [DOI] [PubMed] [Google Scholar]
  • 7.Cichoz-Lach H., Michalak A. Oxidative stress as a crucial factor in liver diseases. World Journal of Gastroenterology. 2014;20(25):8082–8091. doi: 10.3748/wjg.v20.i25.8082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chen Z., Ma X., Zhu Y., et al. Paeoniflorin ameliorates ANIT-induced cholestasis by activating Nrf2 through an PI3K/Akt-dependent pathway in rats. Phytotherapy Research. 2015;29(11):1768–1775. doi: 10.1002/ptr.5431. [DOI] [PubMed] [Google Scholar]
  • 9.Hoek J. B., Pastorino J. G. Ethanol, oxidative stress, and cytokine-induced liver cell injury. Alcohol. 2002;27(1):63–68. doi: 10.1016/S0741-8329(02)00215-X. [DOI] [PubMed] [Google Scholar]
  • 10.Moi P., Chan K., Asunis I., Cao A., Kan Y. W. Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(21):9926–9930. doi: 10.1073/pnas.91.21.9926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Schäfer M., Werner S. Nrf2—a regulator of keratinocyte redox signaling. Free Radical Biology and Medicine. 2015;88:243–252. doi: 10.1016/j.freeradbiomed.2015.04.018. [DOI] [PubMed] [Google Scholar]
  • 12.Buendia I., Michalska P., Navarro E., Gameiro I., Egea J., León R. Nrf2-ARE pathway: an emerging target against oxidative stress and neuroinflammation in neurodegenerative diseases. Pharmacology and Therapeutics. 2016;157:84–104. doi: 10.1016/j.pharmthera.2015.11.003. [DOI] [PubMed] [Google Scholar]
  • 13.Al-Sawaf O., Clarner T., Fragoulis A., et al. Nrf2 in health and disease: current and future clinical implications. Clinical Science. 2016;129(12):989–999. doi: 10.1042/cs20150436. [DOI] [PubMed] [Google Scholar]
  • 14.Zhang H., Davies K. J., Forman H. J. Oxidative stress response and Nrf2 signaling in aging. Free Radical Biology and Medicine. 2015;88:314–336. doi: 10.1016/j.freeradbiomed.2015.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chen B., Lu Y., Chen Y., Cheng J. The role of Nrf2 in oxidative stress-induced endothelial injuries. Journal of Endocrinology. 2015;225(3):R83–R99. doi: 10.1530/joe-14-0662. [DOI] [PubMed] [Google Scholar]
  • 16.Abdul-Aziz A., Macewan D. J., Bowles K. M., Rushworth S. A. Oxidative stress responses and NRF2 in human leukaemia. Oxidative Medicine and Cellular Longevity. 2015;2015:7. doi: 10.1155/2015/454659.454659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jaramillo M. C., Zhang D. D. The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes & Development. 2013;27(20):2179–2191. doi: 10.1101/gad.225680.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bryan H. K., Olayanju A., Goldring C. E., Park B. K. The Nrf2 cell defence pathway: Keap1-dependent and -independent mechanisms of regulation. Biochemical Pharmacology. 2013;85(6):705–717. doi: 10.1016/j.bcp.2012.11.016. [DOI] [PubMed] [Google Scholar]
  • 19.Kaspar J. W., Niture S. K., Jaiswal A. K. Nrf2:INrf2 (Keap1) signaling in oxidative stress. Free Radical Biology and Medicine. 2009;47(9):1304–1309. doi: 10.1016/j.freeradbiomed.2009.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Reddy S. P. The antioxidant response element and oxidative stress modifiers in airway diseases. Current Molecular Medicine. 2008;8(5):376–383. doi: 10.2174/156652408785160925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Espinosa-Diez C., Miguel V., Mennerich D., et al. Antioxidant responses and cellular adjustments to oxidative stress. Redox Biology. 2015;6:183–197. doi: 10.1016/j.redox.2015.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fontana R. J. Acute liver failure including acetaminophen overdose. Medical Clinics of North America. 2008;92(4):761–794. doi: 10.1016/j.mcna.2008.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lee W. M. Acetaminophen and the U.S. Acute Liver Failure Study Group: lowering the risks of hepatic failure. Hepatology. 2004;40(1):6–9. doi: 10.1002/hep.20293. [DOI] [PubMed] [Google Scholar]
  • 24.Mitchell J. R., Jollow D. J., Potter W. Z., Davis D. C., Gillette J. R., Brodie B. B. Acetaminophen induced hepatic necrosis. I. Role of drug metabolism. Journal of Pharmacology and Experimental Therapeutics. 1973;187(1):185–194. [PubMed] [Google Scholar]
  • 25.Hanawa N., Shinohara M., Saberi B., Gaarde W. A., Han D., Kaplowitz N. Role of JNK translocation to mitochondria leading to inhibition of mitochondria bioenergetics in acetaminophen-induced liver injury. Journal of Biological Chemistry. 2008;283(20):13565–13577. doi: 10.1074/jbc.M708916200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Knight T. R., Ho Y.-S., Farhood A., Jaeschke H. Peroxynitrite is a critical mediator of acetaminophen hepatotoxicity in murine livers: protection by glutathione. Journal of Pharmacology and Experimental Therapeutics. 2002;303(2):468–475. doi: 10.1124/jpet.102.038968. [DOI] [PubMed] [Google Scholar]
  • 27.Saito C., Lemasters J. J., Jaeschke H. c-Jun N-terminal kinase modulates oxidant stress and peroxynitrite formation independent of inducible nitric oxide synthase in acetaminophen hepatotoxicity. Toxicology and Applied Pharmacology. 2010;246(1-2):8–17. doi: 10.1016/j.taap.2010.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Goldring C. E. P., Kitteringham N. R., Elsby R., et al. Activation of hepatic Nrf2 in vivo by acetaminophen in CD-1 mice. Hepatology. 2004;39(5):1267–1276. doi: 10.1002/hep.20183. [DOI] [PubMed] [Google Scholar]
  • 29.Aleksunes L. M., Slitt A. L., Maher J. M., et al. Induction of Mrp3 and Mrp4 transporters during acetaminophen hepatotoxicity is dependent on Nrf2. Toxicology and Applied Pharmacology. 2008;226(1):74–83. doi: 10.1016/j.taap.2007.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chan K., Han X.-D., Kan Y. W. An important function of Nrf2 in combating oxidative stress: detoxification of acetaminophen. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(8):4611–4616. doi: 10.1073/pnas.081082098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Okawa H., Motohashi H., Kobayashi A., Aburatani H., Kensler T. W., Yamamoto M. Hepatocyte-specific deletion of the keap1 gene activates Nrf2 and confers potent resistance against acute drug toxicity. Biochemical and Biophysical Research Communications. 2006;339(1):79–88. doi: 10.1016/j.bbrc.2005.10.185. [DOI] [PubMed] [Google Scholar]
  • 32.Reisman S. A., Csanaky I. L., Aleksunes L. M., Klaassen C. D. Altered disposition of acetaminophen in Nrf2-null and Keap1-knockdown mice. Toxicological Sciences. 2009;109(1):31–40. doi: 10.1093/toxsci/kfp047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Mobasher M. A., González-Rodriguez A., Santamaría B., et al. Protein tyrosine phosphatase 1B modulates GSK3b/Nrf2 and IGFIR signaling pathways in acetaminophen-induced hepatotoxicity. Cell Death and Disease. 2013;4(5, article e626) doi: 10.1038/cddis.2013.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kay H. Y., Kim Y. W., Ryu D. H., Sung S. H., Hwang S. J., Kim S. G. Nrf2-mediated liver protection by sauchinone, an antioxidant lignan, from acetaminophen toxicity through the PKCδ-GSK3β pathway. British Journal of Pharmacology. 2011;163(8):1653–1665. doi: 10.1111/j.1476-5381.2010.01095.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lin M., Zhai X., Wang G., et al. Salvianolic acid B protects against Acetaminophen hepatotoxicity by inducing Nrf2 and phase II detoxification gene expression via activation of the PI3K and PKC signaling pathways. Journal of Pharmacological Sciences. 2015;127(2):203–210. doi: 10.1016/j.jphs.2014.12.010. [DOI] [PubMed] [Google Scholar]
  • 36.Noh J., Kim Y., Hwang J. H., et al. Sulforaphane protects against acetaminophen-induced hepatotoxicity. Food and Chemical Toxicology. 2015;80:193–200. doi: 10.1016/j.fct.2015.03.020. [DOI] [PubMed] [Google Scholar]
  • 37.Reisman S. A., Aleksunes L. M., Klaassen C. D. Oleanolic acid activates Nrf2 and protects from acetaminophen hepatotoxicity via Nrf2-dependent and Nrf2-independent processes. Biochemical Pharmacology. 2009;77(7):1273–1282. doi: 10.1016/j.bcp.2008.12.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jadeja R. N., Urrunaga N. H., Dash S., Khurana S., Saxena N. K. Withaferin-a reduces acetaminophen-induced liver injury in mice. Biochemical Pharmacology. 2015;97(1):122–132. doi: 10.1016/j.bcp.2015.07.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gum S. I., Cho M. K. The amelioration of N-acetyl-p-benzoquinone imine toxicity by ginsenoside Rg3: the role of Nrf2-mediated detoxification and Mrp1/Mrp3 transports. Oxidative Medicine and Cellular Longevity. 2013;2013:11. doi: 10.1155/2013/957947.957947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Jaeschke H., Williams C. D., McGill M. R., Xie Y., Ramachandran A. Models of drug-induced liver injury for evaluation of phytotherapeutics and other natural products. Food and Chemical Toxicology. 2013;55:279–289. doi: 10.1016/j.fct.2012.12.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jaeschke H., McGill M. R., Williams C. D., Ramachandran A. Current issues with acetaminophen hepatotoxicity—a clinically relevant model to test the efficacy of natural products. Life Sciences. 2011;88(17-18):737–745. doi: 10.1016/j.lfs.2011.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Du K., McGill M. R., Xie Y., Bajt M. L., Jaeschke H. Resveratrol prevents protein nitration and release of endonucleases from mitochondria during acetaminophen hepatotoxicity. Food and Chemical Toxicology. 2015;81:62–70. doi: 10.1016/j.fct.2015.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gu L., Deng W.-S., Liu Y., et al. Ellagic acid protects Lipopolysaccharide/D-galactosamine-induced acute hepatic injury in mice. International immunopharmacology. 2014;22(2):341–345. doi: 10.1016/j.intimp.2014.07.005. [DOI] [PubMed] [Google Scholar]
  • 44.Li J., Zhang X., Huang H. Protective effect of linalool against lipopolysaccharide/D-galactosamine-induced liver injury in mice. International Immunopharmacology. 2014;23(2):523–529. doi: 10.1016/j.intimp.2014.10.001. [DOI] [PubMed] [Google Scholar]
  • 45.Pan C.-W., Pan Z.-Z., Hu J.-J., et al. Mangiferin alleviates lipopolysaccharide and D-galactosamine-induced acute liver injury by activating the Nrf2 pathway and inhibiting NLRP3 inflammasome activation. European Journal of Pharmacology. 2016;770:85–91. doi: 10.1016/j.ejphar.2015.12.006. [DOI] [PubMed] [Google Scholar]
  • 46.Huang H., Zhang X., Li J. Protective effect of oroxylin A against lipopolysaccharide and/or D-galactosamine-induced acute liver injury in mice. Journal of Surgical Research. 2015;195(2):522–528. doi: 10.1016/j.jss.2015.01.047. [DOI] [PubMed] [Google Scholar]
  • 47.Hwang Y. P., Yun H. J., Chun H. K., et al. Protective mechanisms of 3-caffeoyl, 4-dihydrocaffeoyl quinic acid from Salicornia herbacea against tert-butyl hydroperoxide-induced oxidative damage. Chemico-Biological Interactions. 2009;181(3):366–376. doi: 10.1016/j.cbi.2009.07.017. [DOI] [PubMed] [Google Scholar]
  • 48.Gokila Vani M., Senthil Kumar K. J., Liao J.-W., et al. Antcin C from Antrodia cinnamomea protects liver cells against free radical-induced oxidative stress and apoptosis in vitro and in vivo through Nrf2-dependent mechanism. Evidence-Based Complementary and Alternative Medicine. 2013;2013:17. doi: 10.1155/2013/296082.296082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yang Y.-C., Lii C.-K., Lin A.-H., et al. Induction of glutathione synthesis and heme oxygenase 1 by the flavonoids butein and phloretin is mediated through the ERK/Nrf2 pathway and protects against oxidative stress. Free Radical Biology and Medicine. 2011;51(11):2073–2081. doi: 10.1016/j.freeradbiomed.2011.09.007. [DOI] [PubMed] [Google Scholar]
  • 50.Wu S. C., Yue Y., Tian H., et al. Carthamus red from Carthamus tinctorius L. exerts antioxidant and hepatoprotective effect against CCl4-induced liver damage in rats via the Nrf2 pathway. Journal of Ethnopharmacology. 2013;148(2):570–578. doi: 10.1016/j.jep.2013.04.054. [DOI] [PubMed] [Google Scholar]
  • 51.Farombi E. O., Shrotriya S., Na H.-K., Kim S.-H., Surh Y.-J. Curcumin attenuates dimethylnitrosamine-induced liver injury in rats through Nrf2-mediated induction of heme oxygenase-1. Food and Chemical Toxicology. 2008;46(4):1279–1287. doi: 10.1016/j.fct.2007.09.095. [DOI] [PubMed] [Google Scholar]
  • 52.Lee I.-C., Kim S.-H., Baek H.-S., et al. The involvement of Nrf2 in the protective effects of diallyl disulfide on carbon tetrachloride-induced hepatic oxidative damage and inflammatory response in rats. Food and Chemical Toxicology. 2014;63:174–185. doi: 10.1016/j.fct.2013.11.006. [DOI] [PubMed] [Google Scholar]
  • 53.Lee I.-C., Kim S.-H., Baek H.-S., et al. Protective effects of diallyl disulfide on carbon tetrachloride-induced hepatotoxicity through activation of Nrf2. Environmental Toxicology. 2016;30(5):538–548. doi: 10.1002/tox.21930. [DOI] [PubMed] [Google Scholar]
  • 54.Li J.-P., Gao Y., Chu S.-F., et al. Nrf2 pathway activation contributes to anti-fibrosis effects of ginsenoside Rg1 in a rat model of alcohol- and CCl4-induced hepatic fibrosis. Acta Pharmacologica Sinica. 2014;35(8):1031–1044. doi: 10.1038/aps.2014.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chen S., Zou L., Li L., Wu T. The protective effect of glycyrrhetinic acid on carbon tetrachloride-induced chronic liver fibrosis in mice via upregulation of Nrf2. PLoS ONE. 2013;8(1) doi: 10.1371/journal.pone.0053662.e53662 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Chen M., Gu H., Ye Y., et al. Protective effects of hesperidin against oxidative stress of tert-butyl hydroperoxide in human hepatocytes. Food and Chemical Toxicology. 2010;48(10):2980–2987. doi: 10.1016/j.fct.2010.07.037. [DOI] [PubMed] [Google Scholar]
  • 57.Lim J. H., Park H.-S., Choi J.-K., Lee I.-S., Hyun J. C. Isoorientin induces Nrf2 pathway-driven antioxidant response through phosphatidylinositol 3-kinase signaling. Archives of Pharmacal Research. 2007;30(12):1590–1598. doi: 10.1007/bf02977329. [DOI] [PubMed] [Google Scholar]
  • 58.Esmaeili M. A., Alilou M. Naringenin attenuates CCl4-induced hepatic inflammation by the activation of an Nrf2-mediated pathway in rats. Clinical and Experimental Pharmacology and Physiology. 2014;41(6):416–422. doi: 10.1111/1440-1681.12230. [DOI] [PubMed] [Google Scholar]
  • 59.Choi H. Y., Lee J.-H., Jegal K. H., Cho I. J., Kim Y. W., Kim S. C. Oxyresveratrol abrogates oxidative stress by activating ERK-Nrf2 pathway in the liver. Chemico-Biological Interactions. 2016;245:110–121. doi: 10.1016/j.cbi.2015.06.024. [DOI] [PubMed] [Google Scholar]
  • 60.Hwang Y. P., Jeong H. G. Mechanism of phytoestrogen puerarin-mediated cytoprotection following oxidative injury: estrogen receptor-dependent up-regulation of PI3K/Akt and HO-1. Toxicology and Applied Pharmacology. 2008;233(3):371–381. doi: 10.1016/j.taap.2008.09.006. [DOI] [PubMed] [Google Scholar]
  • 61.Rubiolo J. A., Mithieux G., Vega F. V. Resveratrol protects primary rat hepatocytes against oxidative stress damage: activation of the Nrf2 transcription factor and augmented activities of antioxidant enzymes. European Journal of Pharmacology. 2008;591(1–3):66–72. doi: 10.1016/j.ejphar.2008.06.067. [DOI] [PubMed] [Google Scholar]
  • 62.Leong P. K., Chiu P. Y., Chen N., Leung H., Ko K. M. Schisandrin B elicits a glutathione antioxidant response and protects against apoptosis via the redox-sensitive ERK/Nrf2 pathway in AML12 hepatocytes. Free Radical Research. 2011;45(4):483–495. doi: 10.3109/10715762.2010.550917. [DOI] [PubMed] [Google Scholar]
  • 63.Gao S., Duan X. X., Wang X., et al. Curcumin attenuates arsenic-induced hepatic injuries and oxidative stress in experimental mice through activation of Nrf2 pathway, promotion of arsenic methylation and urinary excretion. Food and Chemical Toxicology. 2013;59:739–747. doi: 10.1016/j.fct.2013.07.032. [DOI] [PubMed] [Google Scholar]
  • 64.Li S., Ding Y., Niu Q., et al. Lutein has a protective effect on hepatotoxicity induced by arsenic via Nrf2 signaling. BioMed Research International. 2015;2015 doi: 10.1155/2015/315205.315205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kalayarasan S., Sriram N., Sureshkumar A., Sudhandiran G. Chromium (VI)-induced oxidative stress and apoptosis is reduced by garlic and its derivative S-allylcysteine through the activation of Nrf2 in the hepatocytes of Wistar rats. Journal of Applied Toxicology. 2008;28(7):908–919. doi: 10.1002/jat.1355. [DOI] [PubMed] [Google Scholar]
  • 66.Senthil Kumar K. J., Liao J.-W., Xiao J.-H., Gokila Vani M., Wang S.-Y. Hepatoprotective effect of lucidone against alcohol-induced oxidative stress in human hepatic HepG2 cells through the up-regulation of HO-1/Nrf-2 antioxidant genes. Toxicology in Vitro. 2012;26(5):700–708. doi: 10.1016/j.tiv.2012.03.012. [DOI] [PubMed] [Google Scholar]
  • 67.Yao P., Nussler A., Liu L., et al. Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. Journal of Hepatology. 2007;47(2):253–261. doi: 10.1016/j.jhep.2007.02.008. [DOI] [PubMed] [Google Scholar]
  • 68.Liu S., Hou W., Yao P., et al. Heme oxygenase-1 mediates the protective role of quercetin against ethanol-induced rat hepatocytes oxidative damage. Toxicology in Vitro. 2012;26(1):74–80. doi: 10.1016/j.tiv.2011.10.013. [DOI] [PubMed] [Google Scholar]
  • 69.Ji L.-L., Sheng Y.-C., Zheng Z.-Y., Shi L., Wang Z.-T. The involvement of p62-Keap1-Nrf2 antioxidative signaling pathway and JNK in the protection of natural flavonoid quercetin against hepatotoxicity. Free Radical Biology and Medicine. 2015;85:12–23. doi: 10.1016/j.freeradbiomed.2015.03.035. [DOI] [PubMed] [Google Scholar]
  • 70.Zhou R., Lin J. J., Wu D. F. Sulforaphane induces Nrf2 and protects against CYP2E1-dependent binge alcohol-induced liver steatosis. Biochimica et Biophysica Acta (BBA)—General Subjects. 2014;1840(1):209–218. doi: 10.1016/j.bbagen.2013.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Xin H.-G., Zhang B.-B., Wu Z.-Q., et al. Treatment with baicalein attenuates methionine-choline deficient diet-induced non-alcoholic steatohepatitis in rats. European Journal of Pharmacology. 2014;738:310–318. doi: 10.1016/j.ejphar.2014.06.006. [DOI] [PubMed] [Google Scholar]
  • 72.Li B., Wang L., Lu Q., Da W. Liver injury attenuation by curcumin in a rat NASH model: an Nrf2 activation-mediated effect? Irish Journal of Medical Science. 2016;185(1):93–100. doi: 10.1007/s11845-014-1226-9. [DOI] [PubMed] [Google Scholar]
  • 73.Qu L. L., Yu B., Li Z., Jiang W. X., Jiang J. D., Kong W. J. Gastrodin ameliorates oxidative stress and proinflammatory response in nonalcoholic fatty liver disease through the AMPK/Nrf2 pathway. Phytotherapy Research. 2016;30(3):402–411. doi: 10.1002/ptr.5541. [DOI] [PubMed] [Google Scholar]
  • 74.Wang Y., Ausman L. M., Greenberg A. S., Russell R. M., Wang X.-D. Dietary lycopene and tomato extract supplementations inhibit nonalcoholic steatohepatitis-promoted hepatocarcinogenesis in rats. International Journal of Cancer. 2010;126(8):1788–1796. doi: 10.1002/ijc24689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen P., Zeng H., Wang Y., et al. Low dose of oleanolic acid protects against lithocholic acid-induced cholestasis in mice: potential involvement of nuclear factor-E2-related factor 2-mediated upregulation of multidrug resistance-associated proteinss. Drug Metabolism and Disposition. 2014;42(5):844–852. doi: 10.1124/dmd.113.056549. [DOI] [PubMed] [Google Scholar]
  • 76.Chen P., Li J., Fan X., et al. Oleanolic acid attenuates obstructive cholestasis in bile duct-ligated mice, possibly via activation of NRF2-MRPs and FXR antagonism. European Journal of Pharmacology. 2015;765:131–139. doi: 10.1016/j.ejphar.2015.08.029.70177 [DOI] [PubMed] [Google Scholar]
  • 77.Oh C. J., Kim J.-Y., Min A.-K., et al. Sulforaphane attenuates hepatic fibrosis via NF-E2-related factor 2-mediated inhibition of transforming growth factor-β/Smad signaling. Free Radical Biology and Medicine. 2012;52(3):671–682. doi: 10.1016/j.freeradbiomed.2011.11.012. [DOI] [PubMed] [Google Scholar]
  • 78.Gantner F., Leist M., Lohse A. W., Germann P. G., Tiegs G. Concanavalin A-induced T-cell-mediated hepatic injury in mice: the role of tumor necrosis factor. Hepatology. 1995;21(1):190–198. doi: 10.1016/0270-9139(95)90428-x. [DOI] [PubMed] [Google Scholar]
  • 79.Heymann F., Hamesch K., Weiskirchen R., Tacke F. The concanavalin a model of acute hepatitis in mice. Laboratory Animals. 2015;49, S1:12–20. doi: 10.1177/0023677215572841. [DOI] [PubMed] [Google Scholar]
  • 80.Liu J., Wu K. C., Lu Y.-F., Ekuase E., Klaassen C. D. NRF2 protection against liver injury produced by various hepatotoxicants. Oxidative Medicine and Cellular Longevity. 2013;2013:8. doi: 10.1155/2013/305861.305861 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Osburn W. O., Yates M. S., Dolan P. D., et al. Genetic or pharmacologic amplification of Nrf2 signaling inhibits acute inflammatory liver injury in mice. Toxicological Sciences. 2008;104(1):218–227. doi: 10.1093/toxsci/kfn079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.González-Rodríguez A., Reibert B., Amann T., Constien R., Rondinone C. M., Valverde A. M. In vivo siRNA delivery of Keap1 modulates death and survival signaling pathways and attenuates concanavalin-A-induced acute liver injury in mice. Disease Models and Mechanisms. 2014;7(9):1093–1100. doi: 10.1242/dmm.015537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tuñón M. J., Alvarez M., Culebras J. M., González-Gallego J. An overview of animal models for investigating the pathogenesis and therapeutic strategies in acute hepatic failure. World Journal of Gastroenterology. 2009;15(25):3086–3098. doi: 10.3748/wjg.15.3086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Weber L. W. D., Boll M., Stampfl A. Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Critical Reviews in Toxicology. 2003;33(2):105–136. doi: 10.1080/713611034. [DOI] [PubMed] [Google Scholar]
  • 85.Randle L. E., Goldring C. E. P., Benson C. A., et al. Investigation of the effect of a panel of model hepatotoxins on the Nrf2-Keap1 defence response pathway in CD-1 mice. Toxicology. 2008;243(3):249–260. doi: 10.1016/j.tox.2007.10.011. [DOI] [PubMed] [Google Scholar]
  • 86.Xu W., Hellerbrand C., Köhler U. A., et al. The Nrf2 transcription factor protects from toxin-induced liver injury and fibrosis. Laboratory Investigation. 2008;88(10):1068–1078. doi: 10.1038/labinvest.2008.75. [DOI] [PubMed] [Google Scholar]
  • 87.Liu F., Ichihara S., Valentine W. M., et al. Increased susceptibility of Nrf2-null mice to 1-bromopropane-induced hepatotoxicity. Toxicological Sciences. 2010;115(2):596–606. doi: 10.1093/toxsci/kfq075. [DOI] [PubMed] [Google Scholar]
  • 88.Lee B.-H., Hsu W.-H., Hsu Y.-W., Pan T.-M. Suppression of dimerumic acid on hepatic fibrosis caused from carboxymethyl-lysine (CML) by attenuating oxidative stress depends on Nrf2 activation in hepatic stellate cells (HSCs) Food and Chemical Toxicology. 2013;62:413–419. doi: 10.1016/j.fct.2013.09.007. [DOI] [PubMed] [Google Scholar]
  • 89.Liu C.-L., Wang J.-M., Chu C.-Y., Cheng M.-T., Tseng T.-H. In vivo protective effect of protocatechuic acid on tert-butyl hydroperoxide-induced rat hepatotoxicity. Food and Chemical Toxicology. 2002;40(5):635–641. doi: 10.1016/S0278-6915(02)00002-9. [DOI] [PubMed] [Google Scholar]
  • 90.Lee D.-S., Kim K.-S., Ko W., et al. The cytoprotective effect of sulfuretin against tert-Butyl Hydroperoxide-induced hepatotoxicity through Nrf2/ARE and JNK/ERK MAPK-mediated Heme Oxygenase-1 expression. International Journal of Molecular Sciences. 2014;15(5):8863–8877. doi: 10.3390/ijms15058863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Hix S., Kadiiska M. B., Mason R. P., Augusto O. In vivo metabolism of tert-Butyl hydroperoxide to methyl radicals. EPR spin-trapping and DNA methylation studies. Chemical Research in Toxicology. 2000;13(10):1056–1064. doi: 10.1021/tx000130l. [DOI] [PubMed] [Google Scholar]
  • 92.Oh J. M., Jung Y. S., Jeon B. S., et al. Evaluation of hepatotoxicity and oxidative stress in rats treated with tert-butyl hydroperoxide. Food and Chemical Toxicology. 2012;50(5):1215–1221. doi: 10.1016/j.fct.2012.01.031. [DOI] [PubMed] [Google Scholar]
  • 93.Jarup L. Hazards of heavy metal contamination. British Medical Bulletin. 2003;68(1):167–182. doi: 10.1093/bmb/ldg032. [DOI] [PubMed] [Google Scholar]
  • 94.Wu K. C., Liu J. J., Klaassen C. D. Nrf2 activation prevents cadmium-induced acute liver injury. Toxicology and Applied Pharmacology. 2012;263(1):14–20. doi: 10.1016/j.taap.2012.05.017. [DOI] [PubMed] [Google Scholar]
  • 95.Liu D., Duan X., Dong D., et al. Activation of the Nrf2 pathway by inorganic arsenic in human hepatocytes and the role of transcriptional repressor Bach1. Oxidative Medicine and Cellular Longevity. 2013;2013:10. doi: 10.1155/2013/984546.984546 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Abiko Y., Shinkai Y., Sumi D., Kumagai Y. Reduction of arsenic-induced cytotoxicity through Nrf2/HO-1 signaling in HepG2 cells. Journal of Toxicological Sciences. 2010;35(3):419–423. doi: 10.2131/jts.35.419. [DOI] [PubMed] [Google Scholar]
  • 97.Wang X.-F., Xing M.-L., Shen Y., Zhu X., Xu L.-H. Oral administration of Cr(VI) induced oxidative stress, DNA damage and apoptotic cell death in mice. Toxicology. 2006;228(1):16–23. doi: 10.1016/j.tox.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 98.Patlolla A. K., Barnes C., Yedjou C., Velma V. R., Tchounwou P. B. Oxidative stress, DNA damage, and antioxidant enzyme activity induced by hexavalent chromium in Sprague-Dawley rats. Environmental Toxicology. 2009;24(1):66–73. doi: 10.1002/tox.20395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.He X. Q., Lin G. X., Chen M. G., Zhang J. X., Ma Q. Protection against chromium (VI)-induced oxidative stress and apoptosis by Nrf2. Recruiting Nrf2 into the nucleus and disrupting the nuclear Nrf2/Keap1 association. Toxicological Sciences. 2007;98(1):298–309. doi: 10.1093/toxsci/kfm081. [DOI] [PubMed] [Google Scholar]
  • 100.Torruellas C., French S. W., Medici V. Diagnosis of alcoholic liver disease. World Journal of Gastroenterology. 2014;20(33):11684–11699. doi: 10.3748/wjg.v20.i33.11684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wu D., Cederbaum A. I. Oxidative stress and alcoholic liver disease. Seminars in Liver Disease. 2009;29(2):141–154. doi: 10.1055/s-0029-1214370. [DOI] [PubMed] [Google Scholar]
  • 102.Cederbaum A. I., Lu Y., Wu D. Role of oxidative stress in alcohol-induced liver injury. Archives of Toxicology. 2009;83(6):519–548. doi: 10.1007/s00204-009-0432-0. [DOI] [PubMed] [Google Scholar]
  • 103.Gong P. E., Cederbaum A. I. Nrf2 is increased by CYP2E1 in rodent liver and HepG2 cells and protects against oxidative stress caused by CYP2E1. Hepatology. 2006;43(1):144–153. doi: 10.1002/hep.21004. [DOI] [PubMed] [Google Scholar]
  • 104.Lamlé J., Marhenke S., Borlak J., et al. Nuclear factor-eythroid 2-related factor 2 prevents alcohol-induced fulminant liver injury. Gastroenterology. 2008;134(4):1159–1168. doi: 10.1053/j.gastro.2008.01.011. [DOI] [PubMed] [Google Scholar]
  • 105.Wu K. C., Liu J., Klaassen C. D. Role of Nrf2 in preventing ethanol-induced oxidative stress and lipid accumulation. Toxicology and Applied Pharmacology. 2012;262(3):321–329. doi: 10.1016/j.taap.2012.05.010. [DOI] [PubMed] [Google Scholar]
  • 106.Azzam H., Malnick S. Non-alcoholic fatty liver disease—the heart of the matter. World Journal of Hepatology. 2015;7(10):1369–1376. doi: 10.4254/wjh.v7.i10.1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Byrne C. D., Targher G. NAFLD: a multisystem disease. Journal of Hepatology. 2015;62(1):S47–S64. doi: 10.1016/j.jhep.2014.12.012. [DOI] [PubMed] [Google Scholar]
  • 108.Williams T. Metabolic syndrome: nonalcoholic fatty liver disease. FP Essentials. 2015;435:24–29. [PubMed] [Google Scholar]
  • 109.Liu W., Baker S. S., Baker R. D., Zhu L. Antioxidant mechanisms in nonalcoholic fatty liver disease. Current Drug Targets. 2015;16(12):1301–1314. doi: 10.2174/1389450116666150427155342. [DOI] [PubMed] [Google Scholar]
  • 110.Serviddio G., Bellanti F., Vendemiale G. Free radical biology for medicine: learning from nonalcoholic fatty liver disease. Free Radical Biology and Medicine. 2013;65:952–968. doi: 10.1016/j.freeradbiomed.2013.08.174. [DOI] [PubMed] [Google Scholar]
  • 111.Kim S., Sohn I., Ahn J.-I., Lee K.-H., Lee Y. S., Lee Y. S. Hepatic gene expression profiles in a long-term high-fat diet-induced obesity mouse model. Gene. 2004;340(1):99–109. doi: 10.1016/j.gene.2004.06.015. [DOI] [PubMed] [Google Scholar]
  • 112.Tanaka Y., Aleksunes L. M., Yeager R. L., et al. NF-E2-related factor 2 inhibits lipid accumulation and oxidative stress in mice fed a high-fat diet. Journal of Pharmacology and Experimental Therapeutics. 2008;325(2):655–664. doi: 10.1124/jpet.107.135822. [DOI] [PubMed] [Google Scholar]
  • 113.Chowdhry S., Nazmy M. H., Meakin P. J., et al. Loss of Nrf2 markedly exacerbates nonalcoholic steatohepatitis. Free Radical Biology and Medicine. 2010;48(2):357–371. doi: 10.1016/j.freeradbiomed.2009.11.007. [DOI] [PubMed] [Google Scholar]
  • 114.Okada K., Warabi E., Sugimoto H., et al. Nrf2 inhibits hepatic iron accumulation and counteracts oxidative stress-induced liver injury in nutritional steatohepatitis. Journal of Gastroenterology. 2012;47(8):924–935. doi: 10.1007/s00535-012-0552-9. [DOI] [PubMed] [Google Scholar]
  • 115.Sugimoto H., Okada K., Shoda J., et al. Deletion of nuclear factor-E2-related factor-2 leads to rapid onset and progression of nutritional steatohepatitis in mice. American Journal of Physiology—Gastrointestinal and Liver Physiology. 2010;298(2):G283–G294. doi: 10.1152/ajpgi.00296.2009. [DOI] [PubMed] [Google Scholar]
  • 116.Jadeja R., Devkar R. V., Nammi S. Herbal medicines for the treatment of nonalcoholic steatohepatitis: Current scenario and future prospects. Evidence-Based Complementary and Alternative Medicine. 2014;2014:18. doi: 10.1155/2014/648308.648308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Xiao J., So K. F., Liong E. C., Tipoe G. L. Recent advances in the herbal treatment of non-alcoholic fatty liver disease. Journal of Traditional and Complementary Medicine. 2013;3(2):88–94. doi: 10.4103/2225-4110.110411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Miloševič N., Milanovič M., Abenavoli L., Milič N. Phytotherapy and nafld-rom goals and challenges to clinical practice. Reviews on Recent Clinical Trials. 2014;9(3):195–203. [PubMed] [Google Scholar]
  • 119.Bahcecioglu I. H., Kuzu N., Metin K., et al. Lycopene prevents development of steatohepatitis in experimental nonalcoholic steatohepatitis model induced by high-fat diet. Veterinary Medicine International. 2010;2010:8. doi: 10.4061/2010/262179.262179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Jaeschke H., Gores G. J., Cederbaum A. I., Hinson J. A., Pessayre D., Lemasters J. J. Mechanisms of hepatotoxicity. Toxicological Sciences. 2002;65(2):166–176. doi: 10.1093/toxsci/65.2.166. [DOI] [PubMed] [Google Scholar]
  • 121.Weerachayaphorn J., Mennone A., Soroka C. J., et al. Nuclear factor-E2-related factor 2 is a major determinant of bile acid homeostasis in the liver and intestine. American Journal of Physiology—Gastrointestinal and Liver Physiology. 2012;302(9):G925–G936. doi: 10.1152/ajpgi.00263.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Aleksunes L. M., Slitt A. L., Maher J. M., et al. Nuclear factor-E2-related factor 2 expression in liver is critical for induction of NAD(P)H:quinone oxidoreductase 1 during cholestasis. Cell Stress and Chaperones. 2006;11(4):356–363. doi: 10.1379/CSC-217.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Okada K., Shoda J., Taguchi K., et al. Nrf2 counteracts cholestatic liver injury via stimulation of hepatic defense systems. Biochemical and Biophysical Research Communications. 2009;389(3):431–436. doi: 10.1016/j.bbrc.2009.08.156. [DOI] [PubMed] [Google Scholar]
  • 124.Tan K. P., Wood G. A., Yang M., Ito S. Participation of nuclear factor (erythroid 2-related), factor 2 in ameliorating lithocholic acid-induced cholestatic liver injury in mice. British Journal of Pharmacology. 2010;161(5):1111–1121. doi: 10.1111/j.1476-5381.2010.00953.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lukivskaya O., Zavodnik L., Knas M., Buko V. Antioxidant mechanism of hepatoprotection by ursodeoxycholic acid in experimental alcoholic steatohepatitis. Advances in Medical Sciences. 2006;51:54–59. [PubMed] [Google Scholar]
  • 126.Ishizaki K., Imada T., Tsurufuji M. Hepatoprotective bile acid 'ursodeoxycholic acid (UDCA)': property and difference as bile acids. Hepatology Research. 2005;33(2):174–177. doi: 10.1016/j.hepres.2005.09.029. [DOI] [PubMed] [Google Scholar]
  • 127.Paumgartner G., Beuers U. Mechanisms of action and therapeutic efficacy of ursodeoxycholic acid in cholestatic liver disease. Clinics in Liver Disease. 2004;8(1):67–81. doi: 10.1016/S1089-3261(03)00135-1. [DOI] [PubMed] [Google Scholar]
  • 128.Kawata K., Kobayashi Y., Souda K., et al. Enhanced hepatic Nrf2 activation after ursodeoxycholic acid treatment in patients with primary biliary cirrhosis. Antioxidants and Redox Signaling. 2010;13(3):259–268. doi: 10.1089/ars.2009.2903. [DOI] [PubMed] [Google Scholar]
  • 129.Lu Y.-F., Wan X.-L., Xu Y., Liu J. Repeated oral administration of oleanolic acid produces cholestatic liver injury in mice. Molecules. 2013;18(3):3060–3071. doi: 10.3390/molecules18033060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Plaa G. L., Priestly B. G. Intrahepatic cholestasis induced by drugs and chemicals. Pharmacological Reviews. 1976;28(3):207–273. [PubMed] [Google Scholar]
  • 131.Korenori Y., Tanigawa S., Kumamoto T., et al. Modulation of Nrf2/Keap1 system by Wasabi 6-methylthiohexyl isothiocyanate in ARE-mediated NQO1 expression. Molecular Nutrition and Food Research. 2013;57(5):854–864. doi: 10.1002/mnfr.201200689. [DOI] [PubMed] [Google Scholar]
  • 132.Joung E.-J., Li M.-H., Lee H. G., et al. Capsaicin induces heme oxygenase-1 expression in HepG2 cells via activation of PI3K-Nrf2 signaling: NAD(P)H:quinone oxidoreductase as a potential target. Antioxidants and Redox Signaling. 2007;9(12):2087–2098. doi: 10.1089/ars.2007.1827. [DOI] [PubMed] [Google Scholar]
  • 133.Chen C.-C., Chen H.-L., Hsieh C.-W., Yang Y.-L., Wung B.-S. Upregulation of NF-E2-related factor-2-dependent glutathione by carnosol provokes a cytoprotective response and enhances cell survival. Acta Pharmacologica Sinica. 2011;32(1):62–69. doi: 10.1038/aps.2010.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Jun Y.-J., Lee M., Shin T., Yoon N., Kim J.-H., Kim H.-R. Eckol enhances heme oxygenase-1 expression through activation of Nrf2/JNK pathway in HepG2 cells. Molecules. 2014;19(10):15638–15652. doi: 10.3390/molecules191015638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Granado-Serrano A. B., Martín M. A., Haegeman G., Goya L., Bravo L., Ramos S. Epicatechin induces NF-κB, activator protein-1 (AP-1) and nuclear transcription factor erythroid 2p45-related factor-2 (Nrf2) via phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) and extracellular regulated kinase (ERK) signalling in HepG2 cells. British Journal of Nutrition. 2010;103(2):168–179. doi: 10.1017/S0007114509991747. [DOI] [PubMed] [Google Scholar]
  • 136.Krajka-Kuźniak V., Paluszczak J., Celewicz L., Barciszewski J., Baer-Dubowska W. Phloretamide, an apple phenolic compound, activates the Nrf2/ARE pathway in human hepatocytes. Food and Chemical Toxicology. 2013;51(1):202–209. doi: 10.1016/j.fct.2012.09.033. [DOI] [PubMed] [Google Scholar]
  • 137.Tanigawa S., Fujii M., Hou D.-X. Action of Nrf2 and Keap1 in ARE-mediated NQO1 expression by quercetin. Free Radical Biology and Medicine. 2007;42(11):1690–1703. doi: 10.1016/j.freeradbiomed.2007.02.017. [DOI] [PubMed] [Google Scholar]
  • 138.Krajka-Kuźniak V., Paluszczak J., Baer-Dubowska W. Xanthohumol induces phase II enzymes via Nrf2 in human hepatocytes in vitro. Toxicology in Vitro. 2013;27(1):149–156. doi: 10.1016/j.tiv.2012.10.008. [DOI] [PubMed] [Google Scholar]
  • 139.Yap W. H., Khoo K. S., Ho A. S. H., Lim Y. M. Maslinic acid induces HO-1 and NOQ1 expression via activation of Nrf2 transcription factor. Biomedicine and Preventive Nutrition. 2012;2(1):51–58. doi: 10.1016/j.bionut.2011.12.005. [DOI] [Google Scholar]
  • 140.Chitturi S., Farrell G. C. Herbal hepatotoxicity: an expanding but poorly defined problem. Journal of Gastroenterology and Hepatology. 2000;15(10):1093–1099. doi: 10.1046/j.1440-1746.2000.02349.x. [DOI] [PubMed] [Google Scholar]
  • 141.Mennen L. I., Walker R., Bennetau-Pelissero C., Scalbert A. Risks and safety of polyphenol consumption. The American Journal of Clinical Nutrition. 2005;81(1, supplement):326S–329S. doi: 10.1093/ajcn/81.1.326S. [DOI] [PubMed] [Google Scholar]
  • 142.Skibola C. F., Smith M. T. Potential health impacts of excessive flavonoid intake. Free Radical Biology and Medicine. 2000;29(3-4):375–383. doi: 10.1016/S0891-5849(00)00304-X. [DOI] [PubMed] [Google Scholar]
  • 143.Liu J., Lu Y.-F., Zhang Y., Wu K. C., Fan F., Klaassen C. D. Oleanolic acid alters bile acid metabolism and produces cholestatic liver injury in mice. Toxicology and Applied Pharmacology. 2013;272(3):816–824. doi: 10.1016/j.taap.2013.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Frenzel C., Teschke R. Herbal hepatotoxicity: clinical characteristics and listing compilation. International Journal of Molecular Sciences. 2016;17(5):p. 588. doi: 10.3390/ijms17050588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bunchorntavakul C., Reddy K. R. Review article: herbal and dietary supplement hepatotoxicity. Alimentary Pharmacology and Therapeutics. 2013;37(1):3–17. doi: 10.1111/apt.12109. [DOI] [PubMed] [Google Scholar]
  • 146.Muqeet Adnan M., Khan M., Hashmi S., Hamza M., AbdulMujeeb S., Amer S. Black cohosh and liver toxicity: is there a relationship? Case Reports in Gastrointestinal Medicine. 2014;2014:3. doi: 10.1155/2014/860614.860614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lim T. Y., Considine A., Quaglia A., Shawcross D. L. Subacute liver failure secondary to black cohosh leading to liver transplantation. BMJ Case Reports. 2013;2013 doi: 10.1136/bcr-2013-009325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Teschke R., Schwarzenboeck A. Suspected hepatotoxicity by Cimicifugae racemosae rhizoma (black cohosh, root): critical analysis and structured causality assessment. Phytomedicine. 2009;16(1):72–84. doi: 10.1016/j.phymed.2008.09.009. [DOI] [PubMed] [Google Scholar]
  • 149.Lynch C. R., Folkers M. E., Hutson W. R. Fulminant hepatic failure associated with the use of black cohosh: a case report. Liver Transplantation. 2006;12(6):989–992. doi: 10.1002/lt.20778. [DOI] [PubMed] [Google Scholar]

Articles from Oxidative Medicine and Cellular Longevity are provided here courtesy of Wiley

RESOURCES