Next Article in Journal
Comparison of Simple Eudragit Microparticles Loaded with Prednisolone and Eudragit-Coated Chitosan-Succinyl-Prednisolone Conjugate Microparticles: Part II. In Vivo Evaluation of Efficacy, Toxicity, and Biodisposition Characteristics
Next Article in Special Issue
Current Status of Herbal Medicines in Chronic Liver Disease Therapy: The Biological Effects, Molecular Targets and Future Prospects
Previous Article in Journal
Application of CRISPR/Cas9 Technology to HBV
Previous Article in Special Issue
The Role of HMGB1 Signaling Pathway in the Development and Progression of Hepatocellular Carcinoma: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Oxidative Stress and Antioxidants in Liver Diseases

School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2015, 16(11), 26087-26124; https://doi.org/10.3390/ijms161125942
Submission received: 12 August 2015 / Revised: 20 September 2015 / Accepted: 19 October 2015 / Published: 2 November 2015
(This article belongs to the Special Issue Molecular Mechanisms of Human Liver Diseases)

Abstract

:
A complex antioxidant system has been developed in mammals to relieve oxidative stress. However, excessive reactive species derived from oxygen and nitrogen may still lead to oxidative damage to tissue and organs. Oxidative stress has been considered as a conjoint pathological mechanism, and it contributes to initiation and progression of liver injury. A lot of risk factors, including alcohol, drugs, environmental pollutants and irradiation, may induce oxidative stress in liver, which in turn results in severe liver diseases, such as alcoholic liver disease and non-alcoholic steatohepatitis. Application of antioxidants signifies a rational curative strategy to prevent and cure liver diseases involving oxidative stress. Although conclusions drawn from clinical studies remain uncertain, animal studies have revealed the promising in vivo therapeutic effect of antioxidants on liver diseases. Natural antioxidants contained in edible or medicinal plants often possess strong antioxidant and free radical scavenging abilities as well as anti-inflammatory action, which are also supposed to be the basis of other bioactivities and health benefits. In this review, PubMed was extensively searched for literature research. The keywords for searching oxidative stress were free radicals, reactive oxygen, nitrogen species, anti-oxidative therapy, Chinese medicines, natural products, antioxidants and liver diseases. The literature, including ours, with studies on oxidative stress and anti-oxidative therapy in liver diseases were the focus. Various factors that cause oxidative stress in liver and effects of antioxidants in the prevention and treatment of liver diseases were summarized, questioned, and discussed.

Graphical Abstract

1. Introduction

Free radicals are atoms or molecules that have unpaired electrons, usually unstable and highly reactive [1]. In biology system, oxygen based radicals and nitrogen based radicals are two types of free radicals. Oxygen free radicals, such as superoxide, hydroxyl radicals, and peroxyl radicals, with the addition of non-radicals, such as hydrogen peroxide, hypochlorous acid and ozone, are known as reactive oxygen species (ROS), which are generated during the metabolism process of oxygen. Reactive nitrogen species (RNS), including nitrogen based radicals and non-radicals, such as nitrogen dioxide, nitric oxide radicals and peroxynitrite, are derived from nitric oxide and superoxide via inducible nitric oxide synthase (iNOS) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, respectively [2,3]. Due to their special chemical characteristics, ROS/RNS can initiate lipid peroxidation, cause DNA strand breaks, and indiscriminately oxidize virtually all molecules in biological membranes and tissues, resulting in injury. However, since the body is able to remove ROS/RNS to a certain degree, these reactive species are not necessarily a threat to the body under physiological conditions [3,4]. As a matter of fact, ROS are required at certain level in the body to perform its important physiological functions. The generation of ROS is a natural part of aerobic life, which is responsible for the manifestation of cellular functions including signal transduction pathways, defense against invading microorganisms and gene expression to the promotion of growth or death [1]. Oxidative/nitrosative stress represents the bodies’ imbalance in the production and the elimination of reactive oxygen and nitrogen species as well as decreased production of antioxidants. In terms of oxidative stress, in specific physiological conditions, it is actually useful. For example, it could strengthen biological defense mechanisms during appropriate physical exercise and ischemia, and induce apoptosis to prepare the birth canal for delivery [2,3]. However, this is confined to particular situations, and in most other cases, large levels of ROS and oxidative stress will induce cell death through necrotic and/or apoptotic mechanisms, leading to cellular and tissue injury.
Liver is a major organ attacked by ROS [5]. Parenchymal cells are primary cells subjected to oxidative stress induced injury in the liver. The mitochondrion, microsomes and peroxisomes in parenchymal cells can produce ROS, regulating on PPARα, which is mainly related to the liver fatty acid oxidation gene expression. Moreover, Kupffer cells, hepatic stellate cells and endothelial cells are potentially more exposed or sensitive to oxidative stress-related molecules. A variety of cytokines like TNF-α can be produced in Kupffer cells induced by oxidative stress, which might increase inflammation and apoptosis. With regard to hepatic stellate cells, the proliferation and collagen synthesis of hepatic stellate cells is triggered by lipid peroxidation caused by oxidative stress [6,7,8]. In mammals, a sophisticated antioxidant system has been developed to maintain the redox homeostasis in the liver (Figure 1). When the ROS is excessive, the homeostasis will be disturbed, resulting in oxidative stress, which plays a critical role in liver diseases and other chronic and degenerative disorders [9]. The oxidative stress not only triggers hepatic damage by inducing irretrievable alteration of lipids, proteins and DNA contents and more importantly, modulating pathways that control normal biological functions. Since these pathways regulate genes transcription, protein expression, cell apoptosis, and hepatic stellate cell activation; oxidative stress is regarded as one of the pathological mechanisms that results in initiation and progression of various liver diseases, such as chronic viral hepatitis, alcoholic liver diseases and non-alcoholic steatohepatitis [10,11]. It has also been suggested that there are complicated cross-talks among pathological factors, inflammation, free radicals and immune responses [11,12]. The general mechanism scheme of oxidative stress induced by various factors on liver disease is concluded in Figure 2. Moreover, systemic oxidative stress arising during liver disease can also cause damage to extra-hepatic organs, such as brain impairment and kidney failure [13]. It was suggested systemic oxidative stress might be a significant “first hit”, acting synergistically with ammonia to induce brain edema in chronic liver failure [14]. With regard to kidney failure, systemic oxidative stress is considered to play a critical role in the pathophysiology of several kidney diseases [15,16].
Figure 1. The redox homeostasis in the liver.
Figure 1. The redox homeostasis in the liver.
Ijms 16 25942 g001
Figure 2. The general mechanism scheme of oxidative stress induced by various factors on liver disease.
Figure 2. The general mechanism scheme of oxidative stress induced by various factors on liver disease.
Ijms 16 25942 g002
Both enzymatic and non-enzymatic antioxidant system are essential for cellular response in order to deal with oxidative stress under physiological condition. Therefore, antioxidant enzyme such as CAT, SOD, and GSH-Px and non-enzymatic electron receptors such as GSH are affected and used as indexes to evaluate the level of oxidative stress [12,17,18,19]. Notably, erythroid 2-related factor 2 (Nrf2) is a major regulator of cellular redox balance [20]. Under physiological condition, Nrf2 binds to kelch-like ECH-associated protein-1 (Keap1) in the cytoplasm, and the ones remaining are inactivated and easily to be degraded. Under oxidative stress, however, Nrf2 dissociates form Keap1 by Keap1 modification or Nrf2 phosphorylation and are thus activated. The activated Nrf2 translocates into the nucleus and interacts with antioxidant response element (ARE), promoting the expression of cytoprotective target genes including antioxidant enzymes and phase II detoxifying enzymes [21]. The enhanced activation of Nrf2 by pharmacologic molecules or genetic engineering has been shown to protect the liver in different oxidative stress models [22]. For example, in terms of pharmacologic activation of Nrf2, the use of small molecules, such as BHA, oleanolic acid, ursolic acid and CDDO-Im have been reported to show hepatoprotection against liver damage induced by acetaminophen, a famous drug possessing hepatotoxicity. During the process where mitochondria convert acetate into ATP, a significant amount of free radicals are generated, which results in cellular injuries, especially to mitochondria themselves. Activation of Nrf2 protects mitochondria from oxidative stress via a variety of mechanisms depending on different circumstances, such as increasing antioxidant levels, protecting against mitochondrial permeability transition pore opening, maintaining the mitochondrial redox state, enhancing mitochondrial biogenesis by promoting transcription of nuclear respiratory factor 1 (Nrf1). For fatty liver disease, activation of Nrf2 could facilitate fatty acid metabolism in liver by directly regulating fatty acid metabolism related genes, such as CD36 [20,22]. Furthermore, the enhanced antioxidant signaling regulated by activated Nrf2 protects mitochondria from oxidative damages, which further ensures competent hepatic fatty acid catabolism.
Regarding the vital role of oxidative stress in chain of liver diseases, various anti-oxidative therapy and antioxidants are proposed to prevent and treat liver diseases [9,12]. A series of studies have tested the effectiveness of some antioxidants in the treatment of patients with various liver diseases, such as chronic hepatitis C virus infection, alcoholic hepatitis or cirrhosis, and non-alcoholic fatty liver disease (NAFLD). The clinical effects of antioxidants as adjuvants including vitamin E/C, mitoquinone, N-acetylcysteine, polaprezinc silymarin, silibinin and some antioxidant cocktail on chronic hepatitis C patients have been examined has shown clear benefit of antioxidants to interferon based therapy of HCV [23,24]. However, despite some positive results were obtained, it cannot reach to the conclusion that antioxidants are useful therapeutic agents for chronic hepatitis C partly due to the sample scale and treatment duration. Vitamins E/C, N-acetylcysteine, polyenylphosphatidylcholine, silymarin, and antioxidants cocktail have been attempted for the treatment of alcoholic hepatitis or cirrhosis patients [24,25,26]. Although some promise has been shown, results indicated that many antioxidants failed to improve the outcome of patients [27]. Additionally, a great deal of studies has investigated the therapeutic effects of vitamins E/C and N-acetylcysteine on NAFLD. It is worth noting that vitamin E has been demonstrated clinically to be a rather promising drug for the treatment of non-alcoholic steatohepatitis [28,29]. Although data from clinical studies is yet to prove the efficacy of antioxidant, application of antioxidants is a rational curative strategy for prevention and treatment of liver diseases involving oxidative stress [17,30]. Natural antioxidants have been found in many edible (such as fruits, vegetables, cereals and tea) and medicinal plants, which often possess strong antioxidant and free radical scavenging abilities as well as anti-inflammatory action [9]. Several well-elaborated reviews concerning antioxidants as therapeutic agents for diverse liver diseases in clinic have been published [11,31,32], therefore, in this review, particular attention will be drawn on the factors causing oxidative stress in liver and in vivo effects of antioxidants for the prevention and treatment of liver diseases. Moreover, although oxidative stress has been suggested to exist in almost all liver diseases, since the fact that there are no animal models with virus-induced liver disease, including hepatitis A, hepatitis B, and hepatitis C, the role of oxidative stress in viral hepatitis are not included in this review.

2. Oxidative Stress in Liver Diseases

2.1. Oxidative Stress Caused by Alcohol

Alcohol beverages are widely consumed all over the world; however, excessive alcohol consumption may cause a series of health problems. It was reported that alcohol consumption accounting for an estimated 3.8% of global mortality. Alcoholic liver disease (ALD) is one of the most important causes of liver-related death, which is associated with increased dose and time of alcohol intake. In 2003, it has been reported that age- and sex-adjusted mortality rate of ALD was 4.4/100,000. Although reductions in overall ALD mortality were observed in several reports on a country scale, it is more likely due to advances in disease management rather than a decrease in the prevalence of ALD, which could be supported by increases in hospital admissions for alcoholic hepatic failure and alcoholic hepatitis [33,34,35]. ALD may progress from steatosis to more severe liver diseases form, such as hepatitis, fibrosis, and cirrhosis [36,37]. As a matter of fact, more than 90% heavy drinkers develops fatty liver, and about 30% of heavy drinkers further develops advance forms of ALD. Although pathogenesis of ALD has not been fully elaborated, the direct consequence of ethanol metabolism seems to be related to ROS production, mitochondrial injury and steatosis, which are the common features of acute and chronic alcohol exposure [32,38,39]. It is well illustrated that at least three distinct enzymatic pathways are involved in the process of ethanol oxidation [15]. The primary pathway for the ethanol metabolism is dehydrogenase system. It is initiated by alcohol dehydrogenase (ADH), a NAD+-requiring enzyme expressed at high levels in hepatocytes, which oxidizes ethanol to acetaldehyde. Then, acetaldehyde enters the mitochondria where it is oxidized to acetate by aldehyde dehydrogenases (ALDH). The second major pathway to oxidize ethanol is the microsomal ethanol oxidizing system (MEOS), which involves an NADPH-requiring enzyme, the cytochrome P450 enzyme CYP2E1. The MEOS pathway is prompted in individuals who consume alcohol chronically. In addition, infrequently, ethanol can also be oxidized by catalase in peroxisomes. Since this oxidation pathway requires the presence hydrogen peroxide (H2O2), under normal conditions, this pathway plays no major role in alcohol metabolism [15,16,17]. During the metabolism processes via dehydrogenase system and MEOS system, NADH or NADP+ will be produced in bulk, leading to the increase of ROS, which cause oxidative stress resulting in hepatocyte injury, and finally trigger various liver diseases (Figure 3).
Figure 3. The metabolic process of ethanol in hepatocyte and the generation of ROS contributing to the liver diseases.
Figure 3. The metabolic process of ethanol in hepatocyte and the generation of ROS contributing to the liver diseases.
Ijms 16 25942 g003
Studies have demonstrated that enzymatic as well as non-enzymatic systems which maintaining cellular homeostasis are remarkably affected by alcohol in diverse models. In particular, the activities of SOD, CAT, GSH-Px, GRD, and GST, as well as the level of lipid peroxidation were changed in animals treated with alcohol [19,40,41,42]. For example, SOD and CAT activities were decreased and the lipid peroxidation level was significantly increased in the liver of 30 days alcohol-treated diabetic rats [40]. An increase of lipid peroxidation and hepatic cytochrome P450, and decrease of hepatic SOD, GSH-Px, GRD, GST, and GSH were also observed in mice treated with dimethoate in combination with ethanol [41]. Furthermore, oxidative stress and antioxidant enzyme were measured in patients with ALD [32]. It was found that as the severity of the disease increased, followed by elevation of serum level of lipid peroxidation indicator malondialdehyde (MDA) and the concentrations of serum vitamins E and C, which act as indexes of antioxidant status, were decreased in ALD patients. The pro-oxidant and antioxidant status in chronic alcoholics have been detected in several studies. The significant decreases of GSH levels in liver and blood of patients with alcoholic liver disease were observed when compared to controls. However, the activity/content of SOD and CAT after alcohol exposure are rather controversial, with reports of increases, no changes, or decreases, depending on the amount and time of alcohol consumption [43,44]. Nevertheless, the increased oxidative stress in patients with ALD has been demonstrated. It was argued that the increases of antioxidants enzymes such as SOD, CAT and GSH-Px might be a compensatory regulatory response to increased oxidative stress [45]. The level of ALT was increased significantly while the level of AST was decreased significantly in patients with ALD [32,46,47].

2.2. Oxidative Stress Caused by Drugs

The liver is the most frequently targeted organ in terms of drug toxicity. The production of radical species, specifically ROS and RNS, has been proposed as an early event of drugs hepatotoxicity and as an indicator of hepatotoxic potential [48]. It has been discovered that a lot of drugs could induce oxidative stress including increase of cellular oxidants and lipid peroxidation, depletion of antioxidants in the liver, such as anti-inflammation drugs, anti-analgesic drugs, anti-cancer drugs and antidepressants. For example, sulfasalazine, a drug to treat inflammatory bowel diseases, has been found to induce hepatic oxidative damage [49]. Oral sulfasalazine administration could reduce SOD but increase CAT activity significantly. It is also suggested that oxidative damage is involved in hepatotoxicity of sulfasalazine treatment. As for zoledronic acid, it is a nitrogen-bearing bisphosphonate, and used to treat the cancer-associated hypercalcemia. It has been shown that zoledronic acid significantly elevated MDA and nitric oxide levels, whereas reduced GSH levels, which indicated that zoledronic acid could induce oxidative stress and decrease antioxidant level in liver [18]. Furthermore, liver antioxidant capacity in hepatic injury induced by paracetamol, an extensively used analgesic compound in mice was evaluated [50]. It was shown that paracetamol induced a remarkable increase of MDA and nitrite as well as nitrate in the liver, with potent decrease of total SOD and Cu/Zn-SOD activity. Samarghandian et al. [51] studied effect of long-term treatment of morphine on enzymes, oxidative stress indices and antioxidant status in male rat liver. The results showed that the levels of ALT, AST and lactate dehydrogenase (LDH) in serum as well as MDA in liver were significantly elicited, while the activities of SOD, glutathione-s-transfrase and CAT were remarkably reduced by morphine. Oxidative stress generated by anticancer drugs including doxorubicin, paclitaxel and docetaxel in the liver of rats have been indicated. It was found that all three drugs increased thiobarbituric acid-reactive substances (TBARS), and the administration of docetaxel significantly decreased the activity of SOD. Furthermore, combined administration of two drugs generated greater changes in oxidative stress related molecules than single agents [52]. Nimesulide, nonsteroidal anti-inflammatory drug, could increase the activities of ALT, AST, ALP and the content of bilirubin in the serum. The activities of SOD and CAT and GSH-Px in the liver were decreased by nimesulide in mice [53]. Chronic administration of fluoxetine (15 mg/kg/day) or clozapine (20 mg/kg/day) was measured in rats exposed to chronic social isolation and controls. The increased serum ALT activity, MDA, decreased GSH levels and compromised SOD expression suggests a link between drugs and hepatic oxidative stress [54]. Anti-tuberculosis agent isoniazid (INH) resulted in both oxidative and nitrosative stress, but the correlation of hepatotoxicity severity with RNS rather than ROS suggested that ONOO generation and mitochondrial dysfunction are responsible mechanisms for hepatotoxicity of INH in vivo [55,56].
Although hepatotoxicity induced by various drugs in humans has been demonstrated in a great number of clinical trials, report concerning the role of oxidative stress in patients with drug induced liver disease is limited by far. For example, mitochondrial dysfunction and DNA damage are found to be critical events in the underling mechanism of paracetamol induced hepatotoxicity in patients, which is supposed to partly attribute to oxidative stress, but, accurate and direct evidence to show the status and role oxidative stress in patients is lacking [57]. As a matter of fact, currently, in addition to animal model study, the investigation of hepatotoxicity induced by drugs is mainly based on the results of retrospective study, whereas there are few clinical studies with large numbers of patients. Moreover, models using human cells have been attempted to mimic pathogenesis of drug induced hepatotoxicity in humans [55]. Overall, clinical data and appropriate experimental model, which could closely resemble the human pathophysiology, is critical for future study of antioxidant treatment for hepato-toxicity caused by drugs.

2.3. Oxidative Stress Caused by Environmental Pollutants

Environmental pollutants such as heavy metals and microcystin have been shown to cause oxidative damage in liver of animal models. Antioxidant defense system in rat liver was damaged after mercury chloride treatment [58]. Mercury chloride at the dose of 0.1 mg/kg could induce a significant decrease in both Mn-dependent SOD and Cu- and Zn-dependent SOD activities, and progressive changes of CAT, GSH-Px, GRD and glucose-6-phosphate dehydrogenase activities. This is also accompanied by a minor increase in serum ALT and γ glutamyltransferase. The results showed that low dose of mercury could incur oxidative stress and hepatic damage. Besides mercury, lead was also found to exacerbate liver lipid peroxidation in protein-undernutrited rats, in which the study also suggested that free radicals is a pathological mechanism for hepatotoxicity of lead [59]. Microcystins are algae toxins produced by cyanobacteria, kind of cyclic nonribosomal peptides, possessing hepatotoxicity that may cause severe injury to the liver. The effect of microcystin LR, the most studied toxic variants, on antioxidant enzymes and lipid peroxidation was investigated in liver rats after acute exposure [60]. The reduction of enzymes activities of GSH-Px, GRD, SOD and CAT as well as significant increase of lipid peroxidation levels were observed in the liver of microcystin LR-treated rat. These results showed that acute exposure of microcystin LR could result in perturbation of the antioxidant enzymes, suggesting the involvement of oxidative stress in the pathogenesis of microcystin LR-induced toxicity.

2.4. Oxidative Stress Caused by Other Factors

Other factors such as radiation and temperature may also induce hepatic oxidative stress. The oxidative stress induced through exposure of mobile phone-like radiation has been investigated in the liver of guinea pigs [61]. The results showed that after radiation exposure, the levels of MDA and total nitric oxide were significantly increased and the activities of SOD, myeloperoxidase and GSH-Px were reduced in the liver of guinea pigs. Additionally, the severity of oxidative damage was increased along with the duration of radiation exposure. The results suggested that mobile phone-like radiofrequency radiation could induce oxidative damage in liver, implying the adverse effect of mobile phone use. Moreover, study observed that cold stress could lead to decrease in CAT, SOD and GSH-Px activities in rat liver when the rats were kept at 10 °C for a week, which indicated that cold stress may cause hepatic damage which is associated with oxidative stress [62].
Benzoyl peroxide is a substance with strong oxidizing capacity, and broadly used as flour bleaching agent. The hepatic antioxidant status and ATPases were affected by benzoyl peroxide in mice [63]. Following benzoyl peroxide exposure, SOD activity was reduced significantly, whereas the content of MDA was increased in liver tissue. The activities of Ca2+-ATPase and Mg2+-ATPase in liver were also significantly decreased by benzoyl peroxide. In another study, the effect of ZnO2 nanoparticles, a common cosmetic component, on cellular oxidative stress in mouse liver was investigated [64]. After exposure to ZnO2 nanoparticles, viability of hepatic cells was decreased in concentration-dependent manner, and decrease in antioxidant enzyme levels as well as increase in DNA adduct.
Studies have suggested that maternal high-fat diet feeding could raise the incidence of metabolism-related diseases in offspring, including chronic liver disease. Zhang et al. [65] found that maternal high-fat diet increased the level of plasma triglyceride and hepatic TBARS significantly. The size of lipid droplets in the liver of rat offspring was also increased. Expression of antioxidant defense genes, such as GSH-Px-1, Cu/Zn-SOD, and paraoxonase enzymes, were significantly lowered in the liver. Up-regulation of the inhibitor of cyclooxygenase-2 and cyclin dependent kinase 4a, and down-regulation of cyclin D1 and phosphorylation of retinoblastoma protein were found in the offspring. These results suggested that maternal high-fat diet might reduce the capacity of antioxidant defense and speed up cellular senescence in hepatic tissue of older offspring. In another study, the effect of high dietary salt on hepatic antioxidant defensing enzyme of fructose-fed rats was investigated [66]. Feeding fructose-fed rats with high-salt diet could trigger hyperinsulinemia and insulin resistance resulting in membrane perturbation. This potentially enhanced hepatic lipid peroxidation in the presence of steatosis, and led to decrease in antioxidant defenses, as observed by reduction of GSH, SOD and CAT activities. These results indicated that consumption of salt-rich diet by insulin-resistant subjects could lead to sodium reabsorption, which may aggravate hepatic lipid peroxidation related to damage antioxidant defenses.
In addition to those liver injury induced by exogenous substances, hepatic oxidative stress has been revealed in other liver diseases and functional disorders. For instance, Messarah et al. [67] has found that thyroid dysfunction would increase lipid peroxidation and oxidative stress status in rat liver. In another study, oxidative stress and antioxidant status in patients with autoimmune cholestatic liver diseases (AC) or autoimmune hepatitis (AIH) were investigated [68]. Several markers of oxidative injury and antioxidant components in whole blood, serum, and urine of 49 patients with AC and 36 patients with AIH as well as healthy subjects were assessed. The results showed that both AC and AIH patients had increased levels in oxidation products of lipid and protein while significant decreased of whole blood GSH level. Protein carbonyl and isoprostane levels were increased and GSH level was gradually decreased with disease severity level (mild to severe fibrosis and cirrhosis) in both AC and AIH patients. In addition, AIH patients had higher levels of aldehydes and GSH-Px activity and lower protein carbonyl levels compared to AC patients. In patients with nonalcoholic fatty liver disease (NAFLD), the oxidative stress and antioxidant status were changed as well [69]. It was shown that level of TBARS in NAFLD patients was significantly higher than subjects with viral hepatitis or healthy controls. Moreover, the ferric reducing ability of plasma in patients with NAFLD was significantly higher than healthy controls, and diseased control group of patients. These results implied that lipid peroxidation and oxidative stress were significantly increased in patients with NAFLD. Although existence of hepatic oxidative stress in various liver diseases was commonly observed, the relationship between oxidative damage and diseases are causal and not strictly defined.

3. Antioxidants for Prevention and Treatment of Liver Diseases

3.1. Antioxidants for Prevention and Treatment of Alcoholic Liver Diseases

An obvious avenue of alcoholic liver diseases (ALD) prevention would be abstinence; however, abstinence is not easy to maintain due to the high rate of recidivism in alcoholics [14]. As mentioned above, ALD develops from simple steatosis to more severe disease forms including hepatitis, fibrosis, cirrhosis, and even hepatocellular carcinoma, which implies that preventing disease development at the early stage would be more effective than receiving treatment at end-stage of liver disease. Notably, TNF, a group of cytotoxic pro-inflammatory cytokines, is thought to play a vital role in initiation of liver damage [70]. Increasing evidence has indicated that oxidative stress might act together with endotoxins to increase TNF production. Increased circulating TNF-α stimulates TNF-α receptors of cell surface, which leads to activation of the stress-related protein kinases JNK and IKKβ, resulting in increased production of additional inflammatory cytokines, and reduced insulin sensitivity. Consequently, the inhibition of TNF is regarded as a therapy to block fatty liver and relieve liver injury [70,71]. Pharmacological and genetic manipulation of TNF have been attempted to treat liver disease. For example, anti-TNF antibodies or knocking out TNF-R1 have been treated to mice to protect against the development of ALD. However, since liver regeneration requires low “basal” contents of TNF, down regulating but not blocking totally TNF activity is a preferred therapeutic intervention for liver disease [71,72]. With better understanding of the mechanism that regulates the initiation and advancement of ALD, antioxidant therapy could be developed as directed therapy to prevent or treat ALD [32,37,73,74]. It has been demonstrated that many food and plants, such as vegetables, fruits, tea, cereals, medicinal plants, microalgae, edible macro-fungi, and wild flowers, have abundant natural antioxidants, and possess the ability of eliminating free radicals and protecting the liver from oxidative stress [75,76,77,78,79,80,81,82,83], and thus might be beneficial for liver diseases.
In recent years, a great number of natural plants has been attempted to eliminate hepatic damage induced by ethanol in animal models, and the bioactive compounds that are responsible for relieving oxidative stress are usually indistinctly ascribed to polyphenols and flavonoids compounds [42,84,85,86,87]. For example, it has been found that green tea, containing abundant water-soluble antioxidants, showed positive effect on the antioxidant abilities in rat liver with chronic ethanol treatment [84]. It was shown that significant reduction of enzymatic and non-enzymatic antioxidants levels, as well as increased levels of lipid and protein modifications was induced by ethanol diet. After administration of green tea, interestingly, the enzymes activity and level of non-enzymatic antioxidants as well as lipid and protein oxidation products were partly normalized. The effects of some natural products on hepatic alcoholic damage associated with oxidative stress were summarized in Table 1, which indicate that anti-oxidative treatment is an encouraging method to reduce alcoholic liver injury. Besides phenolic compounds, more specific bioactive compounds should be further identified and isolated in the future.
Table 1. The effects of antioxidants/plants on alcoholic liver damage. Up-arrow means increase and up-regulation, and down-arrows means decrease and down-regulation.
Table 1. The effects of antioxidants/plants on alcoholic liver damage. Up-arrow means increase and up-regulation, and down-arrows means decrease and down-regulation.
Models (Prevent/Treatemnt)MaterialsEffectDose (Dose-Effect)Bioactive CompoundsReferences
Rats treated with ethanol diet (Prevent)Green tea↑ Enzymes, non-enzymatic antioxidants; ↓ lipid and protein oxidation7 g/L in ethanol Lieber-DeCarli dietEpicatechin, epicatechin gallate[84]
Rats treated with ethanol (Prevent)Ziziphus mauritiana leaf↓ ALT, AST, ALP, total bilirubin, CAT; ↑ GSH-Px, glutathione reductase and SOD200 and 400 mg/kg b.w. (Dose-effect)Tannins, saponins and phenolic compounds[42]
Rats sub-chronically exposed to ethanol (Prevent)Amaranthus hypochondriacus seed↓ MDA, NADPH; ↑ Cu, Zn-SOD140 g/kg in dietTotal phenols[87]
Mice with acute alcohol-induced liver injury (Prevent)Peduncles of Hoveniadulcis↓ ALT, AST, MDA; ↑ SOD, GSH-Px100, 350 and 600 mg/kg b.w. (Dose-effect)Non-starch polysaccharide[86]
Rats treated with ehanol (Prevent)Methanolic extract from Hammada scoparia leaves↓ Aminotransferase, glycogen synthase kinase-3 β, lipid peroxidation; ↑ GSH-Px200 mg/kg b.w.Phenolic compounds[85]
Mice with chronic alcoholic liver damage (Prevent)Jujube honey↓ Lipoprotein oxidation, AST, ALT, MAD, 8-hydroxy-2-deoxyguanosine; ↑ GSH-Px27 and 54 g /kg b.w. (Dose-effect)Phenolic acids[88]
Mice with alcohol-induced hepatotoxicity (Treatment)Freeze-dried, germinated and fermented mung bean↑ Antioxidant levels, NO200 and 1000 mg/kg b.w. [89]
Chronic ethanol exposure in rats (Prevent)Virgin olive oil↓ Transaminases levels, hepatic lipid peroxidation; ↑ GSH-Px, SOD and CAT5% (wt/wt) in dietTocopherols, chlorophyll, total polyphenols[90]
In addition to these natural products, many single compounds have been investigated for their role in eliminating oxidative stress, such as l-theanine, vitamin E, N-acetyl cysteine, raxofelast and betaine [91]. l-theanine, a unique amino acid in green tea, has been proven to possess the ability to prevent alcoholic hepatic damage via augmenting antioxidant capacities [91]. The ethanol-stimulated increase of ALT, AST, and MDA and reduction of antioxidant enzymes activities including the activities of SOD, and CAT, as well as level of GSH were significantly inhibited by l-theanine. The regulation of l-theanine on alcohol-induced fat droplets was further confirmed by histopathological examination. Besides, vitamin E is considered to be beneficial for prevention of diseases associated with oxidative stress because of its remarkable anti-oxidative properties. Kaur et al. [92] has proven that vitamin E could restore the redox status, prevent oxidative stress and reduce apoptosis, and could be used as a prospective curative agent for ethanol-induced hepatic oxidative injury. Moreover, raxofelast, an analog of vitamin E, possesses the ability to inhibit lipid peroxidation in mice exposed to ethanol [93]. Raxofelast diminished the increased hepatic NF-κB activity, reduced serum ALT and liver triglycerides, lowered hepatic MAD levels, prevented liver GSH depletion, decreased Toll-like receptor-4, TNF-α, IL-6 and intercellular adhesion molecule-1 hepatic gene expression. It has been suggested that raxofelast blunted the inflammatory cascade and liver damage during chronic ethanol exposure. N-acetyl cysteine, a scavenger of ROS, may reverse alcoholic liver damage, and alter activities of matrix metalloproteinases [94]. Furthermore, it was shown that the ethanol-induced oxidative stress could be inhibited effectively by betaine, which is also responsible to its hepatoprotection [95].
Betulinic acid is a pentacycliclupane-type triterpene, and has a wide range of bioactivities. Yi et al. [96] has reported that pre-treatment of betulinic acid could significantly reduce the serum levels of ALT, AST, total cholesterol, and triacylglycerides in the mice treated with alcohol. Hepatic levels of GSH, SOD, GSH-Px, and CAT were remarkably increased, while MDA contents and microvesicular steatosis in the liver were decreased by betulinic acid. It was suggested that the hepatoprotective effect of betulinic acid is associated with the improvement of antioxidant enzymes capacity, primarily via enhancement of the tissue redox system and protection of the antioxidant system in the liver. Demethyleneberberine, a natural mitochondria-targeted antioxidant found in Chinese herb Cortex Phellodendri chinensis, has been demonstrated the ability of inhibiting oxidative stress and steatosis in acutely/chronically ethanol-fed mice [97].

3.2. Antioxidants for Prevention and Treatment of Non-Alcoholic Fatty Liver Diseases

NAFLD is characterized by abnormal fatty acids deposition in the liver cells of patients without excessive alcohol intake, viral infection or other hepatoxins, including a broad spectrum of histological irregularities [98]. Notably, obesity is considered to be the main risk factor for the development of NAFLD and the main driver of rapid rise of NAFLD prevalence [99]. The oxidative stress of endoplasmic reticulum induced by free fatty acid in the liver might contribute to the hepatic injury, progressive fibrosis and even cirrhosis [100]. In Table 2, certain antioxidants or plants were attempted to reduce liver injury induced by high fat diet in experimental animals, which indicated that most of them showed both antioxidant and hepato-protective effects. Furthermore, in a clinical trial that aims to systematically evaluate the effect of antioxidant supplements, it was found that AST levels, but not of ALT levels were reduced significantly in patients with NAFLD by antioxidant intervention. It should be pointed out that, however, data obtained is so far insufficient to figure out whether dietary supplements is beneficial or useless for patients with NAFLD [98]. To address this issue, large-scaled of prospective randomized clinical studies on this topic is quite necessary.
It has also been indicated that insulin resistance, oxidative stress, and the inflammatory cascade play a vital role in the pathogenesis of NAFLD by animal study. Data from clinic trial indicated that insulin resistance is a high risk factor of NAFLD. Recent studies have shown that insulin resistance is present in surrounding tissue and live of almost all NAFLD patients [44]. The severity of insulin resistance is correlated with the progression of disease. However, the role of oxidative stress and inflammation in the pathogenesis of NAFLD cascade need to be further studied in human. In the setting of obesity, increased fatty acids and other related metabolites enhance oxidative phosphorylation and ATP generation, leads to increase ROS/RNS production and oxidative stress. Multiple stress-sensitive kinase signaling cascades, such as JNK and IKKβ, are activated by the increased oxidative stress. Once activated, these kinases are able to phosphorylate multiple targets, including the insulin receptor and the family of IRS proteins [101]. Insulin action is impaired by the abnormal serine/threonine phosphorylation in insulin receptor and IRS proteins such as IRS-1 and IRS-2, resulting in insulin resistance. In hyperglycemia caused by insulin resistance, intensive redox reactions occur during the process of protein glycation, generating a great deal of ROS [102]. Additionally, hyperglycemia and high insulin levels stimulate fatty acids synthesis and result in increasing lipid droplets storage within hepatocytes. The excessive intracellular levels of lipid can induce hepatocytes dysfunction or death. The increased ROS also act on large molecules such as poly-unsaturated fatty acids to initiate lipid per-oxidation, which further change the fluidity and permeability of the cell membrane. The inflammatory infiltration induced by lipid per-oxidation may also result in liver inflammation and necrosis, and even fibrosis. In mitochondrion, lipid peroxidation reduces the activity of mitochondrial respiratory chain, and thereby produces more ROS and increase oxidative stress. The prolonged oxidative stress may favor insulin resistance circularly, acting like a vicious circle. Then, the persistent exposure of oxidative stress and hyperglycemia contribute to NAFLD [103,104]. In addition to obesity, other risk factors such as drugs, re-feeding syndrome and other disorders are considered. For example, streptozotocin-induced diabetic rats constitutes as the model of oxidative stress. It was indicated that supplementation of alpha-tocopherol increased alpha-tocopherol in liver, but not in plasma [105]. Diet supplementation of acai, a promising source of natural antioxidants, could increase mRNA levels of gamma-glutamylcysteinesynthetase and GSH-Px in liver tissue, and decrease ROS produced by neutrophils. In addition, supplementation with acai could decrease thiobarbituric acid-reactive substances levels, and increase reduced GSH content in the liver. Moreover, the effect of dietary supplementation of vitamins C and E on oxidative stress and antioxidant redox systems was studied in streptozotocin-induced aged diabetic rats [106]. GSH-Px activity and the concentration of vitamin E in liver were lower, whereas lipid peroxidation levels in liver, and contents of ALT and AST in plasma were higher in the diabetic group than in the control group and were mostly restored by vitamins C and E treatment. Furthermore, the combined treatment with vitamin C, vitamin E, and Se showed a curative effect against the liver injury in streptozotocin-induced diabetic rats [107]. The effects of some antioxidants/plants on liver of streptozotocin-induced diabetic rats are summarized in Table 3.
Table 2. The effects of some antioxidants/plants on NAFLD.
Table 2. The effects of some antioxidants/plants on NAFLD.
Models (Prevent/Treatment)Antioxidant/PlantsEffectsDose (Dose-Effect)Bioactive CompoundsReferences
Diabetic rats fed on a high fat thermolyzed diet (Prevent)Omega 3-polyunsaturated fatty acids↑ SOD, CAT; ↓ triglycerides, non-esterified fatty acid, lipoperoxidation3.0% in dietOmega 3-polyunsaturated fatty acids[108]
Mice fed with high-fat diet (Prevent and treatment)Moringa oleifera leaves; haw pectic oligosaccharide; Thymbra spicata↑ GSH; ↓ ALT, AST, ALP, lipid peroxidation50, 150 and 300 mg/kg b.w. (No dose–effect)Haw pectic oligosaccharide[109,110,111]
Liver damage in diet-induced atherosclerotic rats (Prevent)Tulbaghia violacea rhizomes↓ LDH, AST, ALT, ALP, bilirubin antioxidation100 mg/kg b.w. [112]
Rabbits with high-fat diet (Prevent)Apolipoprotein A–I↑ SOD, GSH-Px; ↓ iNOS, MDA15 mg/kg b.w. [113]
WeRats fed a high-fat diet (Prevent)Black cabbage sprout↑ SOD, CAT, NADPH, GSH-Px, GRD GST250 and 500 mg/kg b.w. (Dose–effect) [114]
Table 3. The effects of some antioxidants/plants on liver of streptozotocin-induced diabetic rat.
Table 3. The effects of some antioxidants/plants on liver of streptozotocin-induced diabetic rat.
Models (Prevent/Treatment)MaterialsEffectsDose (Dose-Effect)References
Streptozotocin-induced diabetic aged rats (Prevent)Vitamins C and EAntioxidation, hepatoprotection [106]
Streptozotocin-induced diabetic rats (Prevent)AcaiAntioxidation, hepatoprotection2% (w/w) in standard diet[115]
Streptozotocin-induced diabetic rats (Prevent)Herba bidentisAntioxidation, hepatoprotection5 mL/kg[116]
Streptozotocin-induced diabetic rats (Prevent)(−)-EpicatechinAntioxidation15 and 30 mg/kg (Dose–effect)[117]
Streptozotocin-induced diabetic rats (Treatment)Stobadine 24.7 mg/kg[118]
Streptozotocin-induced diabetic mice (Prevent)Terminalia glaucescens leavesAntioxidation100 and 300 mg/kg (Dose–effect)[119]
Streptozotocin-induced diabetic rats (Treatment)BerberineAntioxidation75, 150 and 300 mg/kg (Dose–effect)[120]
Streptozotocin-induced diabetic rats (Prevent)Aloe vera leaves 300 mg/kg[121]
Streptozotocin-induced diabetic rats (Treatment)N-AcetylcysteineAntioxidation1.5 g/kg[122]
Streptozotocin-induced diabetic rats (Treatment)Oroxylum indicum stem barkAntioxidation250 mg/kg[123]
Streptozotocin-induced diabetic rats (Treatment)Maslinic acidAntioxidation40, 80 and 160 mg/kg (Dose–effect)[124]
Streptozotocin-induced diabetic rats (Treatment)ResveratrolAntioxidation20 mg/kg[125]
Streptozotocin-nicotinamide induced diabetic rats (Prevent)Stevia rebaudianaAntioxidation [126]

3.3. Antioxidants for Prevention and Treatment of Liver Diseases Induced by Other Factors

Since liver is an essential organ for detoxification and metabolism, and all pharmaceuticals make their way to the liver, for storage and therefore it is more prone to damage [127,128]. Paracetamol is widely used to relieve pain and reduce fever. Although use of paracetamol at its recommended dose is generally safe, overdose could still cause severe hepatic damage in many cases. As mentioned above, paracetamol may induce a remarkable increase of MDA and nitrite as well as nitrate in the liver, apart from a significant reduction in total SOD and Cu/Zn-SOD activity. Models of paracetamol-induced liver damage in mice/rats are widely used to study antioxidant and hepatoprotective effects of antioxidants/plants. For example, Rasool et al. [129] studied hepatoprotective and antioxidant effects of Gallic acid in paracetamol-induced liver damage in mice. It was shown that Gallic acid possessed antioxidant and hepatoprotective effects. In addition to paracetamol, some other drugs such as doxorubicin, tert-butyl hydroperoxide and d-galactosamine may also induce liver injury, which is possibly associated with the rise of oxidative stress. The effects of certain antioxidants/plants on paracetamol and other drugs-induced liver damage are summarized in Table 4. As seen from Table 4, a conclusion could be drawn that materials possessing antioxidant activity also hold capacity of hepatoprotection in animal model, which implies the correlation between antioxidative property of these compounds and their hepatoprotective effect.
Table 4. The effects of some antioxidants/plants on drugs-induced liver damage.
Table 4. The effects of some antioxidants/plants on drugs-induced liver damage.
Models (Prevent/Treatment)MaterialsEffectsDose (Dose-Effect)References
Paracetamol-induced liver toxicity in mice (Prevent)Gallic acidAntioxidation, hepatoprotection100 mg/kg[129]
Paracetamol-induced liver toxicity in mice (Prevent)SauchinoneAntioxidation, hepatoprotection30 mg/kg[130]
Paracetamol-induced liver toxicity in mice (Prevent)GenisteinAntioxidation, hepatoprotection50, 100 and 200 mg/kg (Dose-effect)[131]
Paracetamol-induced liver toxicity in mice (Prevent)Phyllanthus niruriAntioxidation, hepatoprotection100 mg/kg[132]
Paracetamol-induced liver toxicity in mice (Prevent)Polyalthia longifolia leavesAntioxidation, hepatoprotection200 mg/kg[133]
Paracetamol-induced liver damage in rats (Prevent)Boerhaavia diffusa leavesAntioxidation, hepatoprotection100, 200, 300 and 400 mg/kg/day (No dose-effect)[134]
Paracetamol-induced liver damage in rats (Prevent)Saponarin from Gypsophila trichotomaAntioxidation, hepatoprotection80 mg/kg/week[135]
Lipopolysaccharide-induced liver injury in rats (Prevent)Carnosic acidAntioxidation, hepatoprotection15, 30 and 60 mg/kg (Dose-effect)[136]
d-Galactosamine-induced liver injury in rats (Prevent)Combination of selenium, ascorbic acid, β-carotene, and α-tocopherolAntioxidation, hepatoprotection [137]
d-Galactosamine-induced liver injury in rats (Prevent)LeucasasperaAntioxidation, hepatoprotection200 and 400 mg/kg (No dose-effect)[138]
d-Galactosamine-induced liver injury in rats (Prevent)Swertiamarin from Enicostemma axillareAntioxidation, hepatoprotection100 and 200 mg/kg (No dose-effect)[139]
Lipopolysaccharide/d-galactosamineinduced liver injury in rats (Prevent)CurcuminAntioxidation, hepatoprotection100 mg/kg[140]
Lipopolysaccharide/d-galactosamineinduced liver injury in rats (Prevent)betulinic acidAntioxidation, hepatoprotection20 and 50 mg/kg (No dose-effect)[141]
Lipopolysaccharide/d-galactosamine induced hepatitis in rats (Prevent)TridaxprocumbensAntioxidation300 mg/kg[142]
Doxorubicin-induced liver injury in ratsN-acetylcysteineAntioxidation, hepatoprotection10 mg/kg[143]
Cisplatin-induced liver injury in rats (Prevent)Tomato juiceAntioxidation, hepatoprotection [144]
Tert-butyl hydroperoxide-induced liver injury in rats (Prevent)PropolisAntioxidation, hepatoprotection50 and 100 mg/kg (No dose-effect)[145]
Tamoxifen-induced liver injury in mice (Prevent)CatechinAntioxidation40 mg/kg[146]
Hepatic steatosis stimulated with tunicamycin (Treatment)Melatonin↓ ER stress, expression of miR-23a [147]
Ethionine-induced liver injury in mice (Prevent)MelatoninAntioxidation, hepatoprotection3 mg/kg[148]
Many pollutants and toxic substances could cause oxidative stress/damage of liver as mentioned above. Among pollutants and toxins that have been used to model hepatic injury in animals for studying effects of antioxidants/plants on pollutant-induced liver damage, carbon tetrachloride (CCl4) is most widely used. In CCl4-induced liver injury model, oxidative stress could be provoked, which prompts lipid peroxidation that injure hepatocellular membrane, followed by substantial release of pro-inflammatory chemokines and cytokines, which in consequence of liver damage [10]. A large amount of plants, especially medicinal plants, has been investigated to eliminate the hepatic damage stimulated by CCl4. For example, Coptidis rhizome, a traditional Chinese medicinal plant used to clear heat and scavenge toxins, belongs to liver meridian in Chinese medicinal practice [149,150]. The effect of Coptidis rhizome and its bioactive compound berberine on CCl4-induced chronic and acute hepatotoxicity in rats has been thoroughly studied by our research group [10,30,127]. We have found that Coptidis rhizome might act as an antioxidant to relieve CCl4-induced oxidative stress and hepatic damage. The mechanism may partly be ascribed to the reduced phosphorylation of Erk1/2 expression when exposed to oxidative stress [10]. The effects of some antioxidants/plants on toxic substances-induced liver damage are summarized in Table 5. It is particularly worth noting that Nrf2 could be activated by several antioxidants/plants in dimethylnitrosamine or cadmium induced hepatic injury models [151,152,153]. Antioxidant could induce both modification of inhibitor of Nrf2 (INrf2) cysteine 151 and PKC-mediated phosphorylation of Nrf2 serine 40 to release Nrf2 from INrf2. The dissociated and activated Nrf2 then translocates to the nucleus, binds to ARE and up-regulates antioxidants gene expression, which protects cells and relieves injury induced by oxidative stress [154]. Although most of the studies shown in Table 5 suggested the simultaneous role of these natural products as antioxidative and hepatoprotective agents, the related mechanisms and signal pathways have not yet fully studied.
Accumulating evidence demonstrated that ROS could lead to protein modification, lipid peroxidation, DNA damage and therefore acts as the initiator or promoter of carcinogenesis [155,156,157]. As the first line defense in suppressing tumor initiation, antioxidants are treated as one of the promising strategies to prevent liver cancer. Furthermore, it has been reported that the combination of certain chemotherapeutic drugs and antioxidants could reduce drug resistance, sensitizing the liver cancer cells to chemotherapeutics and therefore improving the efficacy of anti-cancer therapy [158]. Our previous studies demonstrated that Coptidis rhizome and berberine are promising agents to fight against liver cancer due to their hepatoprotective and antioxidant properties [155,157,159,160]. In all, cumulative evidence from epidemiological and clinical studies showed that consumption of suitable antioxidants from natural sources may beneficial in fighting against cancer without obvious adverse effects. Besides liver cancer, oxidative injury-associated liver damage induced by other disorders has also been mentioned for confirming the use of antioxidants in the related diseases. For example, it was found that taking catechin from green tea could reduce injury of liver in cholestatic rats induced by bile duct ligation [161]. Allopurinol, a competitive xanthine oxidase inhibitor, has also been used to reduce systemic oxidative stress. The xanthine oxidase over-activity is suggested to play a role in the altered intestinal permeability in cirrhosis, it was found in an open-label pilot study that changes in intestinal permeability correlated to changes in MDA serum values after allopurinol treatment [162]. Additionally, treatment with allopurinol in bile-duct ligation rats and TAA induced liver injury was shown to reduce ROS and thus attenuate brain edema [163]. Effects of certain antioxidants/plants on other substances-induced liver damage are summarized in Table 6, which suggested that some antioxidants possess anti-tumor and hepatoprotective effects collectively in vivo, but the relationship and mechanisms need further exploration.
Notably, melatonin, N-acethyl-5-metoxytryptamine, a famous hormone synthesized mainly by the pineal gland, has been demonstrated as having striking antioxidant properties in numerous studies. It has the remarkable capability to scavenge both ROS and RNS, and block transcriptional factors of pro-inflammatory cytokines. Recently, it has been applied to the treatment of liver disease in terms of reducing oxidative stress [164]. A variety of liver disease models, such as streptozocin-induced diabetic rats and TAA-induced or bile-duct ligated fibrosis rats, melatonin administration showed hepato-protection partially via improving oxidative damage. As a matter of fact, it has been demonstrated that melatonin is even better antioxidant than vitamin E and C in the contexts of certain disease. A comparative study of the protective effects of melatonin and vitamin E on extra-hepatic bile duct ligation in rats indicated that melatonin is much more efficient than vitamin E in reducing the cholestasis parameters, decreasing lipid peroxidation and restoring anti-oxidative enzymes [165,166]. Further investigations are required to evaluate antioxidant and hepato-protective effect of melatonin in clinic.
Table 5. The effects of some antioxidants/plants on toxins-induced liver damage.
Table 5. The effects of some antioxidants/plants on toxins-induced liver damage.
Model (Prevent/Treatment)Antioxidant/PlantEffectsDose/(Dose–Effect)Bioactive CompoundsReferences
CCl4-induced liver damage in rats (Prevent)Coptidis rhizome and berberine↑ SOD; ↓ ALT, AST, Erk1/2Berberine: 120 mg/kg b.w. Extract: 800 mg/kg b.w.Berberine[10]
CCl4-induced liver damage in rats (Prevent)Friedelin isolated from Azima tetracantha leaves↑ SOD, CAT, GSH, GSH-Px; ↓ ALT, AST, LDH [59]
CCl4-induced liver damage in rats (Treatment)N-butanol fraction of Actinidias deliciosa roots↑ GSH; ↓ ALT, AST, MDA(Dose–effect)Oleanolic acid[167]
CCl4-induced liver damage in rats (Prevent)Silybum marianum seeds↑ GSH; HDL/LDL; hepatoprotection100 mg/kg b.w. [168]
CCl4-induced liver damage in rats (Prevent)Dioclea reflexa seeds↑ SOD, CAT; ↓ Transaminases, MDA5 mg/kg (acute) 2.5 mg/kg b.w. (chronic) [169]
CCl4-induced liver damage in rats (Prevent)Morus bombycis, 2,5-dihydroxy-4,3′-di (β-d-glucopyranosyloxy)-trans-stilbene↓ Lipid peroxidation; hepatoprotection100, 300 and 500 mg/kg b.w. (No dose–effect) [170,171]
CCl4-induced liver damage in rats (Prevent)Nigella sativa, Urticadioica↑ Antioxidant enzyme; ↓ lipid peroxidation; hepatoprotectionNigella sativa: 0.2 mg/mL Urtica dioica: 0.2 mg/mL [172]
CCl4-induced liver damage in rats (Prevent)Pleurotusostreatus (oyster mushroom)↑ GSH, CAT, SOD, GSH-Px; ↓ ALT, AST, ALP, MDA200 mg/kg b.w. [173]
CCl4-induced liver damage in rats (Prevent)Cytisusscoparius↑ GSH, CAT, SOD, GSH-Px, GST, GRD; ↓ ALT, AST, LDH250 and 500 mg/kg (No dose–effect) [174]
CCl4-induced liver damage in rats (Prevent)Ethanol extract of Phellinusmerrillii↑ CAT, SOD, GSH-Px; ↓ ALT, AST0.5, 1 and 2 g/kg b.w. (No dose–effect) [175]
CCl4-induced liver damage in rats (Prevent)Ginkgo biloba↑ GSH, SOD, CAT, GSH-Px, GRD, albumin; hepatoprotection25 and 50 mg/kg b.w. (No dose–effect) [176]
CCl4-induced liver damage in mice (Prevent)Protein isolate from Phyllanthus niruri↑ SOD, CAT; ↓ ALT, ALP; lipid peroxidation5 mg/kg b.w. [177]
CCl4-induced liver damage in mice (Prevent)Kahweol and cafestol (Coffee)↓ ALT, AST, cytochrome P450 2E1, lipid peroxidationKahweol or cafestol: 10–100 mg/kg b.w. (Dose–effect)Kahweol and cafestol[178]
CCl4-induced liver damage in rats (Prevent)Cirsium setidens↑ GSH-Px; SOD; hepatoprotection500 mg/kg b.w. [179]
CCl4-induced liver damage in rats (Prevent)Curcumin and saikosaponin A↑ SOD, GSH; ↓ MDA; hepatoprotection [180]
CCl4-induced liver damage in rats (Prevent)Ethanolic extract of Momordica tuberosa tubersAntioxidation, hepatoprotection [181]
CCl4-induced liver damage in rats (Prevent)Oregano and rosemary↓ AST, ALT, ALP; antioxidation20 g/kg b.w. [182]
CCl4-induced liver damage in rats (Prevent)Enicostemma axillareAntioxidation, hepatoprotection100 and 200 mg/kg b.w. (No dose–effect) [139]
CCl4-induced liver damage in rats (Prevent)Ficuscarica leaves and fruits, Morus alba root barks↑ CAT, SOD, GSH; ↓ MDA, AST, ALT, ALP50 and 150 mg/kg b.w. (No dose–effect) [183]
CCl4-induced liver damage in rats (Prevent)Podophyllum hexandrum↑ GSH, GSH-Px, GRD, SOD, GST; ↓ AST, ALT, LDH20, 30 and 50 mg/kg b.w. (No dose–effect) [184]
CCl4-induced liver damage in rats (Prevent)Ficusreligiosa roots↑ CAT, GSH-Px, GRD, SOD, GST; ↓ lipid peroxidation; hepatoprotection [185]
CCl4-induced liver damage in rats (Prevent)Dehydroabietylamine, Carthamus tinctorious↓ AST, ALT, ALP; antioxidation [186]
CCl4-induced liver damage in rats (Prevent)Artemetin, Vitexglabrata↑ SOD, CAT, GSH-Px; ↓ AST, ALT, ALP, lipid peroxidation, TB [187]
CCl4-induced liver damage in mice (Prevent)Blueberry anthocyanins↑ SOD, CAT, GRD, glycogen; ↓ AST, ALT, MDA [188]
CCl4-induced liver damage in rats (Prevent)Matricaria chamomilla↑ SOD, CAT, GSH-Px, GSH; ↓ AST, ALT, MDA50, 100 and 200 mL/kg b.w. (No dose–effect) [189]
CCl4-induced liver damage in mice (Prevent)Lysimachia clethroides↑ SOD; ↓ AST, ALT, MDA150, 300 and 600 mg/kg b.w. (No dose–effect) [190]
CCl4-induced liver damage in rats (Prevent)Garcinia indica fruit rind↑ SOD, CAT, GRD, GSH-Px, GSH; ↓ AST, ALT, MDA400 and 800 mg/kg b.w. (No dose–effect) [191]
CCl4-induced liver damage in rats (Prevent)Agaricus blazei↑ GSH, GRD; ↓ AST, ALT, MDA500 mg/kg b.w. [192]
CCl4-induced liver damage in rats (Prevent)Nerium oleander flowers↑ SOD; ↓ AST, ALT, ALP, MDA100, 200 and 400 mg/kg b.w. (No dose–effect) [193]
CCl4-induced liver damage in rats (Prevent)Hybanthus enneaspermus↓ AST, ALT, ALP, TB; antioxidation200 and 400 mg/kg b.w. (No dose–effect) [194]
CCl4-induced liver damage in mice (Treatment)Anthocyanins in black rice bran↑ SOD, GSH-Px; hepatoprotection200, 400 and 800 mg/kg b.w. (No dose–effect) [195]
CCl4-induced liver damage in rats (Prevent)Roureainduta↑ SOD, CAT, GSH, GSH-Px; ↓ AST, ALT, total bilirubin;500 mg/kg b.w. [196]
CCl4-induced liver damage in rats (Prevent)Proanthocyanidins extracted from grape seeds↑ SOD, GSH, GSH-Px, CAT; ↓ lipid accumulation, liver injury, DNA damage400 mg/kg b.w.Proanthocyanidins[197]
CCl4-induced liver damage in mice (Prevent)Veronica ciliata↑ SOD, GSH; ↓ ALT, AST, ALP150, 300 and 600 mg/kg b.w. (No dose–effect) [198]
CCl4-induced liver damage in rats (Prevent)Subereamollis↑ SOD, GSH, GSH-Px, CAT; ↓ ALT, AST, ALP, MDA100, 200 and 400 mg/kg b.w. (No dose–effect) [199]
CCl4-induced liver damage in rats (Prevent)Solanum xanthocarpum leaves↑ SOD, CAT, GSH, GST; ↓ ALT, AST, ALP, LDH100 and 200 mg/kg b.w. (No dose–effect) [200]
CCl4-induced liver damage in rats (Prevent)AllopurinolModulation of NF-κB, cytokine production and oxidative stress50 mg/kg b.w.
CCl4 and H2O2 induced liver damage in goat (Prevent)Ocimumbasilicum, Trigonellafoenum-graecumAntioxidation [201]
TAA-induced liver injury (Prevent)Genistein↑ GSH; ↓ MDA, ALT, AST, TB0.5, 1.0 and 2.0 mg/kg b.w. (No dose–effect) [202]
TAA-Induced liver Cirrhosis in rats (Prevent)Andrographis paniculata LeafHepato-protection, ↓ ROS, LDH250 and 500 mg/kg b.w. (No dose–effect) [203]
TAA-induced hepatotoxicity in rats (Prevent)corianderAntioxidant; ↓ ALT, AST, ALP, TBARS, MPO, NO Phenolic compounds[204]
TAA-induced fibrosis in mice (Treatment)Ger-Gen-Chyn-Lian-TangAntioxidant; anti-fibrosis; modulation on TGF-β/TGF-β receptor signaling100 and 300 mg/kg b.w. (Dose–effect) [205]
TAA-induced hepatotoxicity in rats (Treatment)Trigonella foenum-graecumAntioxidant; hepato-protection; ↓ ALP, MDA [206]
TAA-induced hepatotoxicity in rats (Treatment)AllopurinolRegulating cellular redox-sensitive transcription factors [163]
Cigarette smoke-induced oxidative damage in liver of rats (Treatment)Sesbania grandiflora leaves↑ SOD, GSH, GSH-Px, CAT, GST, GRD, glucose-6-phosphate dehydrogenase; ↓ AST, ALT, ALP1000 mg/kg b.w. [207]
Cigarette smoking induced oxidative damage in liver of mice (Prevent)Vitamin E and selenium↑ GSH-Px, Se-GSH-Px [208]
Atrazine exposure rats (Prevent)Vitamin E↑ SOD, GSH-Px, CAT, GST; ↓ lipid peroxidation [209]
Methidathion-induced liver injury in rats (Prevent)Vitamins C and E↓ AST, ALT, ALP, MDA;Vitamin E: 50 mg/kg b.w.;Vitamin C: 20 mg/kg b.w. [210]
Pesticide (chlorpyriphos and cypermethrin) induced hepatic damage in mice (Prevent)Black tea↑ SOD, GSH, GSH-Px, CAT, GRD, GST; ↓ AST, ALT, ALP200 mg/mL b.w. [211]
Polychlorinated biphenyls induced hepatic damage in rats (Prevent)α-TocopherolAntioxidation50 mg/kg. b.w. [212]
Aflatoxin-induced hepatic injury in rats (Prevent)Urticadioica seed↑ SOD, GSH-Px, CAT, GRD, GST; ↓ lipid peroxides, hydroxyl radical and hydrogen peroxides2 mL/rat/day [213]
Thioacetamide-induced hepatic damage in rats (Prevent)eugenol↑ COX-2; ↓ AST, ALT, ALP, bilirubin, CYP2E1, lipid peroxidation; antioxidation10.7 mg/kg b.w. [214]
Lead-induced liver damage in rats (Prevent)Ginger↑ SOD, CAT; ↓ MDA100 mg/kg b.w. [215]
Dimethylnitrosamine-induced hepatic damage in rats (Prevent)Anthocyanins from purple sweet potato↑ Nrf2, NADPH, GSH, GST; ↓ yclooxygenase-2, MDA50, 100 and 200 mg/kg b.w. (No dose–effect)Anthocyanins[151]
Cadmium-induced hepatic injury in rats (Prevent)Heated garlic juice, ascorbic acid↑ Nrf2, SOD, CAT; ↓ MDAHeated garlic juice: 100 mg/kg b.w.; Ascorbic acid: 100 mg/kg b.w.Ascorbic acid[152]
Potassium bromate-induced hepatotoxicity of rat (Prevent)Launaea procumbens↑ SOD, CAT, GSH, GSH-Px, GRD, GST200 mg/kg b.w. [216]
Dimethylnitrosamine induced liver fibrosis in rats (Prevent)Platycodi radix root↑ Nrf2, heme oxygenase-1, NADPH, NQO1, GST; ↓ ALT, AST; anti-fibrotic action200 mg/kg b.w.Changkil[153]
As2O3-induced hepatotoxicity in cat (Prevent)Resveratrol↑ GSH; ↓ ROS, MDA3 mL/kg b.w. [217]
Sodiumarsenite induced liver damage in rats (Prevent)Emblica officinalisAntioxidation500 mg in 0.1 mL water, 100 g b.w. [218]
Trichloroacetic acid induced liver injury in rats (Prevent)Date palm fruit↑ SOD, CAT, GSH-Px; ↓ MDA0.5 and 2 g/L b.w. (No dose–effect) [219]
Table 6. Effects of some antioxidants/plants on other related liver disease.
Table 6. Effects of some antioxidants/plants on other related liver disease.
Stress (Prevent/Treatment)Antioxidant/PlantsEffectsDose (Dose–Effect)Bioactive CompoundsReferences
Human liver cancer cell lineMorinda pubescens leavesAntioxidation, cytotoxicity25, 50, 100 and 250 μg/mL b.w. (Dose–effect)Hyoscyamine[220]
Liver cancer of rats (Prevent)Chlorella vulgarisAntioxidation, antitumour50, 150 and 300 mg/mL b.w. (Dose–effect) [221]
Hepatocellular carcinomaCaesalpinia bonducella leaves↑ SOD, GSH, CAT; ↓ MDA, AST, ALT, ALP; anticancer Flavonoids, triterpenoids[222]
Liver cancer of mice (Prevent)Pleurotus pulmonarius (edible mushroom)Antioxidation, anti-tumor [158]
Rat with secondary biliary cirrhosis (Prevent)SilybinAntioxidation0.4 g/kg b.w. [223]
Cholestatic rats with bile duct ligation (Treatment)Green tea catechinAntioxidation, reducing hepatic fibrosis50 mg/kg b.w. [161]
Bile duct-ligated cholestatic rats (Treatment)Epigallocatechin-3-gallateAnti-fibrotic effects, ↓ phosphorylation of Smad2/3 and Akt5 mg/kg b.w. [224]
Bile duct-ligated cholestatic rats (Treatment)Holothuria arenicola↑ SOD, GSH, GST, CAT; ↓ MDA, AST, ALT, ALP200 mg/kg b.w.Phenolic compounds, chlorogenic acid, pyrogallol, rutin, coumaric acid[225]
Bile-duct ligated Rats (Treatment)Garlic↑ GSH; ↓ LDH, TB, MDA, MPO; ↓ TNF-α, TGF-β, MMP-13 [226]
Bile-duct ligated Rats (Treatment)thymoquinone↑ SOD, GSH-Px; ↓ MDA50 mg/kg b.w. [227]
Bile-duct ligated Rats (Treatment)N-acetylcysteine↑ GSH, CAT; ↓ MDA, ALT300 mg/kg b.w. [228]
Bile-duct ligated Rats (Prevent)Phaseolus trilobus↑ SOD; ↓ AST, ALT, ALP, LDH, TB, TBARS;125, 250 and 500 mg/kg b.w. (Dose–effect) [229]
Bile-duct ligated Rats (Treatment)Melatonin↓ TBARS, MPO10 and 100 mg/kg b.w. (Dose–effect) [230]
Ischemia/reperfusion in obese rats with fatty liverMelatonin↑ Antioxidant enzymes; ↓ AST, ALT, MAD, NOx metabolites10 mg/kg b.w. [231]
Bile-duct ligated Rats (Treatment)Allopurinol↓ ROS, brain edema100 mg/kg b.w. [232]
Restraint stress-induced liver injury in mice (Prevent)Astragali radix and Salviae radixAntioxidation, hepatoprotection50, 100 and 200 mg/kg b.w. (No dose–effect)Myelophil[233]

4. Current Anti-Oxidative Therapy in Clinical Trials

Clinical trials are extremely vital and indispensable for the development of anti-oxidative therapy. We looked up the related information of current anti-oxidative therapy in clinic at http://www.ClinicalTrials.gov website. Vitamins, especially vitamin E, are the most frequently studied antioxidant as dietary supplement in clinical trials for liver disease, primarily in phase 2/3. Some other nutritional antioxidants such as zinc and Coenzyme Q10 were studied in phase 2. Compounds including silymarin, metadoxine, N-acetylcystein, propofol, and mitoquinone mesylate, which partially act as antioxidant, have been used as drugs or supplement for liver disease. Some of them, such as silymarin, metadoxine and N-acetylcysteine, are studied for NAFLD or NASH or ALD in phase 4. For example, the application of antioxidants supplement consisted of siliphos, selenium, methionine, and alpha lipoic acid has been approved in patients with fatty liver and non-alcoholic steatohepatitis in Mexico. Plants and foods such as ginger, green tea extract, and chocolate have been adopted as food supplement for their anti-oxidative properties for liver disease. Furthermore, quercetin and resveratrol, two well-known bioactive compounds isolated from plants, have been studied as food supplement as antioxidants for liver disease in phase 3. Despite certain promising results have obtained in clinical trials, anti-oxidative therapy still has a long way to go. As a matter of fact, many antioxidants are highly effective for prevention or treatment in animal models, but in humans it does not appear to be effective for the treatment of established disease. For example, anti-TNF, which shows desirable treatment effects in animal model, appears not to be effective in patients with acute alcoholic hepatitis. Therefore, translational research is highly important for the application of antioxidant therapy in clinic. In the future, natural plants and bio-active compounds isolated from plants as well as endogenous antioxidants such as melatonin, which have shown strong anti-oxidative ability and hepato-protection effects, should be studied by clinic trials with large patient samples and longer duration time.

5. Conclusions and Prospects

Anti-oxidative therapy, mainly using natural and synthetic antioxidants, represents a reasonable therapeutic approach for the prevention and treatment of liver diseases due to the role of oxidative stress in contributing to initiation and progression of hepatic damage. However, although concept of anti-oxidative therapy has been raised for decades and intensive efforts have been paid, there is a long way to go for the application of antioxidants in liver disease. In current clinical trials, mechanisms by which drugs or compounds treat liver disease might partly attribute to anti-oxidative ability, but plain antioxidants mainly used as dietary supplement to prevent the progress of disease or improve the outcome of patients might also be effective. The complex role of oxidative stress in physiological and pathological processes, lacking studies of underlying mechanisms in humans, and other difficulties in translational research are challenges ahead. In current studies, intervention of antioxidants is explored widely in prevention models rather than treatment model, without elaborated underlying mechanism investigation. For natural plants study, the dose used, especially content of antioxidants, is always blurry, not to mention the shift dose for humans. For those studies in which dose–effect has been investigated, only small portion of plants antioxidant showed dose–effect manner for reducing liver injury, suggesting the complex role of oxidative stress in pathogenesis. In animal study, antioxidants are given to animals via oral or intraperitoneal injection. The route of administration is also an influence for absorption and bio-availability of antioxidants. Additionally, since liver is a central organ for metabolism, oxidative stress in liver diseases interacts with many other diseases such as kidney failure and diabetes, certain models in animal study should be improved. These limitations in current study might result in antioxidants that showed desirable effects for prevention or treatment in animal models, but in humans they do not appear to be effective for the treatment of established disease, which is a barrier for the development of anti-oxidative therapy in clinic. Therefore, translational research is of great importance for anti-oxidative therapy. Considering ROS and oxidative stress act positively in certain circumstances and the difference between animals and humans, the effective dose and safe dose, duration of treatment, absorption and bio-availability of antioxidants require thorough investigation. Furthermore, in the future, large-scale samples and appropriate duration of anti-oxidative treatment for liver diseases should be performed.

Acknowledgments

This research was partially supported by the research council of the University of Hong Kong (project codes: 104002889 and 104003422), Wong’s donation (project code: 200006276) and the donation of Gaia Family Trust, New Zealand (project code: 200007008).

Author Contributions

Sha Li wrote the manuscript; All authors commented on the manuscript and discussed the manuscript; Yibin Feng designed, revised and finalized the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AC
Autoimmune cholestatic liver diseases
ADH
Alcohol dehydrogenase
AIH
Autoimmune hepatitis
ALD
Alcoholic liver disease
ALP
Alkaline phosphatase
ALT
Alanine transaminase
ALDH
Aldehyde dehydrogenases
ARE
Antioxidant response element
AST
Aspartate aminotransferase
BHA
Butylated hydroxyanisole
bw
Body weight
CAT
Catalase
CCl4
Carbon tetrachloride
ER
Endoplasmic reticulum
GSH-Px
Glutathione peroxidase
GSH
Glutathione
GRD
Glutathione reductase
GST
Glutathione S-transferase
HDL
High density lipoprotein
HCV
Hepatitis C virus
IL-6
Interleukin 6
INH
Anti-tuberculosis agent isoniazid
iNOS
Inducible nitric oxide synthase (iNOS)
INrf2
Inhibitor of Nrf2
IKKβ
IκB kinase-β
IRS
Insulin receptor substrate
JNK
c-Jun N-terminal kinases
Keap1
kelch-like ECH-associated protein-1
LDH
lactate dehydrogenase
LDL
Low density lipoprotein
MDA
Malondialdehyde
MEOS
Microsomal ethanol oxidizing system
NADPH
Nicotinamide adenine dinucleotide phosphate-oxidase
NAFLD
Non-alcoholic fatty liver disease NAFLD
NO
Nitric Oxide
NQO1
NAD(P)H Dehydrogenase, Quinone 1
Nrf1
Nuclear respiratory factor 1
Nrf2
Erythroid 2-related factor 2
PKC
protein kinase C
PPARα
Peroxisome proliferator activated receptor α
RNS
Reactive nitrogen species
ROS
Reactive oxygen species (ROS)
SOD
Superoxide dismutases
TAA
Thioacetamide
TB
Total bilirubin
TBARS
Thiobarbituric acid-reactive substances
TNF
Tumor necrosis factor

References

  1. Finkel, T.; Holbrook, N.J. Oxidants, oxidative stress and the biology of ageing. Nature 2000, 408, 239–247. [Google Scholar] [CrossRef] [PubMed]
  2. Apel, K.; Hirt, H. Reactive oxygen species: Metabolism, oxidative stress, and signal transduction. Annu. Rev. Plant Biol. 2004, 55, 373–399. [Google Scholar] [CrossRef] [PubMed]
  3. McCord, J.M. The evolution of free radicals and oxidative stress. Am. J. Med. 2000, 108, 652–659. [Google Scholar] [CrossRef]
  4. Mittler, R. Oxidative stress, antioxidants and stress tolerance. Trends Plant Sci. 2002, 7, 405–410. [Google Scholar] [CrossRef]
  5. Sanchez-Valle, V.; Chavez-Tapia, N.C.; Uribe, M.; Mendez-Sanchez, N. Role of oxidative stress and molecular changes in liver fibrosis: A review. Curr. Med. Chem. 2012, 19, 4850–4860. [Google Scholar] [CrossRef] [PubMed]
  6. Sakaguchi, S.; Takahashi, S.; Sasaki, T.; Kumagai, T.; Nagata, K. Progression of alcoholic and non-alcoholic steatohepatitis: Common metabolic aspects of innate immune system and oxidative stress. Drug Metab. Pharmacokinet. 2011, 26, 30–46. [Google Scholar] [CrossRef] [PubMed]
  7. Cichoz-Lach, H.; Michalak, A. Oxidative stress as a crucial factor in liver diseases. World J. Gastroenterol. 2014, 20, 8082–8091. [Google Scholar] [CrossRef] [PubMed]
  8. Wu, D.; Cederbaum, A.I. Oxidative stress and alcoholic liver disease. Semin Liver Dis. 2009, 29, 141–154. [Google Scholar] [CrossRef] [PubMed]
  9. Li, A.N.; Li, S.; Zhang, Y.J.; Xu, X.R.; Chen, Y.M.; Li, H.B. Resources and biological activities of natural polyphenols. Nutrients 2014, 6, 6020–6047. [Google Scholar] [CrossRef] [PubMed]
  10. Feng, Y.; Wang, N.; Ye, X.; Li, H.; Feng, Y.; Cheung, F.; Nagamatsu, T. Hepatoprotective effect and its possible mechanism of Coptidis rhizoma aqueous extract on carbon tetrachloride-induced chronic liver hepatotoxicity in rats. J. Ethnopharmacol. 2011, 138, 683–690. [Google Scholar] [CrossRef] [PubMed]
  11. Singal, A.K.; Jampana, S.C.; Weinman, S.A. Antioxidants as therapeutic agents for liver disease. Liver Int. 2011, 31, 1432–1448. [Google Scholar] [CrossRef] [PubMed]
  12. Medina, J.; Moreno-Otero, R. Pathophysiological basis for antioxidant therapy in chronic liver disease. Drugs 2005, 65, 2445–2461. [Google Scholar] [CrossRef] [PubMed]
  13. Palma, H.E.; Wolkmer, P.; Gallio, M.; Correa, M.M.; Schmatz, R.; Thome, G.R.; Pereira, L.B.; Castro, V.S.; Pereira, A.B.; Bueno, A.; et al. Oxidative stress parameters in blood, liver, and kidney of diabetic rats treated with curcumin and/or insulin. Mol. Cell. Biochem. 2014, 386, 199–210. [Google Scholar] [CrossRef] [PubMed]
  14. Bosoi, C.R.; Yang, X.; Huynh, J.; Parent-Robitaille, C.; Jiang, W.; Tremblay, M.; Rose, C.F. Systemic oxidative stress is implicated in the pathogenesis of brain edema in rats with chronic liver failure. Free Radic. Biol. Med. 2012, 52, 1228–1235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wang, H.; Chen, X.; Su, Y.; Paueksakon, P.; Hu, W.; Zhang, M.Z.; Harris, R.C.; Blackwell, T.S.; Zent, R.; Pozzi, A. p47phox contributes to albuminuria and kidney fibrosis in mice. Kidney Int. 2015, 87, 948–962. [Google Scholar] [CrossRef] [PubMed]
  16. Valente, M.J.; Carvalho, F.; Bastos, M.; de Pinho, P.G.; Carvalho, M. Contribution of oxidative metabolism to cocaine-induced liver and kidney damage. Curr. Med. Chem. 2012, 19, 5601–5606. [Google Scholar] [CrossRef] [PubMed]
  17. Dey, A.; Lakshmanan, J. The role of antioxidants and other agents in alleviating hyperglycemia mediated oxidative stress and injury in liver. Food Funct. 2013, 4, 1148–1184. [Google Scholar] [CrossRef] [PubMed]
  18. Karabulut, A.B.; Gui, M.; Karabulut, E.; Kiran, T.R.; Ocak, S.G.; Otlu, O. Oxidant and antioxidant activity in rabbit livers treated with zoledronic acid. Transplant. Proc. 2010, 42, 3820–3822. [Google Scholar] [CrossRef] [PubMed]
  19. Mallikarjuna, K.; Shanmugam, K.R.; Nishanth, K.; Wu, M.C.; Hou, C.W.; Kuo, C.H.; Reddy, K.S. Alcohol-induced deterioration in primary antioxidant and glutathione family enzymes reversed by exercise training in the liver of old rats. Alcohol 2010, 44, 523–529. [Google Scholar] [CrossRef] [PubMed]
  20. Tang, W.; Jiang, Y.F.; Ponnusamy, M.; Diallo, M. Role of Nrf2 in chronic liver disease. World J. Gastroenterol. 2014, 20, 13079–13087. [Google Scholar] [CrossRef] [PubMed]
  21. Zhang, Y.K.; Wu, K.C.; Klaassen, C.D. Genetic activation of Nrf2 protects against fasting-induced oxidative stress in livers of mice. PLoS ONE 2013, 8, e59122. [Google Scholar] [CrossRef] [PubMed]
  22. Klaassen, C.D.; Reisman, S.A. Nrf2 the rescue: Effects of the antioxidative/electrophilic response on the liver. Toxicol. Appl. Pharmacol. 2010, 244, 57–65. [Google Scholar] [CrossRef] [PubMed]
  23. Emerit, I.; Huang, C.Y.; Serejo, F.; Filipe, P.; Fernandes, A.; Costa, A.; Freitas, J.; Baptista, A.; Carneiro de Moura, M. Oxidative stress in chronic hepatitis C: A preliminary study on the protective effects of antioxidant flavonoids. Hepatogastroenterology 2005, 52, 530–536. [Google Scholar] [PubMed]
  24. Esrefoglu, M. Oxidative stress and benefits of antioxidant agents in acute and chronic hepatitis. Hepat. Mon. 2012, 12, 160–167. [Google Scholar] [CrossRef] [PubMed]
  25. Farias, M.S.; Budni, P.; Ribeiro, C.M.; Parisotto, E.B.; Santos, C.E.; Dias, J.F.; Dalmarco, E.M.; Frode, T.S.; Pedrosa, R.C.; Wilhelm Filho, D. Antioxidant supplementation attenuates oxidative stress in chronic hepatitis C patients. Gastroenterol. Hepatol. 2012, 35, 386–394. [Google Scholar] [CrossRef] [PubMed]
  26. Groenbaek, K.; Friis, H.; Hansen, M.; Ring-Larsen, H.; Krarup, H.B. The effect of antioxidant supplementation on hepatitis C viral load, transaminases and oxidative status: A randomized trial among chronic hepatitis C virus-infected patients. Eur. J. Gastroenterol. Hepatol. 2006, 18, 985–989. [Google Scholar] [CrossRef] [PubMed]
  27. Zhao, J.; Fan, Y.C.; Zhang, F.; Yang, Y.; Zhao, Z.H.; Sun, F.K.; Wang, K. Oxidative stress in chronic hepatitis C patients. Zhonghua Shi Yan He Lin Chuang Bing Du Xue Za Zhi 2013, 27, 47–49. (In Chinese) [Google Scholar] [PubMed]
  28. Kawanaka, M.; Mahmood, S.; Niiyama, G.; Izumi, A.; Kamei, A.; Ikeda, H.; Suehiro, M.; Togawa, K.; Sasagawa, T.; Okita, M.; et al. Control of oxidative stress and reduction in biochemical markers by vitamin E treatment in patients with nonalcoholic steatohepatitis: A pilot study. Hepatol. Res. 2004, 29, 39–41. [Google Scholar] [CrossRef] [PubMed]
  29. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  30. Wang, N.; Feng, Y.B.; Cheung, F.; Chow, O.Y.; Wang, X.B.; Su, W.W.; Tong, Y. A comparative study on the hepatoprotective action of bear bile and coptidis rhizoma aqueous extract on experimental liver fibrosis in rats. BMC Complement. Altern. Med. 2012, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Loomba, R.; Wesley, R.; Pucino, F.; Liang, T.J.; Kleiner, D.E.; Lavine, J.E. Placebo in nonalcoholic steatohepatitis: Insight into natural history and implications for future clinical trials. Clin. Gastroenterol. Hepatol. 2008, 6, 1243–1248. [Google Scholar] [CrossRef] [PubMed]
  32. Masalkar, P.D.; Abhang, S.A. Oxidative stress and antioxidant status in patients with alcoholic liver disease. Clin. Chim. Acta 2005, 355, 61–65. [Google Scholar] [CrossRef] [PubMed]
  33. Louvet, A.; Mathurin, P. Alcoholic liver disease: Mechanisms of injury and targeted treatment. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 231–242. [Google Scholar] [CrossRef] [PubMed]
  34. Zima, T.; Kalousova, M. Oxidative stress and signal transduction pathways in alcoholic liver disease. Alcohol. Clin. Exp. Res. 2005, 29, 110S–115S. [Google Scholar] [CrossRef] [PubMed]
  35. Zhu, H.; Jia, Z.; Misra, H.; Li, Y.R. Oxidative stress and redox signaling mechanisms of alcoholic liver disease: Updated experimental and clinical evidence. J. Dig. Dis. 2012, 13, 133–142. [Google Scholar] [CrossRef] [PubMed]
  36. Banerjee, P.; Jana, S.; Chakraborty, S.; Swarnakar, S. Inflammation and MMPs in alcohol-induced liver diseases and protective action of antioxidants. Indian J. Biochem. Biol. 2013, 50, 377–386. [Google Scholar]
  37. Gao, B.; Bataller, R. Alcoholic liver disease: Pathogenesis and new therapeutic targets. Gastroenterology 2011, 141, 1572–1585. [Google Scholar] [CrossRef] [PubMed]
  38. Beier, J.I.; McClain, C.J. Mechanisms and cell signaling in alcoholic liver disease. Biol. Chem. 2010, 391, 1249–1264. [Google Scholar] [CrossRef] [PubMed]
  39. Diehl, A.M. Recent events in alcoholic liver disease V. Effects of ethanol on liver regeneration. Am. J. Physiol.-Gastrointest. Liver Physiol. 2005, 288, G1–G6. [Google Scholar] [CrossRef] [PubMed]
  40. Shanmugam, K.R.; Mallikarjuna, K.; Reddy, K.S. Effect of alcohol on blood glucose and antioxidant enzymes in the liver and kidney of diabetic rats. Indian J. Pharmacol. 2011, 43, 330–335. [Google Scholar] [PubMed]
  41. Babczynska, A.; Wilczek, G.; Migula, P. Effects of dimethoate on spiders from metal pollution gradient. Sci. Total Environ. 2006, 370, 352–359. [Google Scholar] [CrossRef] [PubMed]
  42. Dahiru, D.; Obidoa, O. Evaluation of the antioxidant effects of Ziziphus mauritiana lam. leaf extracts against chronic ethanol-induced hepatotoxicity in rat liver. Afr. J. Tradit. Complement. Altern. Med. 2007, 5, 39–45. [Google Scholar] [CrossRef] [PubMed]
  43. Das, K.S.; Balakrishnan, V.; Mukherjee, S.; Vasudevan, D.M. Evaluation of blood oxidative stress-related parameters in alcoholic liver disease and non-alcoholic fatty liver disease. Scand. J. Clin. Lab. Investig. 2008, 68, 323–334. [Google Scholar] [CrossRef] [PubMed]
  44. Videla, L.A.; Rodrigo, R.; Orellana, M.; Fernandez, V.; Tapia, G.; Quinones, L.; Varela, N.; Contreras, J.; Lazarte, R.; Csendes, A.; et al. Oxidative stress-related parameters in the liver of non-alcoholic fatty liver disease patients. Clin. Sci. 2004, 106, 261–268. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, Y.L.; Chen, L.J.; Bair, M.J.; Yao, M.L.; Peng, H.C.; Yang, S.S.; Yang, S.C. Antioxidative status of patients with alcoholic liver disease in southeastern Taiwan. World J. Gastroenterol. 2011, 17, 1063–1070. [Google Scholar] [PubMed]
  46. Albano, E. Oxidative mechanisms in the pathogenesis of alcoholic liver disease. Mol. Asp. Med. 2008, 29, 9–16. [Google Scholar] [CrossRef] [PubMed]
  47. Galicia-Moreno, M.; Gutierrez-Reyes, G. The role of oxidative stress in the development of alcoholic liver disease. Revista de Gastroenterología de México 2014, 79, 135–144. (In Spanish) [Google Scholar] [CrossRef] [PubMed]
  48. Videla, L.A. Oxidative stress signaling underlying liver disease and hepatoprotective mechanisms. World J. Hepatol. 2009, 1, 72–78. [Google Scholar] [CrossRef] [PubMed]
  49. Linares, V.; Alonso, V.; Albina, M.L.; Belles, M.; Sirvent, J.J.; Domingo, J.L.; Sanchez, D.J. Lipid peroxidation and antioxidant status in kidney and liver of rats treated with sulfasalazine. Toxicology 2009, 256, 152–156. [Google Scholar] [CrossRef] [PubMed]
  50. Mladenovic, D.; Radosavljevic, T.; Ninkovic, M.; Vucevic, D.; Jesic-Vukicevic, R.; Todorovic, V. Liver antioxidant capacity in the early phase of acute paracetamol-induced liver injury in mice. Food Chem. Toxicol. 2009, 47, 866–870. [Google Scholar] [CrossRef] [PubMed]
  51. Samarghandian, S.; Afshari, R.; Farkhondeh, T. Effect of long-term treatment of morphine on enzymes, oxidative stress indices and antioxidant status in male rat liver. Int. J. Clin. Exp. Med. 2014, 7, 1449–1453. [Google Scholar] [PubMed]
  52. Pieniazek, A.; Czepas, J.; Piasecka-Zelga, J.; Gwozdzinski, K.; Koceva-Chyla, A. Oxidative stress induced in rat liver by anticancer drugs doxorubicin, paclitaxel and docetaxel. Adv. Med. Sci. 2013, 58, 104–111. [Google Scholar] [CrossRef] [PubMed]
  53. Nguyen, V.; Huang, J.; Doan, V.; Lin, X.; Tang, X.; Huang, Y.; Tang, A.; Yang, X.; Huang, R. Hepatoprotective effects of Yulangsan polysaccharide against nimesulide-induced liver injury in mice. J. Ethnopharmacol. 2015, 172, 273–280. [Google Scholar] [CrossRef] [PubMed]
  54. Zlatkovic, J.; Todorovic, N.; Tomanovic, N.; Boskovic, M.; Djordjevic, S.; Lazarevic-Pasti, T.; Bernardi, R.E.; Djurdjevic, A.; Filipovic, D. Chronic administration of fluoxetine or clozapine induces oxidative stress in rat liver: A histopathological study. Eur. J. Pharm. Sci. 2014, 59, 20–30. [Google Scholar] [CrossRef] [PubMed]
  55. Stine, J.G.; Chalasani, N. Chronic liver injury induced by drugs: A systematic review. Liver Int. 2015. [Google Scholar] [CrossRef] [PubMed]
  56. Shuhendler, A.J.; Pu, K.; Cui, L.; Uetrecht, J.P.; Rao, J. Real-time imaging of oxidative and nitrosative stress in the liver of live animals for drug-toxicity testing. Nat. Biotechnol. 2014, 32, 373–380. [Google Scholar] [CrossRef] [PubMed]
  57. O’Riordan, A.; Brummell, Z.; Sizer, E.; Auzinger, G.; Heaton, N.; O’Grady, J.G.; Bernal, W.; Hendry, B.M.; Wendon, J.A. Acute kidney injury in patients admitted to a liver intensive therapy unit with paracetamol-induced hepatotoxicity. Nephrol. Dial. Transplant. 2011, 26, 3501–3508. [Google Scholar] [CrossRef] [PubMed]
  58. Bando, I.; Reus, M.I.; Andres, D.; Cascales, M. Endogenous antioxidant defence system in rat liver following mercury chloride oral intoxication. J. Biochem. Mol. Toxicol. 2005, 19, 154–161. [Google Scholar] [CrossRef] [PubMed]
  59. Adegbesan, B.O.; Adenuga, G.A. Effect of lead exposure on liver lipid peroxidative and antioxidant defense systems of protein-undernourished rats. Biol. Trace Element Res. 2007, 116, 219–225. [Google Scholar] [CrossRef] [PubMed]
  60. Moreno, I.; Pichardo, S.; Jos, A.; Gomez-Amores, L.; Mate, A.; Vazquez, C.M.; Camean, A.M. Antioxidant enzyme activity and lipid peroxidation in liver and kidney of rats exposed to microcystin-LR administered intraperitoneally. Toxicon 2005, 45, 395–402. [Google Scholar] [CrossRef] [PubMed]
  61. Ozgur, E.; Guler, G.; Seyhan, N. Mobile phone radiation-induced free radical damage in the liver is inhibited by the antioxidants N-acetyl cysteine and epigallocatechin-gallate. Int. J. Radiat. Biol. 2010, 86, 935–945. [Google Scholar] [CrossRef] [PubMed]
  62. Yildirim, N.C.; Yurekli, M.; Yildirim, N. Investigation of some antioxidant enzymes activities depending on adrenomedullin treatment and cold stress in rat liver tissue. Turk. J. Biochem. 2010, 35, 138–142. [Google Scholar]
  63. Jia, X.; Wu, Y.; Liu, P. Effects of flour bleaching agent on mice liver antioxidant status and ATPases. Environ. Toxicol. Pharmacol. 2011, 31, 479–484. [Google Scholar] [CrossRef] [PubMed]
  64. Syama, S.; Reshma, S.C.; Sreekanth, P.J.; Varma, H.K.; Mohanan, P.V. Effect of zinc oxide nanoparticles on cellular oxidative stress and antioxidant defense mechanisms in mouse liver. Environ. Toxicol. Pharmacol. 2013, 95, 495–503. [Google Scholar] [CrossRef]
  65. Zhang, X.; Strakovsky, R.; Zhou, D.; Zhang, Y.; Pan, Y.X. A maternal high-fat diet represses the expression of antioxidant defense genes and induces the cellular senescence pathway in the liver of male offspring rats. J. Nutr. 2011, 141, 1254–1259. [Google Scholar] [CrossRef] [PubMed]
  66. Dornas, W.C.; de Lima, W.G.; dos Santos, R.C.; Guerra, J.F.; de Souza, M.O.; Silva, M.; Souza e Silva, L.; Diniz, M.F.; Silva, M.E. High dietary salt decreases antioxidant defenses in the liver of fructose-fed insulin-resistant rats. J. Nutr. Biochem. 2013, 24, 2016–2022. [Google Scholar] [CrossRef] [PubMed]
  67. Messarah, M.; Klibet, F.; Boumendjel, A.; Abdennour, C.; Bouzerna, N.; Boulakoud, M.S.; el Feki, A. Hepatoprotective role and antioxidant capacity of selenium on arsenic-induced liver injury in rats. Exp. Toxicol. Pathol. 2012, 64, 167–174. [Google Scholar] [CrossRef] [PubMed]
  68. Kaffe, E.T.; Rigopoulou, E.I.; Koukoulis, G.K.; Dalekos, G.N.; Moulas, A.N. Oxidative stress and antioxidant status in patients with autoimmune liver diseases. Redox Rep. 2015, 20, 33–41. [Google Scholar] [CrossRef] [PubMed]
  69. Madan, K.; Bhardwaj, P.; Thareja, S.; Gupta, S.D.; Saraya, A. Oxidant stress and antioxidant status among patients with nonalcoholic fatty liver disease (NAFLD). J. Clin. Gastroenterol. 2006, 40, 930–935. [Google Scholar] [CrossRef] [PubMed]
  70. Feagins, L.A.; Flores, A.; Arriens, C.; Park, C.; Crook, T.; Reimold, A.; Brown, G. Nonalcoholic fatty liver disease: A potential consequence of tumor necrosis factor-inhibitor therapy. Eur. J. Gastroenterol. Hepatol. 2015, 27, 1154–1160. [Google Scholar] [CrossRef] [PubMed]
  71. Coffin, C.S.; Fraser, H.F.; Panaccione, R.; Ghosh, S. Liver diseases associated with anti-tumor necrosis factor-alpha (TNF-α) use for inflammatory bowel disease. Inflamm. Bowel Dis. 2011, 17, 479–484. [Google Scholar] [CrossRef] [PubMed]
  72. Frazier, T.H.; Stocker, A.M.; Kershner, N.A.; Marsano, L.S.; McClain, C.J. Treatment of alcoholic liver disease. Ther. Adv. Gastroenterol. 2011, 4, 63–81. [Google Scholar] [CrossRef] [PubMed]
  73. Ding, W.X.; Li, M.; Chen, X.; Ni, H.M.; Lin, C.W.; Gao, W.; Lu, B.; Stolz, D.B.; Clemens, D.L.; Yin, X.M. Autophagy reduces acute ethanol-induced hepatotoxicity and steatosis in mice. Gastroenterology 2010, 139, 1740–1752. [Google Scholar] [CrossRef] [PubMed]
  74. Diehl, A.M.; Chute, J. Underlying potential: Cellular and molecular determinants of adult liver repair. J. Clin. Investig. 2013, 123, 1858–1860. [Google Scholar] [CrossRef] [PubMed]
  75. Deng, G.F.; Xu, X.R.; Zhang, Y.; Li, D.; Gan, R.Y.; Li, H.B. Phenolic compounds and bioactivities of pigmented rice. Crit. Rev. Food Sci. Nutr. 2013, 53, 296–306. [Google Scholar] [CrossRef] [PubMed]
  76. Fu, L.; Xu, B.T.; Gan, R.Y.; Zhang, Y.; Xu, X.R.; Xia, E.Q.; Li, H.B. Total phenolic contents and antioxidant capacities of herbal and tea infusions. Int. J. Mol. Sci. 2011, 12, 2112–2124. [Google Scholar] [CrossRef] [PubMed]
  77. Deng, G.F.; Xu, X.R.; Guo, Y.J.; Xia, E.Q.; Li, S.; Wu, S.; Chen, F.; Ling, W.H.; Li, H.B. Determination of antioxidant property and their lipophilic and hydrophilic phenolic contents in cereal grains. J. Funct. Foods 2012, 4, 906–914. [Google Scholar] [CrossRef]
  78. Deng, G.F.; Lin, X.; Xu, X.R.; Gao, L.L.; Xie, J.F.; Li, H.B. Antioxidant capacities and total phenolic contents of 56 vegetables. J. Funct. Foods 2013, 5, 260–266. [Google Scholar] [CrossRef]
  79. Fu, L.; Xu, B.T.; Xu, X.R.; Gan, R.Y.; Zhang, Y.; Xia, E.Q.; Li, H.B. Antioxidant capacities and total phenolic contents of 62 fruits. Food Chem. 2011, 129, 345–350. [Google Scholar] [CrossRef]
  80. Guo, Y.J.; Deng, G.F.; Xu, X.R.; Wu, S.; Li, S.; Xia, E.Q.; Li, F.; Chen, F.; Ling, W.H.; Li, H.B. Antioxidant capacities, phenolic compounds and polysaccharide contents of 49 edible macro-fungi. Food Funct. 2012, 3, 1195–1205. [Google Scholar] [CrossRef] [PubMed]
  81. Li, H.B.; Cheng, K.W.; Wong, C.C.; Fan, K.W.; Chen, F.; Jiang, Y. Evaluation of antioxidant capacity and total phenolic content of different fractions of selected microalgae. Food Chem. 2007, 102, 771–776. [Google Scholar] [CrossRef]
  82. Li, S.; Li, S.K.; Gan, R.Y.; Song, F.L.; Kuang, L.; Li, H.B. Antioxidant capacities and total phenolic contents of infusions from 223 medicinal plants. Ind. Crops Prod. 2013, 51, 289–298. [Google Scholar] [CrossRef]
  83. Li, A.N.; Li, S.; Li, H.B.; Xu, D.P.; Xu, X.R.; Chen, F. Total phenolic contents and antioxidant capacities of 51 edible and wild flowers. J. Funct. Foods 2014, 6, 319–330. [Google Scholar] [CrossRef]
  84. Augustyniak, A.; Waszkiewicz, E.; Skrzydlewska, E. Preventive action of green tea from changes in the liver antioxidant abilities of different aged rats intoxicated with ethanol. Nutrition 2005, 21, 925–932. [Google Scholar] [CrossRef] [PubMed]
  85. Bourogaa, E.; Nciri, R.; Mezghani-Jarraya, R.; Racaud-Sultan, C.; Damak, M.; el Feki, A. Antioxidant activity and hepatoprotective potential of Hammada scoparia against ethanol-induced liver injury in rats. J. Physiol. Biochem. 2013, 69, 227–237. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, M.; Zhu, P.; Jiang, C.; Ma, L.; Zhang, Z.; Zeng, X. Preliminary characterization, antioxidant activity in vitro and hepatoprotective effect on acute alcohol-induced liver injury in mice of polysaccharides from the peduncles of Hovenia dulcis. Food Chem. Toxicol. 2012, 50, 2964–2970. [Google Scholar] [CrossRef] [PubMed]
  87. Lopez, V.R.L.; Razzeto, G.S.; Gimenez, M.S.; Escudero, N.L. Antioxidant properties of Amaranthus hypochondriacus seeds and their effect on the liver of alcohol-treated rats. Plant Foods Hum. Nutr. 2011, 66, 157–162. [Google Scholar] [CrossRef] [PubMed]
  88. Cheng, N.; Du, B.; Wang, Y.; Gao, H.; Cao, W.; Zheng, J.; Feng, F. Antioxidant properties of jujube honey and its protective effects against chronic alcohol-induced liver damage in mice. Food Funct. 2014, 5, 900–908. [Google Scholar] [CrossRef] [PubMed]
  89. Mohd Ali, N.; Mohd Yusof, H.; Long, K.; Yeap, S.K.; Ho, W.Y.; Beh, B.K.; Koh, S.P.; Abdullah, M.P.; Alitheen, N.B. Antioxidant and hepatoprotective effect of aqueous extract of germinated and fermented mung bean on ethanol-mediated liver damage. BioMed Res. Int. 2013, 2013, 693613. [Google Scholar] [CrossRef] [PubMed]
  90. Kasdallah-Grissa, A.; Nakbi, A.; Koubaa, N.; El-Fazaa, S.; Gharbi, N.; Kamoun, A.; Hammami, M. Dietary virgin olive oil protects against lipid peroxidation and improves antioxidant status in the liver of rats chronically exposed to ethanol. Nutr. Res. 2008, 28, 472–479. [Google Scholar] [CrossRef] [PubMed]
  91. Li, G.L.; Ye, Y.; Kang, J.J.; Yao, X.Y.; Zhang, Y.Z.; Jiang, W.; Gao, M.; Dai, Y.D.; Xin, Y.Q.; Wang, Q.; et al. l-Theanine prevents alcoholic liver injury through enhancing the antioxidant capability of hepatocytes. Food Chem. Toxicol. 2012, 50, 363–372. [Google Scholar] [CrossRef] [PubMed]
  92. Kaur, J.; Shalini, S.; Bansal, M.P. Influence of vitamin E on alcohol-induced changes in antioxidant defenses in mice liver. Toxicol. Mech. Methods 2010, 20, 82–89. [Google Scholar] [CrossRef] [PubMed]
  93. Altavilla, D.; Marini, H.; Seminara, P.; Squadrito, G.; Minutoli, L.; Passaniti, M.; Bitto, A.; Calapai, G.; Calo, M.; Caputi, A.P.; et al. Protective effects of antioxidant raxofelast in alcohol-induced liver disease in mice. Pharmacology 2005, 74, 6–14. [Google Scholar] [CrossRef] [PubMed]
  94. Penumathsa, S.V.; Kode, A.; Rajagopalan, R.; Menon, V.P. Changes in activities of MMP in alcohol and thermally oxidized sunflower oil-induced liver damage: NAC antioxidant therapy. Toxicol. Mech. Methods 2006, 16, 267–274. [Google Scholar] [CrossRef] [PubMed]
  95. Jung, Y.S.; Kim, S.J.; Kwon do, Y.; Ahn, C.W.; Kim, Y.S.; Choi, D.W.; Kim, Y.C. Alleviation of alcoholic liver injury by betaine involves an enhancement of antioxidant defense via regulation of sulfur amino acid metabolism. Food Chem. Toxicol. 2013, 62, 292–298. [Google Scholar] [CrossRef] [PubMed]
  96. Yi, J.; Xia, W.; Wu, J.; Yuan, L.; Wu, J.; Tu, D.; Fang, J.; Tan, Z. Betulinic acid prevents alcohol-induced liver damage by improving the antioxidant system in mice. J. Vet. Sci. 2014, 15, 141–148. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, P.; Qiang, X.; Zhang, M.; Ma, D.; Zhao, Z.; Zhou, C.; Liu, X.; Li, R.; Chen, H.; Zhang, Y. Demethyleneberberine, a natural mitochondria-targeted antioxidant, inhibits mitochondrial dysfunction, oxidative stress, and steatosis in alcoholic liver disease mouse model. J. Pharmacol. Exp. Ther. 2015, 352, 139–147. [Google Scholar] [CrossRef] [PubMed]
  98. Lirussi, F.; Azzalini, L.; Orando, S.; Orlando, R.; Angelico, F. Antioxidant supplements for non-alcoholic fatty liver disease and/or steatohepatitis. Cochrane Database Syst. Rev. 2007, 24, CD004996. [Google Scholar]
  99. Sattar, N.; Forrest, E.; Preiss, D. Non-alcoholic fatty liver disease. BMJ 2014, 349, 8. [Google Scholar] [CrossRef] [PubMed]
  100. Lee, J.H.; Friso, S.; Choi, S.W. Epigenetic mechanisms underlying the link between non-alcoholic fatty liver diseases and nutrition. Nutrients 2014, 6, 3303–3325. [Google Scholar] [CrossRef] [PubMed]
  101. Carter-Kent, C.; Zein, N.N.; Feldstein, A.E. Cytokines in the pathogenesis of fatty liver and disease progression to steatohepatitis: Implications for treatment. Am. J. Gastroenterol. 2008, 103, 1036–1042. [Google Scholar] [CrossRef] [PubMed]
  102. Evans, J.L.; Goldfine, I.D.; Maddux, B.A.; Grodsky, G.M. Are oxidative stress-activated signaling pathways mediators of insulin resistance and β-cell dysfunction? Diabetes 2003, 52, 1–8. [Google Scholar] [CrossRef] [PubMed]
  103. Tamura, Y.; Sato, F.; Kawamori, R. Metabolic syndrome and NAFLD/NASH. Nihon Rinsho 2006, 64, 449–452. [Google Scholar] [PubMed]
  104. Bogdanova, K.; Poczatkova, H.; Uherkova, L.; Riegrova, D.; Rypka, M.; Feher, J.; Marchesini, G.; Vesely, J. Non-alcoholic fatty liver disease (NAFLD)—A novel common aspect of the metabolic syndrome. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 2006, 150, 101–104. [Google Scholar] [CrossRef] [PubMed]
  105. Garcia-Martinez, D.; Ruperez, F.J.; Ugarte, P.; Barbas, C. Tocopherol fate in plasma and liver of streptozotocin-treated rats that orally received antioxidants and Spirulina extracts. Int. J. Vitam. Nutr. Res. 2007, 77, 263–271. [Google Scholar] [CrossRef] [PubMed]
  106. Naziroglu, M.; Butterworth, P.J.; Sonmez, T.T. Dietary vitamin C and E modulates antioxidant levels in blood, brain, liver, muscle, and testes in diabetic aged rats. Int. J. Vitam. Nutr. Res. 2011, 81, 347–357. [Google Scholar] [CrossRef] [PubMed]
  107. Gezginci-Oktayoglu, S.; Basaraner, H.; Yanardag, R.; Bolkent, S. The effects of combined treatment of antioxidants on the liver injury in STZ diabetic rats. Dig. Dis. Sci. 2009, 54, 538–546. [Google Scholar] [CrossRef] [PubMed]
  108. De Assis, A.M.; Rech, A.; Longoni, A.; Rotta, L.N.; Denardin, C.C.; Pasquali, M.A.; Souza, D.O.; Perry, M.L.S.; Moreira, J.C. Omega 3-polyunsaturated fatty acids prevent lipoperoxidation, modulate antioxidant enzymes, and reduce lipid content but do not alter glycogen metabolism in the livers of diabetic rats fed on a high fat thermolyzed diet. Mol. Cell. Biochem. 2012, 361, 151–160. [Google Scholar] [CrossRef] [PubMed]
  109. Li, T.P.; Liu, Y.H.; Dong, Y.P.; Li, S.H.; Zhu, R.G. Anti-fat deposition and antioxidant effects of haw pectic oligosaccharide in the liver of high-fat-fed mice. CyTA-J. Food 2014, 12, 27–31. [Google Scholar] [CrossRef]
  110. Akkol, E.K.; Avci, G.; Kucukkurt, I.; Keles, H.; Tamer, U.; Ince, S.; Yesilada, E. Cholesterol-reducer, antioxidant and liver protective effects of Thymbra spicata L. var. spicata. J. Ethnopharmacol. 2009, 126, 314–319. [Google Scholar] [CrossRef] [PubMed]
  111. Das, N.; Sikder, K.; Ghosh, S.; Fromenty, B.; Dey, S. Moringa oleifera Lam. leaf extract prevents early liver injury and restores antioxidant status in mice fed with high-fat diet. Indian J. Exp. Biol. 2012, 50, 404–412. [Google Scholar] [PubMed]
  112. Olorunnisola, O.S.; Bradley, G.; Afolayan, A.J. Protective effect of Tulbaghia violacea Harv. on aortic pathology, tissue antioxidant enzymes and liver damage in diet-induced atherosclerotic rats. Int. J. Mol. Sci. 2012, 13, 12747–12760. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, W.; Zhou, W.; Wang, B.; Zhu, H.; Ye, L.; Feng, M. Antioxidant effect of apolipoprotein A–I on high-fat diet-induced non-alcoholic fatty liver disease in rabbits. Acta Biochim. Biophys. Sin. 2013, 45, 95–103. [Google Scholar] [CrossRef] [PubMed]
  114. Melega, S.; Canistro, D.; de Nicola, G.R.; Lazzeri, L.; Sapone, A.; Paolini, M. Protective effect of Tuscan black cabbage sprout extract against serum lipid increase and perturbations of liver antioxidant and detoxifying enzymes in rats fed a high-fat diet. Br. J. Nutr. 2013, 110, 988–997. [Google Scholar] [CrossRef] [PubMed]
  115. Guerra, J.F.; Magalhaes, C.L.; Costa, D.C.; Silva, M.E.; Pedrosa, M.L. Dietary acai modulates ROS production by neutrophils and gene expression of liver antioxidant enzymes in rats. J. Clin. Biochem. Nutr. 2011, 49, 188–194. [Google Scholar] [CrossRef] [PubMed]
  116. Xia, X.; Ma, Y.; Xing, X.; Huang, C.; Li, L.; Gui, G.; Liu, Q.; Xue, S. Antioxidant and hepatoprotective effect of different extracts of guizhencao (herba bidentis bipinnatae) against liver injury in hyperlipidemia rats. J. Tradit. Chin. Med. 2013, 33, 518–523. [Google Scholar] [CrossRef]
  117. Quine, S.D.; Raghu, P.S. Effects of (−)-epicatechin, a flavonoid on lipid peroxidation and antioxidants in streptozotocin-induced diabetic liver, kidney and heart. Pharmacol. Rep. 2005, 57, 610–615. [Google Scholar] [PubMed]
  118. Cumaoglu, A.; Cevik, C.; Rackova, L.; Ari, N.; Karasu, C. Effects of antioxidant stobadine on protein carbonylation, advanced oxidation protein products and reductive capacity of liver in streptozotocin-diabetic rats: Role of oxidative/nitrosative stress. Biofactors 2007, 30, 171–178. [Google Scholar] [CrossRef] [PubMed]
  119. Njomen, G.B.; Kamgang, R.; Oyono, J.L.; Njikam, N. Antioxidant potential of the methanol-methylene chloride extract of Terminalia glaucescens leaves on mice liver in streptozotocin-induced stress. Indian J. Pharmacol. 2008, 40, 266–270. [Google Scholar] [PubMed]
  120. Zhou, J.Y.; Zhou, S.W. Protective effect of berberine on antioxidant enzymes and positive transcription elongation factor b expression in diabetic rat liver. Fitoterapia 2011, 82, 184–189. [Google Scholar] [CrossRef] [PubMed]
  121. Ramachandraiahgari, R.M.Y.; Somesula, S.R.; Adi, P.J.; Mannur, I.S.; Enamala, M.; Matcha, B. Protective role of ethanolic extract of aloe vera antioxidant properties on liver and kidney of streptozotocin-induced diabetic rats. Dig. J. Nanomater. Biostruct. 2012, 7, 175–184. [Google Scholar]
  122. Lei, S.; Liu, Y.; Liu, H.; Yu, H.; Wang, H.; Xia, Z. Effects of N-acetylcysteine on nicotinamide dinucleotide phosphate oxidase activation and antioxidant status in heart, lung, liver and kidney in streptozotocin-induced diabetic rats. Yonsei Med. J. 2012, 53, 294–303. [Google Scholar] [CrossRef] [PubMed]
  123. Singh, J.; Kakkar, P. Modulation of liver function, antioxidant responses, insulin resistance and glucose transport by Oroxylum indicum stem bark in STZ induced diabetic rats. Food Chem. Toxicol. 2013, 62, 722–731. [Google Scholar] [CrossRef] [PubMed]
  124. Mkhwanazi, B.N.; Serumula, M.R.; Myburg, R.B.; van Heerden, F.R.; Musabayane, C.T. Antioxidant effects of maslinic acid in livers, hearts and kidneys of streptozotocin-induced diabetic rats: Effects on kidney function. Ren. Fail. 2014, 36, 419–431. [Google Scholar] [CrossRef] [PubMed]
  125. Sadi, G.; Bozan, D.; Yildiz, H.B. Redox regulation of antioxidant enzymes: Post-translational modulation of catalase and glutathione peroxidase activity by resveratrol in diabetic rat liver. Mol. Cell. Biochem. 2014, 393, 111–122. [Google Scholar] [CrossRef] [PubMed]
  126. Ozbayer, C.; Degirmenci, I.; Kurt, H.; Ozden, H.; Civi, K.; Basaran, A.; Gunes, H.V. Antioxidant and free radical-scavenging properties of Stevia rebaudiana (Bertoni) extracts and L-NNA in streptozotocine-nicotinamide induced diabetic rat liver. Turk. Klin. Tip Bilim. Derg. 2011, 31, 51–60. [Google Scholar] [CrossRef]
  127. Ye, X.; Feng, Y.; Tong, Y.; Ng, K.M.; Tsao, S.; Lau, G.K.; Sze, C.; Zhang, Y.; Tang, J.; Shen, J.; et al. Hepatoprotective effects of Coptidis rhizoma aqueous extract on carbon tetrachloride-induced acute liver hepatotoxicity in rats. J. Ethnopharmacol. 2009, 124, 130–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Cheung, F.; Feng, Y.B.; Wang, N.; Yuen, M.F.; Tong, Y.; Wong, V.T. Effectiveness of Chinese herbal medicine in treating liver fibrosis: A systematic review and meta-analysis of randomized controlled trials. Chin. Med. 2012, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Rasool, M.K.; Sabina, E.P.; Ramya, S.R.; Preety, P.; Patel, S.; Mandal, N.; Mishra, P.P.; Samuel, J. Hepatoprotective and antioxidant effects of gallic acid in paracetamol-induced liver damage in mice. J. Pharmacy Pharmacol. 2010, 62, 638–643. [Google Scholar] [CrossRef]
  130. 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 PKC δ-GSK3 β pathway. Br. J. Pharmacol. 2011, 163, 1653–1665. [Google Scholar] [CrossRef] [PubMed]
  131. Fan, Y.J.; Rong, Y.; Li, P.F.; Dong, W.L.; Zhang, D.Y.; Zhang, L.; Cui, M.J. Genistein protection against acetaminophen-induced liver injury via its potential impact on the activation of UDP-glucuronosyltransferase and antioxidant enzymes. Food Chem. Toxicol. 2013, 55, 172–181. [Google Scholar] [CrossRef] [PubMed]
  132. Sabir, S.M.; Rocha, J.B.T. Water-extractable phytochemicals from Phyllanthus niruri exhibit distinct in vitro antioxidant and in vivo hepatoprotective activity against paracetamol-induced liver damage in mice. Food Chem. 2008, 111, 845–851. [Google Scholar] [CrossRef]
  133. Jothy, S.L.; Aziz, A.; Chen, Y.; Sasidharan, S. Antioxidant activity and hepatoprotective potential of polyalthia longifolia and cassia spectabilis leaves against paracetamol-induced liver injury. Evid.-Based Complement. Altern. Med. 2012, 2012, 561284. [Google Scholar] [CrossRef] [PubMed]
  134. Olaleye, M.T.; Akinmoladun, A.C.; Ogunboye, A.A.; Akindahunsi, A.A. Antioxidant activity and hepatoprotective property of leaf extracts of Boerhaavia diffusa Linn against acetaminophen-induced liver damage in rats. Food Chem. Toxicol. 2010, 48, 2200–2205. [Google Scholar] [CrossRef] [PubMed]
  135. Simeonova, R.; Vitcheva, V.; Kondeva-Burdina, M.; Krasteva, I.; Manov, V.; Mitcheva, M. Hepatoprotective and antioxidant effects of saponarin, isolated from Gypsophila trichotoma Wend. on paracetamol-induced liver damage in rats. BioMed Res. Int. 2013, 2013, 757126. [Google Scholar] [CrossRef] [PubMed]
  136. Xiang, Q.; Liu, Z.; Wang, Y.; Xiao, H.; Wu, W.; Xiao, C.; Liu, X. Carnosic acid attenuates lipopolysaccharide-induced liver injury in rats via fortifying cellular antioxidant defense system. Food Chem. Toxicol. 2013, 53, 1–9. [Google Scholar] [CrossRef] [PubMed]
  137. Catal, T.; Bolkent, S. Combination of selenium and three naturally occurring antioxidants administration protects d-galactosamine-induced liver injury in rats. Biol. Trace Elem. Res. 2008, 122, 127–136. [Google Scholar] [CrossRef] [PubMed]
  138. Banu, S.; Bhaskar, B.; Balasekar, P. Hepatoprotective and antioxidant activity of Leucas aspera against d-galactosamine induced liver damage in rats. Pharm. Biol. 2012, 50, 1592–1595. [Google Scholar] [CrossRef] [PubMed]
  139. Jaishree, V.; Badami, S.; Krishnamurthy, P.T. Antioxidant and hepatoprotective effect of the ethyl acetate extract of Enicostemma axillare (Lam). Raynal against CCl4-induced liver injury in rats. Indian J. Exp. Biol. 2010, 48, 896–904. [Google Scholar] [PubMed]
  140. Cerny, D.; Lekic, N.; Vanova, K.; Muchova, L.; Horinek, A.; Kmonickova, E.; Zidek, Z.; Kamenikova, L.; Farghali, H. Hepatoprotective effect of curcumin in lipopolysaccharide/-galactosamine model of liver injury in rats: Relationship to HO-1/CO antioxidant system. Fitoterapia 2011, 82, 786–791. [Google Scholar] [CrossRef] [PubMed]
  141. Zheng, Z.-W.; Song, S.-Z.; Wu, Y.-L.; Lian, L.-H.; Wan, Y.; Nan, J.-X. Betulinic acid prevention of d-galactosamine/lipopolysaccharide liver toxicity is triggered by activation of Bcl-2 and antioxidant mechanisms. J. Pharm. Pharmacol. 2011, 63, 572–578. [Google Scholar] [CrossRef] [PubMed]
  142. Ravikumar, V.; Shivashangari, K.S.; Devaki, T. Effect of Tridax procumbens on liver antioxidant defense system during lipopolysaccharide-induced hepatitis in d-galactosamine sensitised rats. Mol. Cell. Biochem. 2005, 269, 131–136. [Google Scholar] [CrossRef] [PubMed]
  143. Kockar, M.C.; Naziroglu, M.; Celik, O.; Tola, H.T.; Bayram, D.; Koyu, A. N-Acetylcysteine modulates doxorubicin-induced oxidative stress and antioxidant vitamin concentrations in liver of rats. Cell Biochem. Funct. 2010, 28, 673–677. [Google Scholar] [CrossRef] [PubMed]
  144. Avci, A.; Cetin, R.; Erguder, I.B.; Devrim, E.; Kilicoglu, B.; Candir, O.; Ozturk, H.S.; Durak, I. Cisplatin causes oxidation in rat liver tissues: Possible protective effects of antioxidant food supplementation. Turk. J. Med. Sci. 2008, 38, 117–120. [Google Scholar]
  145. Wang, B.J.; Lien, Y.H.; Su, C.L.; Wu, C.P.; Yu, Z.R. Fractionation using supercritical CO2 influences the antioxidant and hepatoprotective activity of propolis against liver damage induced by tert-butyl hydroperoxide. Int. J. Food Sci. Technol. 2006, 41, 68–75. [Google Scholar] [CrossRef]
  146. Tabassum, H.; Parvez, S.; Rehman, H.; Banerjee, B.D.; Raisuddin, S. Catechin as an antioxidant in liver mitochondrial toxicity: Inhibition of tamoxifen-induced protein oxidation and lipid peroxidation. J. Biochem. Mol. Toxicol. 2007, 21, 110–117. [Google Scholar] [CrossRef] [PubMed]
  147. Kim, S.J.; Kang, H.S.; Lee, J.H.; Park, J.H.; Jung, C.H.; Bae, J.H.; Oh, B.C.; Song, D.K.; Baek, W.K.; Im, S.S. Melatonin ameliorates ER stress-mediated hepatic steatosis through miR-23a in the liver. Biochem. Biophys. Res. Commun. 2015, 458, 462–469. [Google Scholar] [CrossRef] [PubMed]
  148. Ferraro, S.M.; Lopez-Ortega, A. Antioxidant activity of melatonin on fatty liver induced by ethionine in mice. Arch. Med. Vet. 2008, 40, 51–57. [Google Scholar]
  149. Feng, Y.; Wang, N.; Tong, Y.; Tsao, S. Berberine: An old drug but new use for liver diseases. Planta Medica 2012, 78, 1091–1091. [Google Scholar] [CrossRef]
  150. Feng, Y.B.; Wang, N.; Zhu, M.F.; Zhang, Z.J.; Tong, Y.; Tsao, S. Interdisciplinary approaches in study of Chinese medicines: Case of coptis. Int. J. Mol. Med. 2010, 26, S21–S21. [Google Scholar]
  151. Hwang, Y.P.; Choi, J.H.; Yun, H.J.; Han, E.H.; Kim, H.G.; Kim, J.Y.; Park, B.H.; Khanal, T.; Choi, J.M.; Chung, Y.C.; et al. Anthocyanins from purple sweet potato attenuate dimethylnitrosamine-induced liver injury in rats by inducing Nrf2-mediated antioxidant enzymes and reducing COX-2 and iNOS expression. Food Chem. Toxicol. 2011, 49, 93–99. [Google Scholar] [CrossRef] [PubMed]
  152. Lawal, A.O.; Lawal, A.F.; Ologundudu, A.; Adeniran, O.Y.; Omonkhua, A.; Obi, F. Antioxidant effects of heated garlic juice on cadmium-induced liver damage in rats as compared to ascorbic acid. J. Toxicol. Sci. 2011, 36, 549–557. [Google Scholar] [CrossRef]
  153. Choi, J.H.; Jin, S.W.; Kim, H.G.; Khanal, T.; Hwang, Y.P.; Lee, K.J.; Choi, C.Y.; Chung, Y.C.; Lee, Y.C.; Jeong, H.G. Platycodi Radix attenuates dimethylnitrosamine-induced liver fibrosis in rats by inducing Nrf2-mediated antioxidant enzymes. Food Chem. Toxicol. 2013, 56, 231–239. [Google Scholar] [CrossRef] [PubMed]
  154. Niture, S.K.; Jain, A.K.; Jaiswal, A.K. Antioxidant-induced modification of INrf2 cysteine 151 and PKC-δ-mediated phosphorylation of Nrf2 serine 40 are both required for stabilization and nuclear translocation of Nrf2 and increased drug resistance. J. Cell Sci. 2009, 122, 4452–4464. [Google Scholar] [CrossRef] [PubMed]
  155. Cheung, F.; Wang, X.B.; Wang, N.; Yuen, M.F.; Ziea, T.C.; Tong, Y.; Wong, V.T.; Feng, Y.B. Chinese medicines as an adjuvant therapy for unresectable Hepatocellular carcinoma during transarterial chemoembolization: A meta-analysis of randomized controlled trials. Evid.-Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, N.; Feng, Y.B. Elaborating the role of natural products-induced autophagy in cancer treatment: Achievements and artifacts in the state of the art. BioMed Res. Int. 2015, 2015. [Google Scholar] [CrossRef] [PubMed]
  157. Tan, H.Y.; Wang, N.; Tsao, S.W.; Zhang, Z.J.; Feng, Y.B. Suppression of vascular endothelial growth factor via inactivation of eukaryotic elongation factor 2 by alkaloids in coptidis rhizome in Hepatocellular carcinoma. Integr. Cancer Ther. 2014, 13, 425–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Xu, W.W.; Li, B.; Lai, E.T.; Chen, L.; Huang, J.J.; Cheung, A.L.; Cheung, P.C. Water extract from Pleurotus pulmonarius with antioxidant activity exerts in vivo chemoprophylaxis and chemosensitization for liver cancer. Nutr. Cancer 2014, 66, 989–998. [Google Scholar] [CrossRef] [PubMed]
  159. Wang, N.; Feng, Y.B.; Zhu, M.F.; Tsang, C.M.; Man, K.; Tong, Y.; Tsao, S.W. Berberine induces autophagic cell death and mitochondrial apoptosis in liver cancer cells: The cellular mechanism. J. Cell. Biochem. 2010, 111, 1426–1436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Wang, N.; Zhu, M.F.; Wang, X.B.; Tan, H.Y.; Tsao, S.W.; Feng, Y.B. Berberine-induced tumor suppressor p53 up-regulation gets involved in the regulatory network of MIR-23a in Hepatocellular carcinoma. Biochim. Biophys. Acta 2014, 1839, 849–857. [Google Scholar] [CrossRef] [PubMed]
  161. Kobayashi, H.; Tanaka, Y.; Asagiri, K.; Asakawa, T.; Tanikawa, K.; Kage, M.; Yagi, M. The antioxidant effect of green tea catechin ameliorates experimental liver injury. Phytomedicine 2010, 17, 197–202. [Google Scholar] [CrossRef] [PubMed]
  162. Spahr, L.; Bresson-Hadni, S.; Amann, P.; Kern, I.; Golaz, O.; Frossard, J.L.; Hadengue, A. Allopurinol, oxidative stress and intestinal permeability in patients with cirrhosis: An open-label pilot study. Liver Int. 2007, 27, 54–60. [Google Scholar] [CrossRef] [PubMed]
  163. Demirel, U.; Yalniz, M.; Aygun, C.; Orhan, C.; Tuzcu, M.; Sahin, K.; Ozercan, I.H.; Bahcecioglu, I.H. Allopurinol ameliorates thioacetamide-induced acute liver failure by regulating cellular redox-sensitive transcription factors in rats. Inflammation 2012, 35, 1549–1557. [Google Scholar] [CrossRef] [PubMed]
  164. Karaaslan, C.; Suzen, S. Antioxidant properties of melatonin and its potential action in diseases. Curr. Top. Med. Chem. 2015, 15, 894–903. [Google Scholar] [CrossRef] [PubMed]
  165. Wahab, M.H.; Akoul, E.S.; Abdel-Aziz, A.A. Modulatory effects of melatonin and vitamin E on doxorubicin-induced cardiotoxicity in Ehrlich ascites carcinoma-bearing mice. Tumori 2000, 86, 157–162. [Google Scholar] [PubMed]
  166. Baydas, G.; Canatan, H.; Turkoglu, A. Comparative analysis of the protective effects of melatonin and vitamin E on streptozocin-induced diabetes mellitus. J. Pineal Res. 2002, 32, 225–230. [Google Scholar] [CrossRef] [PubMed]
  167. Bai, X.; Qiu, A.; Guan, J.; Shi, Z. Antioxidant and protective effect of an oleanolic acid-enriched extract of A. deliciosa root on carbon tetrachloride induced rat liver injury. Asia Pac. J. Clin. Nutr. 2007, 16, 169–173. [Google Scholar] [PubMed]
  168. Shaker, E.; Mahmoud, H.; Mnaa, S. Silymarin, the antioxidant component and Silybum marianum extracts prevent liver damage. Food Chem. Toxicol. 2010, 48, 803–806. [Google Scholar] [CrossRef] [PubMed]
  169. Iliemene, U.D.; Atawodi, S.E.O. In vivo antioxidant and hepatoprotective effects of methanolic extract of dioclea reflexa seed in rats following acute or chronic liver injury. Bangladesh J. Pharmacol. 2014, 9, 112–117. [Google Scholar] [CrossRef]
  170. Jin, Y.S.; Lee, M.J.; Han, W.; Heo, S.I.; Sohn, S.I.; Wang, M.H. Antioxidant effects and hepatoprotective activity of 2,5-dihydroxy-4,3′-di(β-d-glucopyranosyloxy)-trans-stilbene Morus bombycis Koidzumi roots on CCl4-induced liver damage. Free Radic. Res. 2006, 40, 986–992. [Google Scholar] [CrossRef] [PubMed]
  171. Jin, Y.S.; Sa, J.H.; Shim, T.H.; Rhee, H.I.; Wang, M.H. Hepatoprotective and antioxidant effects of Morus bombycis Koidzumi on CCl4-induced liver damage. Biochem. Biophs. Res. Commun. 2005, 329, 991–995. [Google Scholar] [CrossRef] [PubMed]
  172. Kanter, M.; Coskun, O.; Budancamanak, M. Hepatoprotective effects of Nigella sativa L and Urtica dioica L on lipid peroxidation, antioxidant enzyme systems and liver enzymes in carbon tetrachloride-treated rats. World J. Gastroenterol. 2005, 11, 6684–6688. [Google Scholar] [CrossRef] [PubMed]
  173. Jayakumar, T.; Ramesh, E.; Geraldine, P. Antioxidant activity of the oyster mushroom, Pleurotus ostreatus, on CCl4-induced liver injury in rats. Food Chem. Toxicol. 2006, 44, 1989–1996. [Google Scholar] [CrossRef] [PubMed]
  174. Raja, S.; Ahamed, K.F.H.N.; Kumar, V.; Mukherjee, K.; Bandyopadhyay, A.; Mukherjee, P.K. Antioxidant effect of Cytisus scoparius against carbon tetrachloride treated liver injury in rats. J. Ethnopharmacol. 2007, 109, 41–47. [Google Scholar] [CrossRef] [PubMed]
  175. Chang, H.Y.; Peng, W.H.; Sheu, M.J.; Huang, G.J.; Tseng, M.C.; Lai, M.T.; Ho, Y.L.; Chang, Y.S. Hepatoprotective and antioxidant effects of ethanol extract from Phellinus merrillii on carbon tetrachloride-induced liver damage. Am. J. Chin. Med. 2007, 35, 793–804. [Google Scholar] [CrossRef] [PubMed]
  176. Naik, S.R.; Panda, V.S. Hepatoprotective effect of Ginkgoselect Phytosome® in rifampicin induced liver injurym in rats: Evidence of antioxidant activity. Fitoterapia 2008, 79, 439–445. [Google Scholar] [CrossRef] [PubMed]
  177. Bhattacharjee, R.; Sil, P.C. Protein isolate from the herb, Phyllanthus niruri L. (Euphorblaceae), plays hepatoprotective role against carbon tetrachloride induced liver damage via its antioxidant properties. Food Chem. Toxicol. 2007, 45, 817–826. [Google Scholar] [CrossRef] [PubMed]
  178. Lee, K.J.; Choi, J.H.; Jeong, H.G. Hepatoprotective and antioxidant effects of the coffee diterpenes kahweol and cafestol on carbon tetrachloride-induced liver damage in mice. Food Chem. Toxicol. 2007, 45, 2118–2125. [Google Scholar] [CrossRef] [PubMed]
  179. Lee, S.H.; Heo, S.I.; Li, L.; Lee, M.J.; Wang, M.H. Antioxidant and hepatoprotective activities of Cirsium setidens Nakai against CCl4-induced liver damage. Am. J. Chin. Med. 2008, 36, 107–114. [Google Scholar] [CrossRef] [PubMed]
  180. Wu, S.J.; Lin, Y.H.; Chu, C.C.; Tsai, Y.H.; Chao, J.C.J. Curcumin or saikosaponin a improves hepatic antioxidant capacity and protects against CCl4-induced liver injury in rats. J. Med. Food 2008, 11, 224–229. [Google Scholar] [CrossRef] [PubMed]
  181. Kumar, P.; Deval, R.G.; Lakshmayya; Ramachandra, S.S. Antioxidant and hepatoprotective activity of tubers of Momordica tuberosa Cogn. against CCl4 induced liver injury in rats. Indian J. Exp. Biol. 2008, 46, 510–513. [Google Scholar] [PubMed]
  182. Botsoglou, N.A.; Taitzoglou, I.A.; Botsoglou, E.; Zervos, I.; Kokoli, A.; Christaki, E.; Nikolaidis, E. Effect of long-term dietary administration of oregano and rosemary on the antioxidant status of rat serum, liver, kidney and heart after carbon tetrachloride-induced oxidative stress. J. Sci. Food Agric. 2009, 89, 1397–1406. [Google Scholar] [CrossRef]
  183. Singab, A.N.B.; Ayoub, N.A.; Ali, E.N.; Mostafa, N.M. Antioxidant and hepatoprotective activities of Egyptian moraceous plants against carbon tetrachloride-induced oxidative stress and liver damage in rats. Pharm. Biol. 2010, 48, 1255–1264. [Google Scholar] [CrossRef] [PubMed]
  184. Ganie, S.A.; Haq, E.; Masood, A.; Hamid, A.; Zargar, M.A. Antioxidant and protective effect of ethyl acetate extract of Podophyllum hexandrum Rhizome on carbon tetrachloride induced rat liver injury. Evid.-Based Complement. Altern. Med. 2011, 2011. [Google Scholar] [CrossRef] [PubMed]
  185. Gupta, V.K.; Gupta, M.; Sharma, S.K. Evaluation of antioxidant potential of Ficus religiosa (Linn.) roots against carbon tetrachloride-induced liver injury. J. Med. Plants Res. 2011, 5, 1582–1588. [Google Scholar]
  186. Paramesha, M.; Ramesh, C.K.; Krishna, V.; Kumar, Y.S.R.; Parvathi, K.M.M. Hepatoprotective and in vitro antioxidant effect of Carthamus tinctorious L, var Annigeri-2-, an oil-yielding crop, against CCl4-induced liver injury in rats. Pharmacogn. Mag. 2011, 7, 289–297. [Google Scholar] [PubMed]
  187. Sridevi, V.K.; Chouhan, H.S.; Singh, N.K.; Singh, S.K. Antioxidant and hepatoprotective effects of ethanol extract of Vitex glabrata on carbon tetrachloride-induced liver damage in rats. Nat. Prod. Res. 2012, 26, 1135–1140. [Google Scholar] [CrossRef] [PubMed]
  188. Chen, J.; Sun, H.N.; Sun, A.D.; Lin, Q.H.; Wang, Y.; Tao, X.Y. Studies of the protective effect and antioxidant mechanism of blueberry anthocyanins in a CC14-induced liver injury model in mice. Food Agric. Immunol. 2012, 23, 352–362. [Google Scholar] [CrossRef]
  189. Aksoy, L.; Sozbilir, N.B. Effects of Matricaria chamomilla L. on lipid peroxidation, antioxidant enzyme systems, and key liver enzymes in CCl4-treated rats. Toxicol. Environ. Chem. 2012, 94, 1780–1788. [Google Scholar] [CrossRef]
  190. Wei, J.F.; Li, Y.Y.; Yin, Z.H.; Gong, F.; Shang, F.D. Antioxidant activities in vitro and hepatoprotective effects of Lysimachia clethroides Duby on CCl4-induced acute liver injury in mice. Afr. J. Pharm. Pharmacol. 2012, 6, 743–750. [Google Scholar]
  191. Panda, V.S.; Ashar, H.D. Antioxidant and hepatoprotective effects of Garcinia indica choisy fruits in carbon tetrachloride-induced liver injury in rats. J. Food Biochem. 2012, 36, 240–247. [Google Scholar] [CrossRef]
  192. Al-Dbass, A.M.; Al-Daihan, S.K.; Bhat, R.S. Agaricus blazei Murill as an efficient hepatoprotective and antioxidant agent against CCl4-induced liver injury in rats. Saudi J. Biol. Sci. 2012, 19, 303–309. [Google Scholar] [CrossRef] [PubMed]
  193. Singhal, K.G.; das Gupta, G. Hepatoprotective and antioxidant activity of methanolic extract of flowers of Nerium oleander against CCl4-induced liver injury in rats. Asian Pac. J. Trop. Med. 2012, 5, 677–685. [Google Scholar] [CrossRef]
  194. Vuda, M.; D’Souza, R.; Upadhya, S.; Kumar, V.; Rao, N.; Kumar, V.; Boillat, C.; Mungli, P. Hepatoprotective and antioxidant activity of aqueous extract of Hybanthus enneaspermus against CCl4-induced liver injury in rats. Exp. Toxicol. Pathol. 2012, 64, 855–859. [Google Scholar] [CrossRef] [PubMed]
  195. Hou, F.L.; Zhang, R.F.; Zhang, M.W.; Su, D.X.; Wei, Z.C.; Deng, Y.Y.; Zhang, Y.; Chi, J.W.; Tang, X.J. Hepatoprotective and antioxidant activity of anthocyanins in black rice bran on carbon tetrachloride-induced liver injury in mice. J. Funct. Foods 2013, 5, 1705–1713. [Google Scholar] [CrossRef]
  196. Kalegari, M.; Gemin, C.A.; Araujo-Silva, G.; Brito, N.J.; Lopez, J.A.; Tozetto Sde, O.; Almeida, M.; Miguel, M.D.; Stien, D.; Miguel, O.G. Chemical composition, antioxidant activity and hepatoprotective potential of Rourea induta Planch. (Connaraceae) against CCl4-induced liver injury in female rats. Nutrition 2014, 30, 713–718. [Google Scholar] [CrossRef] [PubMed]
  197. Dai, N.; Zou, Y.; Zhu, L.; Wang, H.F.; Dai, M.G. Antioxidant properties of proanthocyanidins attenuate carbon tetrachloride (CCl4)-induced steatosis and liver injury in rats via CYP2E1 regulation. J. Med. Food 2014, 17, 663–669. [Google Scholar] [CrossRef] [PubMed]
  198. Yin, L.; Wei, L.; Fu, R.; Ding, L.; Guo, Y.; Tang, L.; Chen, F. Antioxidant and hepatoprotective activity of Veronica ciliata Fisch. extracts against carbon tetrachloride-induced liver injury in mice. Molecules 2014, 19, 7223–7236. [Google Scholar] [CrossRef] [PubMed]
  199. Abbas, A.T.; El-Shitany, N.A.; Shaala, L.A.; Ali, S.S.; Azhar, E.I.; Abdel-Dayem, U.A.; Youssef, D.T. Red sea Suberea mollis sponge extract protects against CCl4-induced acute liver injury in rats via an antioxidant mechanism. Evid.-Based Complement. Altern. Med. 2014, 2014, 745606. [Google Scholar] [CrossRef] [PubMed]
  200. Jalali Ghassam, B.; Ghaffari, H.; Prakash, H.S.; Kini, K.R. Antioxidant and hepatoprotective effects of Solanum xanthocarpum leaf extracts against CCl4-induced liver injury in rats. Pharm. Biol. 2014, 52, 1060–1068. [Google Scholar] [CrossRef] [PubMed]
  201. Meera, R.; Devi, P.; Kameswari, B.; Madhumitha, B.; Merlin, N.J. Antioxidant and hepatoprotective activities of Ocimum basilicum Linn. and Trigonella foenum-graecum Linn. against H2O2 and CCl4 induced hepatotoxicity in goat liver. Indian J. Exp. Biol. 2009, 47, 584–590. [Google Scholar] [PubMed]
  202. Saleh, D.O.; Abdel Jaleel, G.A.; El-Awdan, S.A.; Oraby, F.; Badawi, M. Thioacetamide-induced liver injury: Protective role of genistein. Can. J. Physiol. Pharmacol. 2014, 92, 965–973. [Google Scholar] [CrossRef] [PubMed]
  203. Abdulaziz Bardi, D.; Halabi, M.F.; Hassandarvish, P.; Rouhollahi, E.; Paydar, M.; Moghadamtousi, S.Z.; Al-Wajeeh, N.S.; Ablat, A.; Abdullah, N.A.; Abdulla, M.A. Andrographis paniculata leaf extract prevents thioacetamide-induced liver cirrhosis in rats. PLoS ONE 2014, 9, e109424. [Google Scholar] [CrossRef] [PubMed]
  204. Moustafa, A.H.; Ali, E.M.; Moselhey, S.S.; Tousson, E.; El-Said, K.S. Effect of coriander on thioacetamide-induced hepatotoxicity in rats. Toxicol. Ind. Health 2014, 30, 621–629. [Google Scholar] [CrossRef] [PubMed]
  205. Chang, Z.Y.; Lee, T.Y.; Huang, T.H.; Wen, C.K.; Chien, R.N.; Chang, H.H. Hepatoprotective effects of Ger-Gen-Chyn-Lian-Tang in thioacetamide-induced fibrosis in mice. J. Chin. Med. Assoc. 2014, 77, 360–366. [Google Scholar] [CrossRef] [PubMed]
  206. Zargar, S. Protective effect of Trigonella foenum-graecum on thioacetamide induced hepatotoxicity in rats. Saudi J. Biol. Sci. 2014, 21, 139–145. [Google Scholar] [CrossRef] [PubMed]
  207. Ramesh, T.; Sureka, C.; Bhuvana, S.; Hazeena Begum, V. Sesbania grandiflora diminishes oxidative stress and ameliorates antioxidant capacity in liver and kidney of rats exposed to cigarette smoke. J. Physiol. Pharmacol. 2010, 61, 467–476. [Google Scholar] [PubMed]
  208. Ozkan, A.; Fiskin, K.; Ayhan, A.G. Effect of vitamin E and selenium on antioxidant enzymes in brain, kidney and liver of cigarette smoke-exposed mice. Biologia 2007, 62, 360–364. [Google Scholar] [CrossRef]
  209. Singh, S.; Mondal, P.; Trigun, S.K. Acute liver failure in rats activates glutamine-glutamate cycle but declines antioxidant enzymes to induce oxidative stress in cerebral cortex and cerebellum. PLoS ONE 2014, 9, e95855. [Google Scholar] [CrossRef] [PubMed]
  210. Sutcu, R.; Altuntas, I.; Yildirim, B.; Karahan, N.; Demirin, H.; Delibas, N. The effects of subchronic methidathion toxicity on rat liver: Role of antioxidant vitamins C and E. Cell biol. Toxicol. 2006, 22, 221–227. [Google Scholar] [CrossRef] [PubMed]
  211. Khan, S.M. Protective effect of black tea extract on the levels of lipid peroxidation and antioxidant enzymes in liver of mice with pesticide-induced liver injury. Cell Biochem. Funct. 2006, 24, 327–332. [Google Scholar] [CrossRef] [PubMed]
  212. Banudevi, S.; Krishnamoorthy, G.; Venkataraman, P.; Vignesh, C.; Aruldhas, M.M.; Arunakaran, J. Role of α-tocopherol on antioxidant status in liver, lung and kidney of PCB exposed male albino rats. Food Chem. Toxicol. 2006, 44, 2040–2046. [Google Scholar] [CrossRef] [PubMed]
  213. Yener, Z.; Celik, I.; Ilhan, F.; Bal, R. Effects of Urtica dioica L. seed on lipid peroxidation, antioxidants and liver pathology in aflatoxin-induced tissue injury in rats. Food Chem. Toxicol. 2009, 47, 418–424. [Google Scholar] [CrossRef] [PubMed]
  214. Yogalakshmi, B.; Viswanathan, P.; Anuradha, C.V. Investigation of antioxidant, anti-inflammatory and DNA-protective properties of eugenol in thioacetamide-induced liver injury in rats. Toxicology 2010, 268, 204–212. [Google Scholar] [CrossRef] [PubMed]
  215. Khaki, A.A.; Khaki, A. Antioxidant effect of ginger to prevents lead-induced liver tissue apoptosis in rat. J. Med. Plants Res. 2010, 4, 1492–1495. [Google Scholar]
  216. Khan, R.A.; Khan, M.R.; Sahreen, S.; Shah, N.A.; Khan, A.M.; Khan, Y.M.; Bokhari, J.; Rashid, U.; Shabbir, B.A.M.; Saeed, N.; et al. Effect of various fractions of Launaea procumbens on antioxidant enzymes in rats liver: Oxidative stress induced by potassium bromate (KBrO3). Afr. J. Pharm. Pharmacol. 2012, 6, 512–515. [Google Scholar] [CrossRef]
  217. Zhang, Z.G.; Gao, L.; Cheng, Y.Y.; Jiang, J.; Chen, Y.; Jiang, H.J.; Yu, H.X.; Shan, A.S.; Cheng, B.J. Resveratrol, a natural antioxidant, has a protective effect on liver injury induced by inorganic arsenic exposure. BioMed Res. Int. 2014, 2014. [Google Scholar] [CrossRef] [PubMed]
  218. Maiti, S.; Chattopadhyay, S.; Acharyya, N.; Deb, B.; Hati, A.K. Emblica officinalis (amla) ameliorates arsenic-induced liver damage via DNA protection by antioxidant systems. Mol. Cell. Toxicol. 2014, 10, 75–82. [Google Scholar] [CrossRef]
  219. El Arem, A.; Saafi, E.B.; Ghrairi, F.; Thouri, A.; Zekri, M.; Ayed, A.; Zakhama, A.; Achour, L. Aqueous date fruit extract protects against lipid peroxidation and improves antioxidant status in the liver of rats subchronically exposed to trichloroacetic acid. J. Physiol. Biochem. 2014, 70, 451–464. [Google Scholar] [CrossRef] [PubMed]
  220. Kumar, D.J.; Santhi, R.J. Antioxidant and cytotoxic effects of hexane extract of Morinda pubescens leaves in human liver cancer cell line. Asian Pac. J. Trop. Med. 2012, 5, 362–366. [Google Scholar] [CrossRef]
  221. Mukti, N.A.; Sulaiman, S.; Saad, S.M.; Basari, J.M.H.; Rahman, M.A.; Ngah, W.Z.W.; Yusof, Y.A.M. Chlorella vulgaris exhibited antioxidant and antitumour effects against liver cancer in in vivo and in vitro studies. Sains Malays. 2009, 38, 773–784. [Google Scholar]
  222. Gupta, M.; Mazumder, U.K.; Kumar, R.S.; Sivakumar, T.; Gomathi, P.; Rajeshwar, Y. Antioxidant defense system induced by a methanol extract of Caesalpinia bonducella in rat liver. Pharm. Biol. 2005, 43, 411–419. [Google Scholar] [CrossRef]
  223. Serviddio, G.; Bellanti, F.; Stanca, E.; Lunetti, P.; Blonda, M.; Tamborra, R.; Siculella, L.; Vendemiale, G.; Capobianco, L.; Giudetti, A.M. Silybin exerts antioxidant effects and induces mitochondrial biogenesis in liver of rat with secondary biliary cirrhosis. Free Radic. Biol. Med. 2014, 73, 117–126. [Google Scholar] [CrossRef] [PubMed]
  224. Yu, D.K.; Zhang, C.X.; Zhao, S.S.; Zhang, S.H.; Zhang, H.; Cai, S.Y.; Shao, R.G.; He, H.W. The anti-fibrotic effects of epigallocatechin-3-gallate in bile duct-ligated cholestatic rats and human hepatic stellate LX-2 cells are mediated by the PI3K/Akt/Smad pathway. Acta Pharmacol. Sin. 2015, 36, 473–482. [Google Scholar] [CrossRef] [PubMed]
  225. Fahmy, S.R. Anti-fibrotic effect of Holothuria arenicola extract against bile duct ligation in rats. BMC Complement. Altern Med. 2015, 15, 14. [Google Scholar] [CrossRef] [PubMed]
  226. Mahmoud, M.F.; Zakaria, S.; Fahmy, A. Aqueous garlic extract alleviates liver fibrosis and renal dysfunction in bile-duct-ligated rats. Z. Naturforsch. C 2014, 69, 133–141. [Google Scholar] [CrossRef] [PubMed]
  227. Oguz, S.; Kanter, M.; Erboga, M.; Erenoglu, C. Protective effects of thymoquinone against cholestatic oxidative stress and hepatic damage after biliary obstruction in rats. J. Mol. Histol. 2012, 43, 151–159. [Google Scholar] [CrossRef] [PubMed]
  228. Galicia-Moreno, M.; Favari, L.; Muriel, P. Antifibrotic and antioxidant effects of N-acetylcysteine in an experimental cholestatic model. Eur. J. Gastroenterol. Hepatol. 2012, 24, 179–185. [Google Scholar] [CrossRef] [PubMed]
  229. Fursule, R.A.; Patil, S.D. Hepatoprotective and antioxidant activity of Phaseolus trilobus, Ait on bile duct ligation induced liver fibrosis in rats. J. Ethnopharmacol. 2010, 129, 416–419. [Google Scholar] [CrossRef] [PubMed]
  230. Ohta, Y.; Kongo, M.; Kishikawa, T. Melatonin exerts a therapeutic effect on cholestatic liver injury in rats with bile duct ligation. J. Pineal Res. 2003, 34, 119–126. [Google Scholar] [CrossRef] [PubMed]
  231. Kireev, R.; Bitoun, S.; Cuesta, S.; Tejerina, A.; Ibarrola, C.; Moreno, E.; Vara, E.; Tresguerres, J.A. Melatonin treatment protects liver of Zucker rats after ischemia/reperfusion by diminishing oxidative stress and apoptosis. Eur. J. Pharmacol. 2013, 701, 185–193. [Google Scholar] [CrossRef] [PubMed]
  232. Aldaba-Muruato, L.R.; Moreno, M.G.; Hernandez-Mercado, E.; Shibayama, M.; Muriel, P. Secondary biliary cirrhosis in the rat is prevented by decreasing NF-κB nuclear translocation and TGF-β expression using allopurinol, an inhibitor of xanthine oxidase. Can. J. Physiol. Pharmacol. 2012, 90, 1469–1478. [Google Scholar] [CrossRef] [PubMed]
  233. Kim, H.G.; Lee, J.S.; Lee, J.S.; Han, J.M.; Son, C.G. Hepatoprotective and antioxidant effects of Myelophil on restraint stress-induced liver injury in BALB/c mice. J. Ethnopharmacol. 2012, 142, 113–120. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Li, S.; Tan, H.-Y.; Wang, N.; Zhang, Z.-J.; Lao, L.; Wong, C.-W.; Feng, Y. The Role of Oxidative Stress and Antioxidants in Liver Diseases. Int. J. Mol. Sci. 2015, 16, 26087-26124. https://doi.org/10.3390/ijms161125942

AMA Style

Li S, Tan H-Y, Wang N, Zhang Z-J, Lao L, Wong C-W, Feng Y. The Role of Oxidative Stress and Antioxidants in Liver Diseases. International Journal of Molecular Sciences. 2015; 16(11):26087-26124. https://doi.org/10.3390/ijms161125942

Chicago/Turabian Style

Li, Sha, Hor-Yue Tan, Ning Wang, Zhang-Jin Zhang, Lixing Lao, Chi-Woon Wong, and Yibin Feng. 2015. "The Role of Oxidative Stress and Antioxidants in Liver Diseases" International Journal of Molecular Sciences 16, no. 11: 26087-26124. https://doi.org/10.3390/ijms161125942

Article Metrics

Back to TopTop