Next Article in Journal
The Beneficial Effect of Proanthocyanidins and Icariin on Biochemical Markers of Bone Turnover in Rats
Next Article in Special Issue
Hepatotoxicity: Molecular Mechanisms and Pathophysiology
Previous Article in Journal
Overexpression of OsGID1 Enhances the Resistance of Rice to the Brown Planthopper Nilaparvata lugens
Previous Article in Special Issue
Lipoic Acid Prevents High-Fat Diet-Induced Hepatic Steatosis in Goto Kakizaki Rats by Reducing Oxidative Stress Through Nrf2 Activation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Mechanisms Involved in Oxidative Stress-Associated Liver Injury Induced by Chinese Herbal Medicine: An Experimental Evidence-Based Literature Review and Network Pharmacology Study

School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(9), 2745; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19092745
Submission received: 28 July 2018 / Revised: 8 September 2018 / Accepted: 10 September 2018 / Published: 13 September 2018
(This article belongs to the Special Issue Hepatotoxicity: Molecular Mechanisms and Pathophysiology)

Abstract

:
Oxidative stress, defined as a disequilibrium between pro-oxidants and antioxidants, can result in histopathological lesions with a broad spectrum, ranging from asymptomatic hepatitis to hepatocellular carcinoma in an orchestrated manner. Although cells are equipped with sophisticated strategies to maintain the redox biology under normal conditions, the abundance of redox-sensitive xenobiotics, such as medicinal ingredients originated from herbs or animals, can dramatically invoke oxidative stress. Growing evidence has documented that the hepatotoxicity can be triggered by traditional Chinese medicine (TCM) during treating various diseases. Meanwhile, TCM-dependent hepatic disorder represents a strong correlation with oxidative stress, especially the persistent accumulation of intracellular reactive oxygen species. Of note, since TCM-derived compounds with their modulated targets are greatly diversified among themselves, it is complicated to elaborate the potential pathological mechanism. In this regard, data mining approaches, including network pharmacology and bioinformatics enrichment analysis have been utilized to scientifically disclose the underlying pathogenesis. Herein, top 10 principal TCM-modulated targets for oxidative hepatotoxicity including superoxide dismutases (SOD), malondialdehyde (MDA), glutathione (GSH), reactive oxygen species (ROS), glutathione peroxidase (GPx), Bax, caspase-3, Bcl-2, nuclear factor (erythroid-derived 2)-like 2 (Nrf2), and nitric oxide (NO) have been identified. Furthermore, hepatic metabolic dysregulation may be the predominant pathological mechanism involved in TCM-induced hepatotoxic impairment.

Graphical Abstract

1. Introduction

It appears in early evolution that oxidative stress commonly associates with either long-term degenerative diseases or acute ischemia–reperfusion injury [1,2,3,4,5,6]. The causes for these pathologies are generally inseparable from the shift of reactive oxygen species (ROS) from mediating normal physiological responses (i.e., redox biology) to invoke inevitable cellular dysfunctions through oxidative damage, especially inefficient oxidative phosphorylation in the mitochondria [7]. ROS, characterized as a byproduct of aerobic metabolism, primarily comprises superoxide anion, hydrogen peroxide, and hydroxyl radicals that confer reactivity to numerous targets in physiological and pathological processes. Despite the fact that mammals possess well-equipped antioxidant systems, including enzymes and non-enzyme antioxidants, it remains inadequate to normalize the severe hepatotoxicity induced by toxic substances with strong pro-oxidant properties.
Treatment strategies of traditional Chinese medicine (TCM) in alleviating different diseases, such as tumor proliferation, diabetic retinopathy, and liver dysfunctions, have had prolonged utilization and are generally considered as multitarget therapies with minimal adverse actions [8,9,10,11]. In contrast to the therapeutic effects, side effects of TCM, such as hepatotoxicity, have rarely been reported. However, accumulating evidence regarding TCM-associated oxidative hepatotoxicity was frequently addressed in recent years, particularly in paradoxical pharmacological activities of toxic and curative actions [12]. The most vital clue is the association between the initiation of TCM administration and hepatotoxicity generation and, of equal importance, to the deceleration following withdrawal. Concordantly, liver is the primary organ susceptible to pathological cascades of oxidative stress. In particular, parenchymal cells are most vulnerable in an oxidative environment. Abundant ROS are produced from mitochondria, and microsomes in parenchymal cells by regulating PPARα-associated signaling pathways. In addition, in Kupffer cells, the hepatic oxygen sensor and resident liver macrophage, has been postulated to trigger the formation of hepatic fibrosis by excessive ROS-induced apoptosis and inflammation [13]. Both hepatic stellate and endothelial cells are all specialized in producing ROS in physiological and pathological systems, and accustomed to suffering from lipid peroxidation [14]. Of note, although studies in the evaluation of mammal’s susceptibility to TCM-induced oxidative hepatotoxicity have been extensively demonstrated, specific identification of potential targets in relation to pathogenesis of TCM-dependent hepatotoxicity is limited. Hereby, we performed a literature review with network pharmacology, aiming to systematically decipher the highly pathogenic molecular targets for oxidative hepatotoxicity regulated by TCM.

2. Molecular Mechanisms Involved in TCM-Induced Oxidative Hepatotoxicity

2.1. Redox Status in Physiology and Pathology

Redox signaling is of essential importance to aerobic metabolism in regulating cell functions, including signal transduction pathways, defense in response to invading microorganisms, and gene expressions for cell physiological activity [15]. Oxidative stress is one of the key pathogenic processes that mainly associates to the disorder of redox homeostasis. A high concentration of redox signaling of ROS is commonly observed with cell damage and metabolic dysregulation, including lipid peroxidation, and irretrievable protein and DNA degeneration [16]. Moreover, the trigger of oxidative stress in a coordinated manner can disseminate the impairment to extrahepatic organs, including the failure of kidney, brain, and lung, which seem to indiscriminately oxidize almost all molecules in tissues [17,18,19]. However, under normal oxygen metabolism, ROS is perceived to be a molecular secondary messenger involved in the signal transduction mechanism in response to cytokines, hormones, and adenosine triphosphate (ATP), regulating the biological and physiological processes [20]. However, excessive ROS can be efficiently scavenged through intracellular redox homeostasis to maintain the cell metabolism and survival. The antioxidant system in our body is sensitive to the alterations in redox state for alleviating potential chain reactions of oxidative stress. Therefore, whether ROS is linked to orchestrated biological processes in routine metabolism or initiation of oxidative stress depends on the steady or imbalance of redox state. Aside from the beneficial effects, disruption of redox homeostasis, by chemicals in general, is correlated to the moderate-to-severe damage in organisms which, in turn, accelerates the progression of oxidative stress-related impairment [21].
To keep the generation of ROS controllable in liver, both enzymatic and non-enzymatic systems are in charge of maintaining the redox homeostasis. The steady-state cellular redox status can malfunction when exposed to pro-oxidant xenobiotics with toxic levels [22]. More specifically, redundant ROS is generally eliminated by a series of the following enzymes, primarily containing superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), whereas non-enzymatic molecules include glutathione (GSH), tocopherol, and beta-carotene. When the antioxidant system is vulnerable, the expression of activated ROS will be enhanced [23]. Ultimately, disruption of redox homeostasis is established with the undesirable elevated pro-oxidants. Subsequently, the physiological functions of several amino acids, such as cysteine, tyrosine, and tryptophan, are impaired. ROS-mediated pathological alterations in these amino acids are greatly susceptible to proteolytic attack via proteasomes in oxidized proteins, since proteins are composed of numerous amino acids and comparatively more prone to have specific targets of ROS [24]. An excess of ROS enhances mitochondrial permeability and functions. Reactive aldehydes, like 4-hydroxynonenal (4-HNE), can be released by ROS that inactivate the mitochondrial respiratory chain by hindering electron stream and activating oxidative stress [25].
In addition, pathological changes in sensitive proteins caused by oxidative stress can be a reversible or irreversible process. Reversible modifications are commonly implicated with cysteine for preventing the ROS-induced irreversible loss of cell function, including the carbonylation of lysine and arginine, Di-tyrosine generation, and protein–protein crosslinks, that exacerbate the degeneration and accumulation of proteins in cytoplasmic inclusions [26]. Therefore, regarding the prevention of oxidative stress in physiological circumstances, interference with superfluous pro-oxidant factors should be controlled through redox homeostasis system, which is typically composed of enzyme and non-enzymatic antioxidants.

2.2. Enzymatic and Non-Enzymatic System in Redox Homeostasis

The human body possesses sophisticated mechanisms to optimally maintain the redox homeostasis and cell functions against oxidative stress. This is achieved by the generation of antioxidative substances derived from endogenous antioxidant system or producing exogenous components of enzymatic and non-enzymatic antioxidants. Enzymatic reactions mediated by antioxidant endogenous enzymes ameliorate cellular oxidative stress-induced cell death principally through scavenging toxic substances, including overproduced intracellular ROS and reactive nitrogen species (RNS), aiming to stabilize the cellular contents of DNA and protein. As shown in Figure 1, the principal antioxidant enzymes concerning the neutralization of ROS are SOD, CAT, GPx, glutathione reductase (GR), and GRx. More specifically, SOD catalyzes the dismutation of superoxide peroxide (O2−) to oxygen (O2) and hydrogen peroxide (H2O2). H2O2 is decomposed into H2O and O2 by the action of CAT and GPx. In this process, CAT is an enzyme that can be found in almost all aerobiotic organs [27], whereas GPx enzymes transform H2O2 by using it to oxidize reduced GSH into oxidized glutathione (GSSG). GR, a flavoprotein (FAD-containing) enzyme, regenerates GSH from GSSG with the reducing power of nicotinamide adenine dinucleotide phosphate oxidase (NAD(P)H) [28,29].
Notably, accumulating superoxide is the vital substance that tends to be more correlated with oxidative stress, rather than redox signaling, in the oxygen consumption of this enzymatic system. Specifically, superoxide normally consists of the one-electron reduction of intracellular oxygen that can be converted by SOD into H2O2. SOD is the main defense substance against superoxide, including two isoforms as SOD1 and SOD2. SOD1 is a homodimeric protein termed Cu-ZnSOD, and mainly distributed in the cytosol as well as mitochondrial intermembrane space. SOD1 comprises the ions of copper and zinc in cytoplasm and nucleus [30]. Copper is necessary for catalytic reaction, whereas zinc is imperative for maintaining protein structure. However, SOD-2 is defined as manganese-associated SOD (MnSOD) and primarily exists in the mitochondrial matrix. Both two isoforms possess an identical mechanism of dismutation of O2− into H2O2.
Non-enzymatic antioxidants capable of rapidly inactivating free radicals and oxidants, can be categorized into two classifications, as follows: endogenous (metabolism) and exogenous (nutrient) antioxidants, in which metabolisms comprise glutathione, lipoic acid, and L-arginine, etc. In terms of their location, these proteins and molecules exert intracellular or extracellular therapeutic mechanisms to counteract excessive ROS and RNS. In addition, compounds that cannot be directly synthesized in vivo but in vitro, such as vitamin C, vitamin D, and trace metals, are identified as exogenous antioxidants [31]. As for vitamin C (ascorbic acid), it is a water-soluble, potent intra- and extracellular antioxidant that eliminates physiological free radicals, including hydroxyl and peroxyl radicals [32]. Moreover, GSH is an affluent thiol-based antioxidant inside cells, rich in live aerobic cells, and involved in both enzymatic and non-enzymatic reactions. As a cofactor for GPx, GSH catalyzes the reduction of H2O2 to H2O and O2. Furthermore, GSH can restrain the formation of the highly toxic substance hydroxyl radical through attenuating genomic instability and preventing lipids, proteins, and DNA from being indiscriminately oxidized [33,34].

2.3. Hepatotoxicity Caused by Specific Pro-Oxidant TCMs

Regarding the indispensable role of liver in the biotransformation of foods and medicines, hepatic disorders commonly result from the imbalance of metabolic homeostasis [35]. Although the ingestion of toxic substances, heterologous compounds, anticancer drugs, and immunosuppressive agents are known as the potential inducers of liver injury, growing evidence illustrates that long-term intake of curative drugs, such as certain anti-inflammatory and anticancer TCMs, may as well cause a large spectrum of hepatotoxicity including acute liver injury, steatosis-hepatitis, and fibrosis, etc. [36,37,38,39,40]. Furthermore, heightened reports of TCM-induced oxidative hepatic damage were particularly emphasized in recent studies [41]. In parallel, liver is the main organ of escalating ROS attack [42]. Although ROS may act in either positive or negative role on cellular functions in terms of the intensity and duration of oxidative stress, ROS production with an intoxicating dosage has been frequently detected by certain TCM treatments. Hepatotoxic TCM can irretrievably alter the biological functions of proteins, DNA contents, lipids, carbohydrates, membranes, which leads to oxidative stress-triggered hepatocyte injury [43]. However, the explicit mechanism by which overloaded ROS causes hepatic injury upon TCM treatment is not fully disclosed. Thus, searching effective approaches for identifying potential TCM with oxidative hepatotoxicity and underlying mechanisms are urgently demanded.

2.4. Literature Search Methodology

To further achieve an in-depth understanding of TCM-induced oxidative hepatotoxicity, series of the terms, including “Chinese medicine” or “Chinese herb” in combination with “hepatotoxicity” or “liver injury”, are firstly utilized to search the database of PubMed, Google Scholar, and Web of Science. After the initial exploration, in-text references related to these screening conditions will be selected manually. The keyword of “oxidative stress” will be further taken in the screening filter. Finally, literature that matched with all aforementioned criteria are accepted, otherwise, the articles were considered irrelevant and excluded. In addition, studies within the past 5 years are incorporated into the construction of both Table 1 and network pharmacology, which provide the up-to-date comprehension of the role of TCM-dependent oxidative hepatotoxicity. Several hepatotoxic TCMs with intensive elucidation are discussed in the following sections.

2.5. Pure Compounds

Tetrandrine, a principal alkaloid isolated from Stephania tetrandra, has revealed multiple therapeutic effects on rheumatism, glaucoma, myocardial infarction, and tumor treatments [44,45,46,47]. However, it is noteworthy that tetrandrine-related liver toxicity has been reported, and overexpression of ROS and disorder of mitochondrial permeability transition (MPT) was found to associate with tetrandrine-induced liver toxicity. MPT is a vital pathogenetic mechanism of drug-induced liver failure, and was identified by the overloaded intramitochondrial Ca2+-induced progressive permeabilization of the inner mitochondrial membrane, resulting in mitochondrial swelling and membrane rupture [48]. Besides, GSH depletion, as well as the activation of the pro-oxidant enzyme cytochrome P450 (CYP450) and especially cytochrome P2E1 (CYP2E1), was observed in tetrandrine-treated hepatocytes. Of note, chronic administration of tetrandrine for more than 3 months, with the dose ranging from 2 to 5 mg/kg, sensitized hepatocytes to oxidative damage [44].
Isoline and retrorsine are the pyrrolizidine alkaloid derived from the Chinese medicine Ligularia duciformis [49]. In spite of the curative effect of anti-inflammation and blocking cough reflex, pyrrolizidine alkaloid is commonly believed to be a representative poisonous alkaloid that disturbs the metabolism of numerous organs, especially in the liver [50]. Potential intoxication of enhanced serum alanine transaminase (ALT) and aspartate aminotransferase (AST) can be detected in clinical application as an antitussive agent. Furthermore, increased expression of GPx-1 and GST-Pi can be recognized in isoline-treated mouse liver, indicating that the self-defense to counteract oxidative stress in the body is activated, because both GPx-1 and GST-Pi are GSH-associated antioxidant enzymes. Concomitantly, upregulated malondialdehyde (MDA) and ROS, in combination with the degradation of glycine N-methyltransferase (maintaining contents of cellular folate), GPx, and CAT can be observed as well, alterations which further confirmed the soline-induced oxidative stress in hepatotoxicity [51]. Severe hepatic GSH depletion can take place as a result of the excessive production of dehydro-retrorsine generated from P450-modulated metabolic activation of retrorsine with the toxic dose of 0.2 mmol/kg, along with the abundant serum AST and ALT. Meanwhile, as covalent binding of reactive metabolites plays a vital role in the mechanism of toxic actions, higher rates of pyrrole–protein adduction than the vehicle group were observed in retrorsine-treated mouse, indicating that additional convincing evidence of Ligularia duciformis-caused hepatotoxicity has been provided [52,53,54].
Geniposide, an iridoid glycoside in Gardenia jasminoides, exerts anti-fibrotic, anti-osteoarthritis, and anti-epilepsy actions by modulation of the expression of transforming growth factor-β (TGF-β)/Smad4, p38 mitogen-activated protein kinase (p38 MAPK), and PI3K/Akt/GSK-3beta signaling pathways, respectively [55,56,57]. Whereas, geniposide manifested a number of pathological phenomena in the liver of SD rat with the dose greater than 574 mg/kg, such as elevated MDA, liver enzymes (ALT, AST, alkaline phosphatase (ALP), total bilirubin), focal necrosis, and downregulated SOD, leading to the onset of oxidative stress caused hepatotoxicity [58]. Meanwhile, exposure to geniposide less than 574 mg/kg was considered non-toxic to the liver, according to the unaltered serum biochemical indicators and liver weight [58].
Saikosaponins, an oleanane-type triterpenoid saponins, is the major bioactive ingredient extracted from Radix bupleuri, which has been used to prevent Alzheimer’s, pulmonary diseases, and even viral hepatitis [59,60,61]. However, in accordance with the cumulative evidence, saikosaponins probably contributed to toxicity in hepatocytes, and in particular, caused acute liver injury with doses of more than 19 g/day for a human being with 70 kg body weight. Metabolic dysregulation of lipids and proteins can be taken place due to excess ROS generation in the treatment of saikosaponins. Saikosaponins dose- and time-dependently evoked the increase of AST, ALT, and lactate dehydrogenase (LDH) [62]. CYP2E1, an important member of cytochrome P450 mixed function oxidase enzymes, plays a critical role in the metabolism of xenobiotics. Abundant ROS was detected, along with the upregulating CYP2E1 that linked to the dysregulated lipid metabolism upon saikosaponins treatment. Saikosaponin-induced oxidative stress was further proved by the dose-dependent depletion of GSH and elevation of MDA and inducible nitric oxide synthase (iNOS) [62].
Vincristine, a major compound of Catharanthus roseus, has been demonstrated to potently attenuate a series of malignancies, including colon cancer, metastatic breast cancer, and rhabdomyosarcoma [63,64,65]. The most therapeutic mechanism of inducing tumor death by vincristine is to break the polymerization of mitotic spindle microtubules and continuous arrests cell division during metaphase. Regarding the signs of vincristine-induced hepatotoxicity, increased levels of serum ALT, ALP, and AST are revealed in relation to the altered liver architecture. Moreover, hepatic content of MDA is enhanced, along with the significant decrease of hepatic SOD, GPx, reduced GSH (GSHr), GST, indicating that oxidative stress is established. Concomitantly, higher mRNA expressions of interleukin-12 (IL-12), interleukin-14 (IL-14), Bax, p53, and cleaved caspase-3 are simultaneously observed in hepatocytes, which stand for the induction of apoptosis. Reduced intracellular mRNA levels of Bcl-2 can be measured as well, suggesting that uptake of vincristine will invoke ROS-triggered apoptotic and inflammatory effects in liver tissues and cells [66].
Oxymatrine is a quinolizidine alkaloid isolated from the Chinese medicine Sophora flavescens and adopted to treat chronic viral hepatitis, plaque psoriasis, and arrhythmia [67,68,69]. Besides the findings of beneficial actions, it is proved to worsen liver damage. When treated with oxymatrine, cell viability will be reduced while the rate of apoptosis will be enhanced, as the intracellular markers for apoptotic pathway containing pro-caspase-3, -8, -9, and Bax are increasing, associated with the decrease of Bcl-2. In addition, expressions of endoplasmic reticulum (ER) stress indicators have been altered as well, including the activation of glucose regulated protein (GRP78), C/EBP homologous protein (CHOP), cleaved caspase-4, phospho-c-Jun N-terminal kinase (p-JNK), inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6) and pancreatic ER kinase (PERK). If there is interference with ER stress inhibition, intercellular levels of ROS, p-JNK, and cleaved caspase-3 will be dramatically dropped. As a consequence, hepatotoxicity of oxymatrine is linked with ER stress-induced ROS production [70].
Triptolide, triptonide, and wilforgine, three principal ingredients of Tripterygium wilfordii, elicit their pharmacodynamics in anti-inflammatory and anti-autoimmune diseases, especially rheumatoid arthritis [71]. At present, the clinical application of Tripterygium wilfordii is confined on account of its narrow therapeutic window and the high incidence of severe hepatotoxicity and nephrotoxicity [72]. Metabolic pathways for triptolide and triptonide are hydroxylation and cysteine conjugation, whereas wilforgine undergoes oxidative metabolism and hydrolysis. However, triptolide was demonstrated to be the major contributor to Tripterygium wilfordii-induced hepatotoxicity through hydroxylated by cytochrome P3A (CYP3A) [73], since CYP3A was specifically picked out as the main isoform responsible for triptolide hydroxylation and activates hepatic P450 which facilities the aggravation of liver toxicity [74,75]. Meanwhile, metabolic eliminations of GSH, as well as the reduction of NAD(P)H and quinine oxidoreductase 1 (NQO1), was verified in triptolide-treated mouse, indicating the dysfunction of the defense system to scavenge the oxidative species [76,77]. The nuclear factor erythroid 2-related factor 2 (Nrf2) is a regulator of cell resistance to pro-oxidant substances, and further mediates the antioxidant response components to control the physiological and pathophysiological outcomes of oxidant exposure. Triptolide-induced hepatotoxicity not only contains the blockade of cytoplasmic and nuclear Nrf2 activation, but inhibits the transition of hepatic influx and efflux transporters responsible for the interchanges of compound across cell membranes, such as P-gp, multidrug resistance-associated protein 2 (MRP2), multidrug resistance protein 4 (MRP4), bile salt export pump, and organic anion transporting polypeptide 2 (OATP2), at transcription level, which exerts more significant efficiency than that at functional level [76]. Meanwhile, blood biochemical levels of ALT and AST are elevated in combination with the failure in hematopoiesis, reproductive, renal, and cardiac systems through 0.2 mg/kg triptolide administration [78].

2.6. Herbal Extracts

Polygonum multiflorum (Heshouwu in Chinese) has been used in China for centuries, with anticancer, anti-inflammation, and reinforcing kidney function effects [79,80,81], However, accumulating evidence of liver cell damage arising from Polygonum multiflorum consumption has been reported, especially acute injury in morphological alteration in a time-dependent manner [82]. In 2013, the department of China Drug and Food Administration released a warning public announcement that more than 100 case reports concerning hepatotoxicity with Polygonum multiflorum treatment Investigation of Liver Injury of Polygonum multiflorum Thunb. Thus, a scientific basis for the toxicological mechanism of Polygonum multiflorum needs to be clarified. Polygonum multiflorum has two medicinal forms, Polygoni multiflori radix and Polygoni multiflori radix prapaerata. Notably, there is growing interest in the paradoxical effect of Polygonum multiflorum regarding whether it is hepatotoxic or not. Both forms share identical therapeutic effects in combating nonalcoholic fatty liver disease (NAFLD), fibrosis, as well as the cirrhosis, when the daily intake is less than 6 g per person [70,83,84]. The frequent mechanism of hepatotoxicity for both forms may contain cell cycle arrest and facilitate the activities of ALT, AST, ALP, creatinine, total bilirubin (TBil), direct bilirubin (DBil), and indirect bilirubin (IBil), and the leakage of LDH, whereas drug metabolic enzymes of cytochrome P3A4 (CYP3A4), cytochrome P2C19 (CYP2C19), CYP2E1, and SOD are attenuated by different pharmacokinetic behaviors [70,85,86,87]. The principal ingredients of Polygonum multiflorum include emodin-O-hex-sulfate, tetrahydroxystilbene-O-(galloyl)-hex. Among these, emodin and cis-stilbene glucoside might be the major responsibilities for liver toxicity [87]. In particular, cis-stilbene glucoside extracted from Polygonum multiflorum can trigger immunological idiosyncratic liver dysfunction in rats with lipopolysaccharide (LPS) intervention by repressing peroxisome proliferator-activated receptor (PPAR-γ) [88]. Aside from that, the ethanol extract is more toxic than the aqueous extract [89]. A method of prolonged decoction was convincingly demonstrated to effectively detoxicate Polygonum multiflorum [59]. Based on these alterations in biomarkers, Polygonum multiflorum-induced disturbance in the metabolic process of fat, bile acid, and amino acid may be the dominant threats to the induction of oxidative hepatotoxicity.
Evodiae fructus, an eminent Chinese herbal medicine, demonstrated its therapeutic capabilities in regard to anti-analgesic antiemetic and anti-inflammatory effects in gastrointestinal and cardiovascular diseases [90,91]. Alkaloids, including evodiamine and rutaecarpine, are the major bioactive ingredient in Evodiae fructus, and were reported to alleviate colorectal carcinoma, atherosclerosis and cardiovascular relaxation [92,93]. Nevertheless, the risk of increasing hepatotoxicity in patients treated with Evodiae fructus was frequently reported. Aqueous extract of Evodiae fructus can distinctly cause MPT in liver mitochondria, enhance the levels of AST, ALT, nitric oxide (NO), nitric oxide synthase (NOS), and MDA and downregulate the levels of MnSOD, GSH, and GPx, which synergistically lead to the occurrence of oxidative stress [94]. The cessation of ATP synthesis in association with induction of cytochrome C (CytC) release in hepatocytes was demonstrated in Evodiae fructus-dependent treatment, indicating the threshold of mitochondrial oxidative damage in liver [95,96].
Genkwa flos is characterized as the flower bud of Daphne genkwa and classified into the Chinese Pharmacopoeia with its wide range of pharmacodynamics actions including anti-herpes, anticancer, and inflammation-related symptoms [97,98,99]. Prior chemical studies have shown that Genkwa flos encompasses various types of constituents involving flavonoids, diterpenoids, lignans, and coumarins [100]. Current emerging evidence indicates that severe lesions to cardiac, renal, hepatic, and cutaneous tissues can be identified by excessive and prolonged administration of Genkwa flos [101,102,103,104]. Hepatotoxicity of Genkwa flos treatment have shown in HL-7702 liver cells, such as the increased hepatic serum makers of ALT, AST, and MDA, and downregulated oxidative stress indexes of CAT and GSH. In particular, phospholipase A2 (PLA2)/lysophosphatidylcholine (LPC) pathway is one of the crucial metabolic pathways involved in the glycerol phospholipid metabolism and participates in oxidative and inflammatory-induced liver injury [105]. It is remarkable that both the contents of PLA2 and LPC are significantly enhanced in Genkwa flos-treated HL-7702 liver cells [100]. The disruption of S1P metabolism mainly focused on Sphk/S1P pathway may occupy a vital position in Genkwa flos-related liver injury, as well [100]. Apart from that, chloroform extract of Genkwa flos demonstrated an inhibitory effect on the transcriptional activity of uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and serum bilirubin, which can enhance the susceptibility to oxidative stress-induced chromosomal aberration during the liver injury [106].
Cassia occidentalis, mainly cultivated in the south of China and Asia, is adopted as a moderate purgative and stomachic herbal medicine. The ethanol extract of Cassia occidentalis has numerous therapeutic activities, including anti-inflammation and anti-anaphylaxis [107]. Several toxic reactions, including loss of weight, myopathy, and hepatocellular necrosis, have been noticed in animals fed with Cassia occidentalis [108,109]. In 2008, WHO reported that intermittent exposure to Cassia occidentalis can ultimately cause hepatomyoencephalopathy with failed muscle, hepatic, and cerebral dysfunction [110]. The transcriptional profiling demonstrated that gene expressions of antioxidant enzymes containing CAT and SOD have been reduced in Cassia occidentalis-treated rats. On the contrary, significant decreases in GPx and GSH expressions have been proofed, along with concomitant increase of lipid peroxidation, which re-emphasizes the involvement of oxidative stress [111,112]. Furthermore, profound reductions in xenobiotic-metabolizing enzymes, including CYP1A1, CYP1A2, CYP2B1, GST, and quinone reductase, have been validated in combination with the impairment of carbohydrate metabolism in Cassia occidentalis-treated rats, as well. Growing apoptotic and inflammatory factors, including Bax, caspase-3, NF-κB, TNF-α, Akt, TGF-β, MAPK-9 and -14, IL-6, JNK, p-38, and FasL, have been observed by microarray analysis in the hepatic tissue of rats fed with Cassia occidentalis, suggesting that multiple pathological pathways are involved and contribute to hepatocyte death [111]. Interestingly, acute administration of Cassia occidentalis alleviates oxidative stress in the kidney of Rattus norvegicus, which initially aims to treat hypertension by enhancing urinary excretion with the elimination of Na+, Cl, and K+. Therefore, exact evidence for the underlying profile of Cassia occidentalis with hepatotoxicity deserves further studies [113].

3. Network Pharmacology-Associated Study

3.1. Network Construction and Targets Discovery

The general analogy of pharmacological actions is like a selective “key” that integrates into the specific “lock” of a drug target. The conception of creating selective ligands to counter undesirable adverse actions has been the dominant paradigm during the innovation of drug discovery. Nevertheless, post-genomic biology has revealed that the proposed schematic of drug action is far more complicated, suggesting that not only various “keys” may be suitable for a single “lock” but one “key” to fit multiple “locks”. Drug actions elaborated by means of network pharmacology can give insights into the pharmacological activities of hepatotoxic TCM, rather than separately enumerate case studies. As an innovative screening approach of drug target identification, network pharmacology prioritizes targets for TCM-induced oxidative hepatotoxicity in a systematic manner, aiming to identify the major pathological mechanism in hepatocytes.
With regard to plotting the visualized network figure, the literature that meets the screening criteria mentioned in the section of “Literature Search Methodology” will be enrolled in the construction of a network. After a large-scale screening, the qualified data are imported in a bioscience software termed “Cytoscape” for network analysis (free access online at “http://www.cytoscape.org/”) [140,141]. Components of nodes in the network (Figure 2) refer to specific factors, including TCM-derived extracts, pure compounds, and TCM-modulated targets (protein or gene), whereas edges that straightforwardly interconnect nodes stand for TCM–target interactions. Those nodes with more shared edges and centripetal position account are more efficacious than those with less. For providing a deeper insight, network analysis has been performed by a statistical plug-in “NetworkAnalyzer” attached in Cytoscape, to calculate the correlation degree for each node with a specific parameter, along with typical size and color. More specifically, as shown in Figure 2, the larger and brighter the node is, the greater the degree of correlation is, which allows for the perception of differentiating the relativity of each node with oxidative hepatotoxicity through the color gradient from bright to dark (i.e., brightest-green; middle-yellow; darkest-red). Noticeably, since the serum aminotransferases of ALT, AST, and ALP are well-accepted indicators for reflecting the severity of liver diseases directly, the measurement of these three targets exists widely in the majority of liver disease studies, which results in the extremely high value of the degree of correlation [142]. Although we cannot postulate that oxidative hepatotoxicity is not associated with the alteration of ALT, AST, and ALP, these three serum aminotransferases with high correlation values will greatly interfere with the judgment of ranking other targets in relation to oxidative liver injury. Thus, during the statistical processing, these three factors will not be enrolled in the ranking of the TCM-regulated targets susceptible to oxidative stress-induced hepatotoxicity. Based on exquisitely programmed procedures by network pharmacology, the top 10 high influential factors modulated by TCM have been shown in Figure 3B, and are ranked in order as the following: SOD, MDA, GSH, ROS, GPx, Bax, caspase-3, Bcl-2, Nrf2, and NO, which play pivotal roles in the initiation and propagation of oxidative hepatotoxicity.

3.2. Hepatotoxic Role of Identified TCM-Regulated Targets by Network Pharmacology

3.2.1. SOD, MDA, GSH, ROS, and GPx

Although oxidative hepatotoxicity caused by various medicines has been verified in numerous clinical trials, reports regarding the vital role of TCM-induced hepatic oxidative damage in patients are far away from satisfactory. In accordance with the principal targets picked up by NetworkAnalyzer, SOD is the most susceptible and regulated target for TCM-induced hepatic oxidative damage. When hepatocytes are attacked by TCM-derived compounds like atractyloside and saikosaponins, SOD will fail in preventing O2 overproduction when cell sensibility to hepatic injury is impaired by inhibiting the formation of peroxynitrite [96,143]. Serum levels of antioxidant enzymes, like SOD1, GPx, and CAT, are decreased in a patient with chronic liver cirrhosis and viral hepatitis when compared to healthy individuals. Inversely, indicators of oxidative stress and liver damage, such as MDA, NO, and ALT, are increased [144]. Upregulated ROS production in the formation of alcoholic liver disease (ALD) facilities the decreased of Cu-ZnSOD activity in association with the positively regulated NAD(P)H oxidase [145,146]. Interestingly, in certain special cases, even if the SOD content is steady or increased under pathological conditions, the possibility of defective SOD-induced oxidative damage may occur, since growing reports demonstrate that mutant forms of SOD may be generated, for example, in amyotrophic lateral sclerosis (ALS) disease. Mutations of SOD are proposed to modify the antioxidant proteins into pro-oxidants which are capable of invoking oxidative injury. However, whether this mechanism may exist in the development of hepatotoxicity or not, further studies are needed [144,147]. As for MDA, similar to 4-HNE, it is the end product of lipid peroxidation, representing a credible biomarker of oxidative stress. The formation of MDA can be extensively detected in ROS-caused degradation of polyunsaturated lipids [148]. Serum MDA levels usually increase in proportion to the severity of oxidative damage. Hereby, measuring MDA content may be considered as a reliable reference point for the degree of hepatic tissue impairment with oxidative stress. In addition, oxidative stress-related cirrhosis has been demonstrated to usually be linked with the negative regulation of MDA, SOD, GSH, and CAT [149]. ROS, undoubtedly acting as a dominant role in aerobic life, can be responsible for the manifestation of chronic liver disorders and stimulating their deterioration simultaneously. Referring to my previous description, ROS consists of various species, such as superoxide anion, hydrogen peroxide, and hydroxyl radicals. All of these substances possess specific inherent chemical characteristics and interact with different physiological and pathological targets. Hepatic cells, including Kupffer, endothelial, and stellate cells, are more susceptible to oxidative stress-induced apoptosis, necrosis, and tumorigenesis, especially by overproduced ROS [150].

3.2.2. Bax, Caspase-3, and Bcl-2

Coincidentally, Bax, caspase-3, and Bcl-2 are all represented as notable biomarkers in cell apoptosis, particularly in mitochondrial apoptotic pathway [151]. It is noteworthy that mitochondrial permeability as well as transition potential can be disturbed by continuous ROS generation, phenomena which may not only result in the release of apoptotic inductors, such as Bax, caspase-3 and cytochrome C, but also the downregulation of Bcl-2, degradation of mitochondrial DNA, and dysregulation of ATP synthesis in an oxidative phosphorylation system [152,153,154]. In detail, Bcl-2 family members are evolutionarily conserved modulators for programmed cell death termed “apoptosis”, and both pro-apoptotic (i.e., Bax) and antiapoptotic (i.e., Bcl-2) members are affiliated in the same family. The ratio of Bax to Bcl-2 is broadly considered as a rheostat to measure the cell susceptibility to apoptosis. Caspase-3 is a cysteine protease that mediates apoptosis by proteolysis of particular substrates, especially by Bax/Bcl-2 [155]. Interestingly, in addition to being the downstream substrate of caspase-3, Bcl-2, with its antiapoptotic property, can be inactivated by caspase-3 and converted into a pro-apoptosis motivator unrelated to Bax/Bcl-2 pathway, suggesting that a feedback loop between Bcl-2 and caspase-3 exists [156]. Considerable evidence supports the idea that enhanced Bax/Bcl-2 ratio, combined with cleavage of caspase-3, takes place in various liver diseases, including chronic hepatitis, alcoholic liver, and hepatocellular carcinoma (HCC) [157,158,159].

3.2.3. Nrf2 and NO

Nrf2, also known as nuclear factor (erythroid-derived 2)-like 2, is the principal modulator of encoding genes that protect cells against electrophilic stress. Thus, Nrf2 is comparatively less active in cells without stress, and coordinated with the basal flow of endogenic electrophile [160]. Activated Nrf2 in an oxidative environment not only illustrates the increasing synthesis of nucleophiles, including GSH and thioredoxin but, also, the enzyme-relevant catalysis for redox transitions [161]. As a sensor of cellular redox state, Nrf2 usually binds to Kelch-like epichlorohydrin-associated protein 1(KEAP1) in the normal physiological state. Increasing ROS, as well as electrophiles, can result in the release of Nrf2 into the nucleus, in order to activate the transcription of cell-protective genes. Antioxidant response element (ARE) can be activated to invoke antioxidant genes during oxidative stress [162]. However, ARE-associated genes, primarily regulated by Nrf2, govern GSH homeostasis and the activities of NAD(P)H quinone oxidoreductase 1 and uridine 5′-diphospho-glucosyltransferase [163]. Therefore, reduction of Nrf2 may make cells less sensitive to oxidative stress. Growing investigations have drawn attention to the alteration of Nrf2ARE feedback loop in counteracting the progression of various liver disorders including viral hepatitis, cirrhosis, hepatocellular carcinoma, and nonalcoholic liver disease [164,165,166,167]. Furthermore, this feedback loop associates with liver regeneration, as well. Overproduced ROS, derived from mitochondria, can be observed in Nrf2-knockout mice fed with pro-oxidant hepatotoxins or high-fat diet, along with more susceptibility to hepatotoxicity. Therefore, targeting Nrf2 as a curative target to treat oxidative stress-related liver diseases is meaningful and promising [168]. As for NO, it is a short-time lived free radical in gaseous state. It plays a variety of roles in the liver and other organs [168]. In healthy liver, NO production primarily originates from endotheliocytes by endothelial NO synthase (eNOS), and the low flow of NO is documented to adequately support the perfusion of liver sinusoids though regulating vascular tone and permeability [169]. Apart from that, endotheliocytes maintain sinusoid perfusion though upregulating NO generation. NO also regulates leukocyte adherence to sinusoidal endothelial cells, which is associated with attenuating the aggregation and adhesion of platelets [170]. In line with accelerating research studies, the expressions of NO and iNOS are positively regulated in almost all hepatocytes containing hepatic stellate cells under chronic hepatitis and endotoxemia [171,172]. In certain circumstances, NO has been deemed either a cytoprotective or a cytotoxic reagent, which depends on the local productive ratio of reactive oxygen intermediates (i.e., oxygen-centralized free radicals). Nevertheless, the predominant role of NO, in combination with the low contents of reactive oxygen intermediates is inclined towards the protective effect [173,174]. However, NO donors are reported to alleviate liver necrosis mainly by suppressing the levels of hydroxyl radical and lipid peroxidation, processes which can be blocked by the interaction between NO and reactive oxygen intermediates (i.e., lipid alkoxyl and lipid hydroperoxyl) and, in turn, attack the liver with overloaded NO [175,176]. In addition, upregulated serum aminotransferases of ALT, AST, and ALP may have strong linkage with oxidative hepatotoxicity, due to their high correlation with hepatic injury [177].

3.3. Bioinformatics Enrichment Analysis

Functional analysis of genes or signaling pathways in physiopathology are traditionally conducted among few clusters or even studied separately at a time. On the contrary, the lists of differential gene activities in most cases can be obtained and analyzed by gene-annotation databases with bioanalysis tools and experimental approaches, including DAVID (database for annotation, visualization and integrated discovery), KEGG (Kyoto encyclopedia of genes and genomes) database, KOBAS (KEGG orthology-based annotation system), microarray, and ChIP-on-CHIPs [178]. All these alternative technologies are indeed the bioinformatics scanning methods with sophisticated algorithms, rather than purely statistical tools. Gene-annotation enrichment study allows researchers to predict genome-wide genes under certain conditions and discern a series of specific biological processes most relevant to their investigation. Notably, the database of DAVID is equipped with powerful exploratory capacity for annotating, visualizing, and integrated discovering bioinformatics resources (available at https://david.ncifcrf.gov/home.jsp). Therefore, all the hepatotoxic TCM-modulated targets identified by network pharmacology have been incorporated into the integrated bioinformatics tool termed “Gene ontology (GO)” and “KEGG pathway analysis” in DAVID for identifying enriched biological pathways and highly correlated diseases (shared with similar participating genes) with kappa statistical analysis. Minus log (−log) transformed p value or q value (adjusted p value) have been input into GO and KEGG analysis, respectively (Figure 3A,C) followed by quantitatively estimating the statistical difference in comparison with background genes (background genes are usually automatically collected from backend database in DAVID). A value of −log10 (p or q) larger than 1.3 (equally in p < 0.05) is regarded as a significant difference. The figure of enriched KEGG pathways (Figure 3A) is plotted by R project (available at https://www.r-project.org)
Regarding this study, the methods of collecting biological information with a mode of “gene-to-annotation” is appropriate for mining primary TCM-modulated targets in triggering oxidative hepatotoxicity. After integrating network identified targets into the analytic tools of GO—Biological Process (BP) and KEGG in DAVID, KEGG result indicates that “metabolic pathway (−log10 (q) = 2.492)”, “pathways in cancers (−log10 (q) = 2.122)” and “P13K-Akt signaling pathway (−log10 (q) = 2.057)”, may have the most relevance to hepatotoxic TCM-modulated targets (Figure 3A). However, GO analysis in BP subunit illustrates that the most underlying pathways regulated hepatotoxic TCMs are “extrinsic apoptotic pathway in absence of ligand (−log10 (q) = 3.619)”, “response to toxic substance (−log10 (q) = 3.222)”, “heterocycle metabolic process (−log10 (q) = 2.602)”, and “drug metabolic process (−log10 (q) = 2.009)” (Figure 3C), indicating that intervention with metabolic process may play a vital role in TCM-dependent oxidative liver injury. Nevertheless, it is worth noting that management with targeting mining in a single database is limited with regard to providing comprehensive evidence. In this sense, heterogeneous databases with diverse enrichment tools should be taken into optimized analytic procedures in further study.

3.4. RUCAM (Roussel Uclaf Causality Assessment Method) in TCM-Induced Hepatotoxicity in Clinical Studies

3.4.1. RUCAM-Based Causality Assessment

To cope with the increasing tendency of herb-induced liver injury (HILI), which includes herbal TCM-associated hepatotoxicity, clinicians have made great efforts to establish valid diagnostic criteria in the face of numerous clinical biomarkers [179]. In accordance with the recommendation in Asian and European regions, a causality assessment of HILI can be achieved by using a diagnostic tool RUCAM (Roussel Uclaf Causality Assessment Method), which provides a high degree of certainty [180,181]. Based on the systematical algorithms and quantitative method for identifying hepatotoxins and pharmacological hallmarks in case series, RUCAM is deemed as a validated mean of assigning key points for perceiving liver-specific clinical symptoms and cases. The resulting causality scores are marked in terms of core elements in updated RUCAM, such as time period and alterations of ALT values, etc. Of note, [182]. RUCAM scale typically ranges from −9 to +14, which can be hierarchically categorized as the following: ≥9 (highly probable); 6–8 (probable); 3–5 (possible); 1–2 (unlikely); ≤0 (excludes causality) [182,183].

3.4.2. Identified Hepatotoxic TCM in Case Reports Using RUCAM

Although our knowledge of characteristics related to HILI has been substantially enriched within decades, little amounts of TCM-associated HILI have been identified, not to mention the causality between TCM consumption and hepatotoxicity in RUCAM-dependent clinical trials [184]. Herein, several updated RUCAM-based case reports containing TCM administration have been reviewed in this study. Melchart et al. performed a prospective and large-scale study concerning TCM-hepatotoxicity on the basis of RUCAM [185]. In the research, a total of 21,470 patients without liver diseases were treated with 11 independent herbal TCMs, such as Bupleuri radix, Scutellaeiae radix, and Glycyrrhizae radix, etc. Finally, 26 patients (0.12%) experienced high values of ALT (≥5 × upper limit of normal). However, RUCAM-related causality grades for TCM-treated patients were probable in 8/26 patients (score = 6–8), possible in 16/26 (score = 3–5), and excluded in 2/26 (score ≤ 0). Therefore, 24 patients (0.11%) might undergo TCM-induced hepatotoxicity and the most suspicious TCMs with hepatotoxic effects in this study were Bupleuri radix and Scutellaeiae radix, suggesting that these two herbs are mainly prone to induce liver injury with the causality grading of “possible”. Additional insights should be focused on the treatment of greater celandine and kava, since several RUCAM-based causality assessments indicated that hepatitis and liver cell necrosis could be generated in either greater celandine- or kava-treated patients. Both greater celandine and kava hepatotoxicity are primarily considered as an idiosyncratic liver injury in most susceptible individuals. More specifically, the causality gradings of 12 patients treated with greater celandine were probable or highly probable, while scores of highly probable, probable, or possible gradings could be marked in 8 patients with kava intervention, indicating that hepatic histological activity is more vulnerable to greater celandine or kava treatment [186,187,188,189]. Hao et al. has analyzed the etiology, clinical features, and prognosis of antipyretic analgesic drugs, antibiotics, and TCM-induced hepatotoxicity, including Tripterygium wilfordii, Eucommia ulmoides, and others. According to RUCAM-associated causality assessments in these HILI cases, TCM has emerged as a potential hepatotoxin (score > 3), with the grading at least “possible”, which is identical to both of the antipyretic analgesic drugs and antibiotics (both scores > 3) [190]. Zhang et al. have conducted literature mining targeting hepatotoxic TCM with RUCAM-based high causality grading. As a result, Polygonum multiflorum was documented to be the high probable TCM in 65 of 114 cases, a finding which was consistent with the experimental evidence discussed in this review [191].

3.5. Ethnopharmacology-Associated Challenges and Threats

Until now, accumulating evidence has especially manifested that liver injury may be the result of a therapeutic drug including TCM treatment. On the basis of both TCM theory and aforementioned findings in this study, principal strict challenges and threats can be summarized in the following seven aspects: (a) The criteria for distinguishing and identification of hepatotoxic TCM is ambiguous. Only a minority of hepatotoxic TCMs have been approved with safety issues by the department of China Drug and Food Administration. (b) Systematic screening and research studies focused on discovering and elaborating potential toxic targets for liver injury are greatly limited and time-dependent. Therefore, the findings gained from a certain period may have a deviation from the truth. (c) Most of the investigations concentrate on TCM-dependent pure compounds rather than TCM formula-caused liver dysfunction. Noticeably, TCM formula, which consists of multiple medicinal herbs, is the predominant form applied for treating diseases by TCM practitioners, rather than using a compound or refined extraction from a single herb. Therefore, growing attention should be drawn on disclosing the hepatotoxic effects involved in TCM formula, which seems to be equally meaningful. (d) A majority of studies regarding TCM-induced hepatotoxicity are still under the experimental stage and inadequate in multicenter clinical trials on a large scale. (e) Hepatotoxic role of TCM is composed of diverse pathological mechanisms and not limited to oxidative damage. Such hepatotoxicity-related pathogenesis as inflammation and apoptosis have crosstalk with oxidative stress. Hereby, it is uncertain which pathology plays a predominant and uncontestable role in hepatotoxicity development. (f) Due to the complexity of biological activities, interpretations of the association between numerous genes and relevant pathways should be performed with various advanced algorithms and annotation terms, aiming to avoid incomprehensive data mining. (g) Last but not least, assessment of risk-to-benefit ratio concerning the pharmacological actions of a novel anti-disease drug, including TCM or Western medicine, is imperative, and should be strictly conducted before any clinical applications.

4. Conclusions and Prospects

Regardless of the “holistic” and “natural” therapeutic modalities of TCM in response to various functional abnormalities, escalating interests are related to the cytotoxic role of TCM in the perturbation of organic physiological activities. Current investigations, including this study, have documented that oxidative stress usually participates in progressive hepatotoxicity, particularly triggered by TCM-regulated targets in hepatocytes (Figure 1). However, although liver is equipped with a well-established defense mechanism to protect hepatocytes from oxidative impairment, the intervention of potential toxic TCM remains to successfully invoke oxidative hepatotoxicity. Depending on the target mining by network pharmacology, 10 predominant factors, susceptible to hepatic oxidative injury upon TCM therapy, have been identified, including SOD, MDA, GSH, ROS, GPx, Bax, Caspase-3, Bcl-2, Nrf2, and NO. In addition, TCM-induced hepatotoxicity may be mainly involved in a metabolic pathway in accordance with bioinformatics enrichment analysis. Thus, practitioners should make an effort to be aware of the underlying hepatotoxic hazards prior to TCM application, but not be limited to focusing attention on the limited findings in this review. Selective TCM therapies to multiple diseases without ineluctable hepatotoxicity are essential and should be further studied.

Author Contributions

N.W. and C.Z. wrote and edited the manuscript; Y.F. designed, revised and finalized the manuscript. C.Z., N.W., Y.X., H.-Y.T., S.L. and Y.F. commented on the manuscript during the writing process.

Funding

This research was funded by the Research Council of the University of Hong Kong (project codes: 104003422, 104004092 and 104004460), Wong’s donation (project code: 200006276), a donation from the Gaia Family Trust of New Zealand (project code: 200007008), a contract research project (project code: 260007482), and the Research Grants Committee (RGC) of Hong Kong, HKSAR (Project Codes: 740608, 766211 and 17152116).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ALDalcoholic liver disease
ALPalkaline phosphatase
ALSamyotrophic lateral sclerosis
ALTalanine transaminase
ASTaspartate aminotransferase
ATF6activating transcription factor 6
ATPadenosine triphosphate
CATcatalase
CHOPC/EBP homologous protein
CytCcytochrome C
CYP2C19cytochrome P2C19
CYP2E1cytochrome P2E1
CYP3Acytochrome P3A
CYP3A4cytochrome P3A4
CYP450cytochrome P450
DAVIDdatabase for annotation, visualization and integrated discovery
DBildirect bilirubin
eNOSendothelial NO synthase
ERendoplasmic reticulum
GPxglutathione peroxidase
GRP78glucose regulated protein
GSHglutathione
GSTglutathione S-transferase
HCChepatocellular carcinoma
HCVhepatitis C virus
HILIherb-induced liver injury
IBilindirect bilirubin
IL-6interleukin 6
IL-12interleukin-12
IL-14interleukin-14
IRE1inositol-requiring enzyme 1
iNOSinducible nitric oxide synthase
JNKc-Jun N-terminal kinases
Keap1Kelch-like ECH-associated protein-1
KEGG Kyoto encyclopedia of genes and genomes
KOBAS KEGG orthology-based annotation system
LDHlactate dehydrogenase
LPClysophosphatidylcholine
LPSlipopolysaccharide
MDAmalondialdehyde
NA(D)PHnicotinamide adenine dinucleotide phosphate oxidase
NAFLDnon-alcoholic fatty liver disease
NOnitric oxide
NOSnitric oxide synthase
NQO1NAD(P)H dehydrogenase, quinone 1
Nrf1nuclear respiratory factor 1
Nrf2nuclear factor (erythroid-derived 2)-like 2
MPTmitochondrial permeability transition
MRP2multidrug resistance-associated protein 2
MRP4multidrug resistance protein 4
OATP2organic anion transporting polypeptide 2
p38 MAPKp38 mitogen-activated protein kinase
p-JNKphospho-c-Jun N-terminal kinase
PERKpancreatic ER kinase
PKCprotein kinase C
PLA2phospholipase A2
PPAR-αperoxisome proliferator activated receptor α
PPAR-γperoxisome proliferator-activated receptor-γ
RNSreactive nitrogen species
ROSreactive oxygen species
RUCAMRoussel Uclaf Causality Assessment Method
SODsuperoxide dismutases
TBiltotal bilirubin
TGF-βtransforming growth factor- β
TNFtumor necrosis factor
UGT1A1uridine diphosphate glucuronosyltransferase 1A1
4-HNE4-hydroxynonenal

References

  1. Mendes-Braz, M.; Martins, J.O. Diabetes Mellitus and Liver Surgery: The Effect of Diabetes on Oxidative Stress and Inflammation. Mediat. Inflamm. 2018, 2018, 2456579–2456590. [Google Scholar] [CrossRef] [PubMed]
  2. Taleb, A.; Ahmad, K.A.; Ihsan, A.U.; Qu, J.; Lin, N.; Hezam, K.; Koju, N.; Hui, L.; Qilong, D. Antioxidant effects and mechanism of silymarin in oxidative stress induced cardiovascular diseases. Biomed. Pharmacother. 2018, 102, 689–698. [Google Scholar] [CrossRef] [PubMed]
  3. Vida, C.; Martinez de Toda, I.; Garrido, A.; Carro, E.; Molina, J.A.; De la Fuente, M. Impairment of Several Immune Functions and Redox State in Blood Cells of Alzheimer’s Disease Patients. Relevant Role of Neutrophils in Oxidative Stress. Front. Immunol. 2017, 8, 1974. [Google Scholar] [CrossRef] [PubMed]
  4. Gao, D.; Jing, S.; Zhang, Q.; Wu, G. Pterostilbene protects against acute renal ischemia reperfusion injury and inhibits oxidative stress, inducible nitric oxide synthase expression and inflammation in rats via the Toll-like receptor 4/nuclear factor-kappaB signaling pathway. Exp. Ther. Med. 2018, 15, 1029–1035. [Google Scholar] [CrossRef] [PubMed]
  5. Kocak, C.; Kocak, F.E.; Akcilar, R.; Akcilar, A.; Savran, B.; Zeren, S.; Bayhan, Z.; Bayat, Z. Ukrain (NSC 631570) ameliorates intestinal ischemia-reperfusion-induced acute lung injury by reducing oxidative stress. Bosn. J. Basic Med. Sci. 2016, 16, 75–81. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, N.; Tan, H.Y.; Li, S.; Xu, Y.; Guo, W.; Feng, Y. Supplementation of Micronutrient Selenium in Metabolic Diseases: Its Role as an Antioxidant. Oxid. Med. Cell. Longev. 2017, 2017, 7478523–7478536. [Google Scholar] [CrossRef] [PubMed]
  7. Brennan, L.; Khoury, J.; Kantorow, M. Parkin elimination of mitochondria is important for maintenance of lens epithelial cell ROS levels and survival upon oxidative stress exposure. Biochim. Biophys. Acta 2017, 1863, 21–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Zhang, C.; Xu, Y.; Tan, H.Y.; Li, S.; Wang, N.; Zhang, Y.; Feng, Y. Neuroprotective effect of He-Ying-Qing-Re formula on retinal ganglion cell in diabetic retinopathy. J. Ethnopharmacol. 2018, 214, 179–189. [Google Scholar] [CrossRef] [PubMed]
  9. Wu, Y.; You, J.; Li, F.; Wang, F.; Wang, Y. MicroRNA-542-3p suppresses tumor cell proliferation via targeting Smad2 inhuman osteosarcoma. Oncol. Lett. 2018, 15, 6895–6902. [Google Scholar] [CrossRef] [PubMed]
  10. Zhao, M.G.; Sheng, X.P.; Huang, Y.P.; Wang, Y.T.; Jiang, C.H.; Zhang, J.; Yin, Z.Q. Triterpenic acids-enriched fraction from Cyclocarya paliurus attenuates non-alcoholic fatty liver disease via improving oxidative stress and mitochondrial dysfunction. Biomed. Pharmacother. 2018, 104, 229–239. [Google Scholar] [CrossRef] [PubMed]
  11. Tan, H.Y.; Wang, N.; Li, S.; Hong, M.; Guo, W.; Man, K.; Cheng, C.S.; Chen, Z.; Feng, Y. Repression of WT1-Mediated LEF1 Transcription by Mangiferin Governs β-Catenin-Independent Wnt Signalling Inactivation in Hepatocellular Carcinoma. Cell. Physiol. Biochem. 2018, 47, 1819–1834. [Google Scholar] [CrossRef] [PubMed]
  12. Tu, C.; Gao, D.; Li, X.F.; Li, C.Y.; Li, R.S.; Zhao, Y.L.; Li, N.; Jia, G.L.; Pang, J.Y.; Cui, H.R.; et al. Inflammatory stress potentiates emodin-induced liver injury in rats. Front. Pharmacol. 2015, 6, 233. [Google Scholar] [CrossRef] [PubMed]
  13. Feng, M.; Ding, J.; Wang, M.; Zhang, J.; Zhu, X.; Guan, W. Kupffer-derived matrix metalloproteinase-9 contributes to liver fibrosis resolution. Int. J. Biol. Sci. 2018, 14, 1033–1040. [Google Scholar] [CrossRef] [PubMed]
  14. Grossini, E.; Bellofatto, K.; Farruggio, S.; Sigaudo, L.; Marotta, P.; Raina, G.; De Giuli, V.; Mary, D.; Pollesello, P.; Minisini, R.; et al. Levosimendan inhibits peroxidation in hepatocytes by modulating apoptosis/autophagy interplay. PLoS ONE 2015, 10, e0124742. [Google Scholar] [CrossRef] [PubMed]
  15. Lopez, C.; Checa, S.K.; Soncini, F.C. CpxR/CpxA-controls scsABCD transcription to counteract copper and oxidative stress in Salmonella Typhimurium. J. Bacteriol. 2018. [Google Scholar] [CrossRef] [PubMed]
  16. Mortezaee, K.; Khanlarkhani, N. Melatonin application in targeting oxidative-induced liver injuries: A review. J. Cell. Physiol. 2018, 233, 4015–4032. [Google Scholar] [CrossRef] [PubMed]
  17. Yuan, J.; Wang, D.; Liu, Y.; Chen, X.; Zhang, H.; Shen, F.; Liu, X.; Fu, J. Hydrogen-rich water attenuates oxidative stress in rats with traumatic brain injury via Nrf2 pathway. J. Surg. Res. 2018, 228, 238–246. [Google Scholar] [CrossRef] [PubMed]
  18. Kovalcikova, A.; Gyuraszova, M.; Vavrincova-Yaghi, D.; Vavrinec, P.; Tothova, L.; Boor, P.; Sebekova, K.; Celec, P. Oxidative stress in the brain caused by acute kidney injury. Metab. Brain Dis. 2018, 33, 961–967. [Google Scholar] [CrossRef] [PubMed]
  19. Carvalho, C.G.; Procianoy, R.S.; Neto, E.C.; Silveira, R.C. Preterm Neonates with Respiratory Distress Syndrome: Ventilator-Induced Lung Injury and Oxidative Stress. J. Immunol. Res. 2018, 2018, 6963754–6963758. [Google Scholar] [CrossRef] [PubMed]
  20. Finkel, T. Signal transduction by reactive oxygen species. J. Cell Biol. 2011, 194, 7–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Souayed, N.; Chennoufi, M.; Boughattas, F.; Haouas, Z.; Maaroufi, K.; Miled, A.; Ben-Attia, M.; Aouam, K.; Reinberg, A.; Boughattas, N.A. Circadian variation in murine hepatotoxicity to the antituberculosis agent <<Isoniazide>>. Chronobiol. Int. 2015, 32, 1201–1210. [Google Scholar] [CrossRef] [PubMed]
  22. He, L.; He, T.; Farrar, S.; Ji, L.; Liu, T.; Ma, X. Antioxidants Maintain Cellular Redox Homeostasis by Elimination of Reactive Oxygen Species. Cell. Physiol. Biochem. 2017, 44, 532–553. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Farzaei, M.H.; Zobeiri, M.; Parvizi, F.; El-Senduny, F.F.; Marmouzi, I.; Coy-Barrera, E.; Naseri, R.; Nabavi, S.M.; Rahimi, R.; Abdollahi, M. Curcumin in Liver Diseases: A Systematic Review of the Cellular Mechanisms of Oxidative Stress and Clinical Perspective. Nutrients 2018, 10, 855. [Google Scholar] [CrossRef] [PubMed]
  24. Lefaki, M.; Papaevgeniou, N.; Chondrogianni, N. Redox regulation of proteasome function. Redox. Biol. 2017, 13, 452–458. [Google Scholar] [CrossRef] [PubMed]
  25. Rivero Osimani, V.L.; Valdez, S.R.; Guinazu, N.; Magnarelli, G. Alteration of syncytiotrophoblast mitochondria function and endothelial nitric oxide synthase expression in the placenta of rural residents. Reprod. Toxicol. 2016, 61, 47–57. [Google Scholar] [CrossRef] [PubMed]
  26. Lu, S.C. Antioxidants in the treatment of chronic liver diseases: Why is the efficacy evidence so weak in humans? Hepatology 2008, 48, 1359–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Nowicki, C.J.; Kashian, D.R. Comparison of lipid peroxidation and catalase response in invasive dreissenid mussels exposed to single and multiple stressors. Environ. Toxicol. Chem. 2018, 37, 1643–1654. [Google Scholar] [CrossRef] [PubMed]
  28. Marchlewicz, M.; Szypulska-Koziarska, D.; Grzegrzolka, A.; Kruk, J.; Duchnik, E.; Wiszniewska, B. [Protection against oxidative stress in male reproductive system]. Pomeranian J. Life Sci. 2016, 62, 44–52. [Google Scholar] [PubMed]
  29. Neale, P.A.; Achard, M.E.S.; Escher, B.I.; Leusch, F.D.L. Exploring the oxidative stress response mechanism triggered by environmental water samples. Environ. Sci. Process Impacts 2017, 19, 1126–1133. [Google Scholar] [CrossRef] [PubMed]
  30. Wang, Y.; Han, T.; Xue, L.M.; Han, P.; Zhang, Q.Y.; Huang, B.K.; Zhang, H.; Ming, Q.L.; Peng, W.; Qin, L.P. Hepatotoxicity of kaurene glycosides from Xanthium strumarium L. fruits in mice. Pharmazie 2011, 66, 445–449. [Google Scholar] [PubMed]
  31. Singh, S.V.; Shrivastava, A.; Jyotshna; Chaturvedi, U.; Singh, S.C.; Shanker, K.; Saxena, J.K.; Bhatia, G.; Pal, A. A mechanism-based pharmacological evaluation of efficacy of Flacourtia indica in management of dyslipidemia and oxidative stress in hyperlipidemic rats. J. Basic Clin. Physiol. Pharmacol. 2016, 27, 121–129. [Google Scholar] [CrossRef] [PubMed]
  32. Nauser, T.; Gebicki, J.M. Physiological Concentrations of Ascorbate Cannot Prevent the Potentially Damaging Reactions of Protein Radicals in Humans. Chem. Res. Toxicol. 2017, 30, 1702–1710. [Google Scholar] [CrossRef] [PubMed]
  33. Calabrese, V.; Cornelius, C.; Rizzarelli, E.; Owen, J.B.; Dinkova-Kostova, A.T.; Butterfield, D.A. Nitric oxide in cell survival: A janus molecule. Antioxid. Redox Signal. 2009, 11, 2717–2739. [Google Scholar] [CrossRef] [PubMed]
  34. Dizdaroglu, M.; Jaruga, P. Mechanisms of free radical-induced damage to DNA. Free Radic. Res. 2012, 46, 382–419. [Google Scholar] [CrossRef] [PubMed]
  35. Krajka-Kuzniak, V.; Szaefer, H.; Ignatowicz, E.; Adamska, T.; Markowski, J.; Baer-Dubowska, W. Influence of Cloudy Apple Juice on N-Nitrosodiethylamine-Induced Liver Injury and Phases I and Ii Biotransformation Enzymes in Rat Liver. Acta Pol. Pharm. 2015, 72, 267–276. [Google Scholar] [PubMed]
  36. Lim, E.J.; Chin, R.; Nachbur, U.; Silke, J.; Jia, Z.; Angus, P.W.; Torresi, J. Hepatitis C-induced hepatocyte apoptosis following liver transplantation is enhanced by immunosuppressive agents. J. Viral Hepat. 2016, 23, 730–743. [Google Scholar] [CrossRef] [PubMed]
  37. Belka, M.; Baczek, T. The Metabolism of Anticancer Drugs by the Liver: Current Approaches to the Drug Development Process. Curr. Drug Metab. 2015, 16, 506–521. [Google Scholar] [CrossRef] [PubMed]
  38. Yuen, M.F.; Tam, S.; Fung, J.; Wong, D.K.; Wong, B.C.; Lai, C.L. Traditional Chinese medicine causing hepatotoxicity in patients with chronic hepatitis B infection: A 1-year prospective study. Aliment. Pharmacol. Ther. 2006, 24, 1179–1186. [Google Scholar] [CrossRef] [PubMed]
  39. Motoyama, H.; Enomoto, M.; Yasuda, T.; Fujii, H.; Kobayashi, S.; Iwai, S.; Morikawa, H.; Takeda, T.; Tamori, A.; Sakaguchi, H.; et al. [Drug-induced liver injury caused by an herbal medicine, bofu-tsu-sho-san]. Nihon Shokakibyo Gakkai Zasshi 2008, 105, 1234–1239. [Google Scholar] [PubMed]
  40. Lee, C.H.; Wang, J.D.; Chen, P.C. Risk of liver injury associated with Chinese herbal products containing radix bupleuri in 639,779 patients with hepatitis B virus infection. PLoS ONE 2011, 6, e16064. [Google Scholar] [CrossRef] [PubMed]
  41. Jung, J.Y.; Park, S.M.; Ko, H.L.; Lee, J.R.; Park, C.A.; Byun, S.H.; Ku, S.K.; Cho, I.J.; Kim, S.C. Epimedium koreanum Ameliorates Oxidative Stress-Mediated Liver Injury by Activating Nuclear Factor Erythroid 2-Related Factor 2. Am. J. Chin. Med. 2018, 46, 469–488. [Google Scholar] [CrossRef] [PubMed]
  42. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yang, F.; Liang, Y.; Xu, L.; Ji, L.; Yao, N.; Liu, R.; Shi, L.; Liang, T. Exploration in the cascade working mechanisms of liver injury induced by total saponins extracted from Rhizoma Dioscorea bulbifera. Biomed. Pharmacother. 2016, 83, 1048–1056. [Google Scholar] [CrossRef] [PubMed]
  44. Qi, X.M.; Miao, L.L.; Cai, Y.; Gong, L.K.; Ren, J. ROS generated by CYP450, especially CYP2E1, mediate mitochondrial dysfunction induced by tetrandrine in rat hepatocytes. Acta Pharmacol. Sin. 2013, 34, 1229–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. He, F.Q.; Qiu, B.Y.; Li, T.K.; Xie, Q.; Cui, D.J.; Huang, X.L.; Gan, H.T. Tetrandrine suppresses amyloid-β-induced inflammatory cytokines by inhibiting NF-kappaB pathway in murine BV2 microglial cells. Int. Immunopharmacol. 2011, 11, 1220–1225. [Google Scholar] [CrossRef] [PubMed]
  46. Xue, Y.; Wang, Y.; Feng, D.C.; Xiao, B.G.; Xu, L.Y. Tetrandrine suppresses lipopolysaccharide-induced microglial activation by inhibiting NF-kappaB pathway. Acta Pharmacol. Sin. 2008, 29, 245–251. [Google Scholar] [CrossRef] [PubMed]
  47. Huang, P.; Xu, Y.; Wei, R.; Li, H.; Tang, Y.; Liu, J.; Zhang, S.S.; Zhang, C. Efficacy of tetrandrine on lowering intraocular pressure in animal model with ocular hypertension. J. Glaucoma 2011, 20, 183–188. [Google Scholar] [CrossRef] [PubMed]
  48. Gao, W.Y.; Li, D.; Cai, D.E.; Huang, X.Y.; Zheng, B.Y.; Huang, Y.H.; Chen, Z.X.; Wang, X.Z. Hepatitis B virus X protein sensitizes HL-7702 cells to oxidative stress-induced apoptosis through modulation of the mitochondrial permeability transition pore. Oncol. Rep. 2017, 37, 48–56. [Google Scholar] [CrossRef] [PubMed]
  49. Chen, Y.; Ji, L.; Xiong, A.; Yang, L.; Wang, Z. Involvement of intracellular glutathione in regulating isoline-induced cytotoxicity in human normal liver L-02 cells. Toxicol. Ind. Health 2013, 29, 567–575. [Google Scholar] [CrossRef] [PubMed]
  50. Liu, T.Y.; Chen, Y.; Wang, Z.Y.; Ji, L.L.; Wang, Z.T. Pyrrolizidine alkaloid isoline-induced oxidative injury in various mouse tissues. Exp. Toxicol. Pathol. 2010, 62, 251–257. [Google Scholar] [CrossRef] [PubMed]
  51. Wang, Z.Y.; Kang, H.; Ji, L.L.; Yang, Y.Q.; Liu, T.Y.; Cao, Z.W.; Morahan, G.; Wang, Z.T. Proteomic characterization of the possible molecular targets of pyrrolizidine alkaloid isoline-induced hepatotoxicity. Environ. Toxicol. Pharmacol. 2012, 34, 608–617. [Google Scholar] [CrossRef] [PubMed]
  52. Yang, X.; Li, W.; Sun, Y.; Guo, X.; Huang, W.; Peng, Y.; Zheng, J. Comparative Study of Hepatotoxicity of Pyrrolizidine Alkaloids Retrorsine and Monocrotaline. Chem. Res. Toxicol. 2017, 30, 532–539. [Google Scholar] [CrossRef] [PubMed]
  53. Li, Y.H.; Tai, W.C.; Khan, I.; Lu, C.; Lu, Y.; Wong, W.Y.; Chan, W.Y.; Wendy Hsiao, W.L.; Lin, G. Toxicoproteomic assessment of liver responses to acute pyrrolizidine alkaloid intoxication in rats. J. Environ. Sci. Health C Environ. Carcinog. Ecotoxicol. Rev. 2018, 36, 65–83. [Google Scholar] [CrossRef] [PubMed]
  54. Tu, M.; Li, L.; Lei, H.; Ma, Z.; Chen, Z.; Sun, S.; Xu, S.; Zhou, H.; Zeng, S.; Jiang, H. Involvement of organic cation transporter 1 and CYP3A4 in retrorsine-induced toxicity. Toxicology 2014, 322, 34–42. [Google Scholar] [CrossRef] [PubMed]
  55. Wei, H.; Duan, G.; He, J.; Meng, Q.; Liu, Y.; Chen, W.; Meng, Y. Geniposide attenuates epilepsy symptoms in a mouse model through the PI3K/Akt/GSK-3β signaling pathway. Exp. Ther. Med. 2018, 15, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
  56. Pan, H.; Li, Y.; Qian, H.; Qi, X.; Wu, G.; Zhang, H.; Xu, M.; Rao, Z.; Li, J.L.; Wang, L.; et al. Effects of Geniposide from Gardenia Fruit Pomace on Skeletal-Muscle Fibrosis. J. Agric. Food Chem. 2018, 66, 5802–5811. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, Y.; Shou, K.; Gong, C.; Yang, H.; Yang, Y.; Bao, T. Anti-Inflammatory Effect of Geniposide on Osteoarthritis by Suppressing the Activation of p38 MAPK Signaling Pathway. Biomed. Res. Int. 2018, 2018, 8384576–8384587. [Google Scholar] [CrossRef] [PubMed]
  58. Ding, Y.; Zhang, T.; Tao, J.S.; Zhang, L.Y.; Shi, J.R.; Ji, G. Potential hepatotoxicity of geniposide, the major iridoid glycoside in dried ripe fruits of Gardenia jasminoides (Zhi-zi). Nat. Prod. Res. 2013, 27, 929–933. [Google Scholar] [CrossRef] [PubMed]
  59. Du, Z.A.; Sun, M.N.; Hu, Z.S. Saikosaponin a Ameliorates LPS-Induced Acute Lung Injury in Mice. Inflammation 2018, 41, 193–198. [Google Scholar] [CrossRef] [PubMed]
  60. Lee, T.H.; Park, S.; You, M.H.; Lim, J.H.; Min, S.H.; Kim, B.M. A potential therapeutic effect of saikosaponin C as a novel dual-target anti-Alzheimer agent. J. Neurochem. 2016, 136, 1232–1245. [Google Scholar] [CrossRef] [PubMed]
  61. Zhao, Y.; Wang, Y.J.; Zhao, R.Z.; Xiang, F.J. Vinegar amount in the process affected the components of vinegar-baked Radix Bupleuri and its hepatoprotective effect. BMC Complement. Altern. Med. 2016, 16, 346. [Google Scholar] [CrossRef] [PubMed]
  62. Li, X.; Li, X.; Lu, J.; Huang, Y.; Lv, L.; Luan, Y.; Liu, R.; Sun, R. Saikosaponins induced hepatotoxicity in mice via lipid metabolism dysregulation and oxidative stress: A proteomic study. BMC Complement. Altern. Med. 2017, 17, 219. [Google Scholar] [CrossRef] [PubMed]
  63. Xu, Z.; Zhang, L. BRCA1 expression serves a role in vincristine resistance in colon cancer cells. Oncol. Lett. 2017, 14, 345–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Zeng, F.; Ju, R.J.; Liu, L.; Xie, H.J.; Mu, L.M.; Lu, W.L. Efficacy in Treating Lung Metastasis of Invasive Breast Cancer with Functional Vincristine Plus Dasatinib Liposomes. Pharmacology 2018, 101, 43–53. [Google Scholar] [CrossRef] [PubMed]
  65. Setty, B.A.; Stanek, J.R.; Mascarenhas, L.; Miller, A.; Bagatell, R.; Okcu, F.; Nicholls, L.; Lysecki, D.; Gupta, A.A. VIncristine, irinotecan, and temozolomide in children and adolescents with relapsed rhabdomyosarcoma. Pediatr. Blood Cancer 2018, 65, e26728. [Google Scholar] [CrossRef] [PubMed]
  66. Shati, A.A.; Elsaid, F.G. Hepatotoxic effect of subacute vincristine administration activates necrosis and intrinsic apoptosis in rats: Protective roles of broccoli and Indian mustard. Arch. Physiol. Biochem. 2018. [Google Scholar] [CrossRef] [PubMed]
  67. He, M.; Wu, Y.; Wang, M.; Chen, W.; Jiang, J. Meta-analysis of the clinical value of oxymatrine on sustained virological response in chronic hepatitis B. Ann. Hepatol. 2016, 15, 482–491. [Google Scholar] [PubMed]
  68. Zhou, H.; Shi, H.J.; Yang, J.; Chen, W.G.; Xia, L.; Song, H.B.; Bo, K.P.; Ma, W. Efficacy of oxymatrine for treatment and relapse suppression of severe plaque psoriasis: Results from a single-blinded randomized controlled clinical trial. Br. J. Dermatol. 2017, 176, 1446–1455. [Google Scholar] [CrossRef] [PubMed]
  69. Cao, Y.G.; Jing, S.; Li, L.; Gao, J.Q.; Shen, Z.Y.; Liu, Y.; Xing, Y.; Wu, M.L.; Wang, Y.; Xu, C.Q.; et al. Antiarrhythmic effects and ionic mechanisms of oxymatrine from Sophora flavescens. Phytother. Res. 2010, 24, 1844–1849. [Google Scholar] [PubMed]
  70. Li, H.; Wang, X.; Liu, Y.; Pan, D.; Wang, Y.; Yang, N.; Xiang, L.; Cai, X.; Feng, Y. Hepatoprotection and hepatotoxicity of Heshouwu, a Chinese medicinal herb: Context of the paradoxical effect. Food Chem. Toxicol. 2017, 108, 407–418. [Google Scholar] [CrossRef] [PubMed]
  71. Cameron, M.; Gagnier, J.J.; Little, C.V.; Parsons, T.J.; Blumle, A.; Chrubasik, S. Evidence of effectiveness of herbal medicinal products in the treatment of arthritis. Part 2: Rheumatoid arthritis. Phytother. Res. 2009, 23, 1647–1662. [Google Scholar] [CrossRef] [PubMed]
  72. Li, X.J.; Jiang, Z.Z.; Zhang, L.Y. Triptolide: Progress on research in pharmacodynamics and toxicology. J. Ethnopharmacol. 2014, 155, 67–79. [Google Scholar] [CrossRef] [PubMed]
  73. Shen, G.; Zhuang, X.; Xiao, W.; Kong, L.; Tan, Y.; Li, H. Role of CYP3A in regulating hepatic clearance and hepatotoxicity of triptolide in rat liver microsomes and sandwich-cultured hepatocytes. Food Chem. Toxicol. 2014, 71, 90–96. [Google Scholar] [CrossRef] [PubMed]
  74. Tai, T.; Huang, X.; Su, Y.; Ji, J.; Su, Y.; Jiang, Z.; Zhang, L. Glycyrrhizin accelerates the metabolism of triptolide through induction of CYP3A in rats. J. Ethnopharmacol. 2014, 152, 358–363. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, H.; Ya, G.; Rui, H. Inhibitory Effects of Triptolide on Human Liver Cytochrome P450 Enzymes and P-Glycoprotein. Eur. J. Drug Metab. Pharmacokinet. 2017, 42, 89–98. [Google Scholar] [CrossRef] [PubMed]
  76. Hou, Z.; Chen, L.; Fang, P.; Cai, H.; Tang, H.; Peng, Y.; Deng, Y.; Cao, L.; Li, H.; Zhang, B.; et al. Mechanisms of Triptolide-Induced Hepatotoxicity and Protective Effect of Combined Use of Isoliquiritigenin: Possible Roles of Nrf2 and Hepatic Transporters. Front. Pharmacol. 2018, 9, 226. [Google Scholar] [CrossRef] [PubMed]
  77. Li, X.X.; Du, F.Y.; Liu, H.X.; Ji, J.B.; Xing, J. Investigation of the active components in Tripterygium wilfordii leading to its acute hepatotoxicty and nephrotoxicity. J. Ethnopharmacol. 2015, 162, 238–243. [Google Scholar] [CrossRef] [PubMed]
  78. Mei, Z.; Li, X.; Wu, Q.; Hu, S.; Yang, X. The research on the anti-inflammatory activity and hepatotoxicity of triptolide-loaded solid lipid nanoparticle. Pharmacol. Res. 2005, 51, 345–351. [Google Scholar] [CrossRef] [PubMed]
  79. Xing, Y.X.; Li, M.H.; Tao, L.; Ruan, L.Y.; Hong, W.; Chen, C.; Zhao, W.L.; Xu, H.; Chen, J.F.; Wang, J.S. Anti-Cancer Effects of Emodin on HepG2 Cells as Revealed by (1)H NMR Based Metabolic Profiling. J. Proteome Res. 2018, 17, 1943–1952. [Google Scholar] [CrossRef] [PubMed]
  80. Park, S.Y.; Jin, M.L.; Kang, N.J.; Park, G.; Choi, Y.W. Anti-inflammatory effects of novel polygonum multiflorum compound via inhibiting NF-kappaB/MAPK and upregulating the Nrf2 pathways in LPS-stimulated microglia. Neurosci. Lett. 2017, 651, 43–51. [Google Scholar] [CrossRef] [PubMed]
  81. Lin, E.Y.; Bayarsengee, U.; Wang, C.C.; Chiang, Y.H.; Cheng, C.W. The natural compound 2,3,5,4′-tetrahydroxystilbene-2-O-β-d glucoside protects against adriamycin-induced nephropathy through activating the Nrf2-Keap1 antioxidant pathway. Environ. Toxicol. 2018, 33, 72–82. [Google Scholar] [CrossRef] [PubMed]
  82. Li, C.L.; Ma, J.; Zheng, L.; Li, H.J.; Li, P. Determination of emodin in L-02 cells and cell culture media with liquid chromatography-mass spectrometry: Application to a cellular toxicokinetic study. J. Pharm. Biomed. Anal. 2012, 71, 71–78. [Google Scholar] [CrossRef] [PubMed]
  83. Hwang, Y.H.; Kang, K.Y.; Kim, J.J.; Lee, S.J.; Son, Y.J.; Paik, S.H.; Yee, S.T. Effects of Hot Water Extracts from Polygonum multiflorum on Ovariectomy Induced Osteopenia in Mice. Evid. Based Complement. Alternat. Med. 2016, 2016, 8970585–8970594. [Google Scholar] [CrossRef] [PubMed]
  84. Ling, S.; Xu, J.W. Biological Activities of 2,3,5,4′-Tetrahydroxystilbene-2-O-β-d-Glucoside in Antiaging and Antiaging-Related Disease Treatments. Oxid. Med. Cell. Longev. 2016, 2016, 4973239–4973253. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, T.; Wang, J.; Jiang, Z.; Zhou, Z.; Li, Y.; Zhang, L.; Zhang, L. [Study on hepatotoxicity of aqueous extracts of Polygonum multiforum in rats after 28-day oral administration-analysis on correlation of cholestasis]. Zhongguo Zhong Yao Za Zhi 2012, 37, 1445–1450. [Google Scholar] [PubMed]
  86. Yu, J.; Xie, J.; Mao, X.J.; Wang, M.J.; Li, N.; Wang, J.; Zhaori, G.T.; Zhao, R.H. Hepatoxicity of major constituents and extractions of Radix Polygoni Multiflori and Radix Polygoni Multiflori Praeparata. J. Ethnopharmacol. 2011, 137, 1291–1299. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, M.; Lin, L.; Lin, H.; Qu, C.; Yan, L.; Ni, J. Interpretation the Hepatotoxicity Based on Pharmacokinetics Investigated Through Oral Administrated Different Extraction Parts of Polygonum multiflorum on Rats. Front. Pharmacol. 2018, 9, 505. [Google Scholar] [CrossRef] [PubMed]
  88. Meng, Y.K.; Li, C.Y.; Li, R.Y.; He, L.Z.; Cui, H.R.; Yin, P.; Zhang, C.E.; Li, P.Y.; Sang, X.X.; Wang, Y.; et al. Cis-stilbene glucoside in Polygonum multiflorum induces immunological idiosyncratic hepatotoxicity in LPS-treated rats by suppressing PPAR-gamma. Acta Pharmacol. Sin. 2017, 38, 1340–1352. [Google Scholar] [CrossRef] [PubMed]
  89. Wu, X.; Chen, X.; Huang, Q.; Fang, D.; Li, G.; Zhang, G. Toxicity of raw and processed roots of Polygonum multiflorum. Fitoterapia 2012, 83, 469–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Fan, X.; Zhu, J.Y.; Sun, Y.; Luo, L.; Yan, J.; Yang, X.; Yu, J.; Tang, W.Q.; Ma, W.; Liang, H.P. Evodiamine Inhibits Zymosan-Induced Inflammation In Vitro and In Vivo: Inactivation of NF-kappaB by Inhibiting IkappaBalpha Phosphorylation. Inflammation 2017, 40, 1012–1027. [Google Scholar] [CrossRef] [PubMed]
  91. Schramm, A.; Hamburger, M. Gram-scale purification of dehydroevodiamine from Evodia rutaecarpa fruits, and a procedure for selective removal of quaternary indoloquinazoline alkaloids from Evodia extracts. Fitoterapia 2014, 94, 127–133. [Google Scholar] [CrossRef] [PubMed]
  92. Chien, C.C.; Wu, M.S.; Shen, S.C.; Ko, C.H.; Chen, C.H.; Yang, L.L.; Chen, Y.C. Activation of JNK contributes to evodiamine-induced apoptosis and G2/M arrest in human colorectal carcinoma cells: A structure-activity study of evodiamine. PLoS ONE 2014, 9, e99729. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, Y.; Liu, Q.; Xu, Y.; Liu, C.; Wang, X.; He, X.; Zhu, N.; Liu, J.; Wu, Y.; Li, Y.; et al. Rutaecarpine suppresses atherosclerosis in ApoE-/- mice through upregulating ABCA1 and SR-BI within RCT. J. Lipid Res. 2014, 55, 1634–1647. [Google Scholar] [CrossRef] [PubMed]
  94. Li, X.Y.; Sun, R. [Study on efficacy accompanied by side effects of water extraction components of Evodiae Fructus based on syndrome model]. Zhongguo Zhong Yao Za Zhi 2015, 40, 2753–2759. [Google Scholar] [PubMed]
  95. Cai, Q.; Wei, J.; Zhao, W.; Shi, S.; Zhang, Y.; Wei, R.; Zhang, Y.; Li, W.; Wang, Q. Toxicity of Evodiae fructus on rat liver mitochondria: The role of oxidative stress and mitochondrial permeability transition. Molecules 2014, 19, 21168–21182. [Google Scholar] [CrossRef] [PubMed]
  96. Huang, W.; Li, X.; Sun, R. [“Dose-time-toxicity” relationship study on hepatotoxicity caused by multiple dose water extraction components of Evodiae Fructus to mice]. Zhongguo Zhong Yao Za Zhi 2012, 37, 2223–2227. [Google Scholar] [PubMed]
  97. Uyangaa, E.; Choi, J.Y.; Ryu, H.W.; Oh, S.R.; Eo, S.K. Anti-herpes Activity of Vinegar-processed Daphne genkwa Flos via Enhancement of Natural Killer Cell Activity. Immune Netw. 2015, 15, 91–99. [Google Scholar] [CrossRef] [PubMed]
  98. Li, S.; Chou, G.; Hseu, Y.; Yang, H.; Kwan, H.; Yu, Z. Isolation of anticancer constituents from flos genkwa (Daphne genkwa Sieb.et Zucc.) through bioassay-guided procedures. Chem. Cent. J. 2013, 7, 159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Yun, J.W.; Kim, S.H.; Kim, Y.S.; You, J.R.; Kwon, E.; Jang, J.J.; Park, I.A.; Kim, H.C.; Kim, H.H.; Che, J.H.; et al. Evaluation of subchronic (13 week) toxicity and genotoxicity potential of vinegar-processed Genkwa Flos. Regul. Toxicol. Pharmacol. 2015, 72, 386–393. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, Z.; Chu, Y.; Zhang, Y.; Chen, Y.; Zhang, J.; Chen, X. Investigation of potential toxic components based on the identification of Genkwa Flos chemical constituents and their metabolites by high-performance liquid chromatography coupled with a Q Exactive high-resolution benchtop quadrupole Orbitrap mass spectrometer. J. Sep. Sci. 2018, 41, 3328–3338. [Google Scholar] [PubMed]
  101. Shu, Y.; Liang, Y.; Liang, Z.; Zhao, X.; Zhu, X.; Feng, W.; Liang, J.; Ito, Y. Studies on a Simple and Efficient Method for Large-Scale Preparation of Genkwanin from Daphne Genkwa Sieb. Et Zucc. Using Normal-Phase Flash Chromatography. J. Liq. Chromatogr. Relat. Technol. 2014, 37, 773–785. [Google Scholar] [CrossRef] [PubMed]
  102. Yu, J.G.; Guo, J.; Zhu, K.Y.; Tao, W.; Chen, Y.; Liu, P.; Hua, Y.; Tang, Y.; Duan, J.A. How impaired efficacy happened between Gancao and Yuanhua: Compounds, targets and pathways. Sci. Rep. 2017, 7, 3828. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, Y.; Duan, J.A.; Guo, J.; Shang, E.; Tang, Y.; Qian, Y.; Tao, W.; Liu, P. Yuanhuapine-induced intestinal and hepatotoxicity were correlated with disturbance of amino acids, lipids, carbohydrate metabolism and gut microflora function: A rat urine metabonomic study. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2016, 1026, 183–192. [Google Scholar] [CrossRef] [PubMed]
  104. Tian, Y.; Li, C.P. [Anti-mite activities of 25 kinds of traditional Chinese medicines for Demodex folliculorum]. Zhong Yao Cai 2006, 29, 1013–1015. [Google Scholar] [PubMed]
  105. Shi, J.; Zhou, J.; Ma, H.; Guo, H.; Ni, Z.; Duan, J.; Tao, W.; Qian, D. An in vitro metabolomics approach to identify hepatotoxicity biomarkers in human L02 liver cells treated with pekinenal, a natural compound. Anal. Bioanal. Chem. 2016, 408, 1413–1424. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, N.; Miao, P.P.; Guo, C.E.; Chen, H.Y.; Ma, P.K.; Li, H.P.; Zhu, H.Y.; Gao, X.; Zhang, Y.J. [In vitro effects of Genkwa Flos chloroform extract on activity of human liver microsomes UGTs and UGT1A1]. Zhongguo Zhong Yao Za Zhi 2016, 41, 3296–3302. [Google Scholar] [PubMed]
  107. Sreejith, G.; Latha, P.G.; Shine, V.J.; Anuja, G.I.; Suja, S.R.; Sini, S.; Shyama, S.; Pradeep, S.; Shikha, P.; Rajasekharan, S. Anti-allergic, anti-inflammatory and anti-lipidperoxidant effects of Cassia occidentalis Linn. Indian J. Exp. Biol. 2010, 48, 494–498. [Google Scholar] [PubMed]
  108. Panigrahi, G.; Tiwari, S.; Ansari, K.M.; Chaturvedi, R.K.; Khanna, V.K.; Chaudhari, B.P.; Vashistha, V.M.; Raisuddin, S.; Das, M. Association between children death and consumption of Cassia occidentalis seeds: Clinical and experimental investigations. Food Chem. Toxicol. 2014, 67, 236–248. [Google Scholar] [CrossRef] [PubMed]
  109. Oliveira-Filho, J.P.; Cagnini, D.Q.; Badial, P.R.; Pessoa, M.A.; Del Piero, F.; Borges, A.S. Hepatoencephalopathy syndrome due to Cassia occidentalis (Leguminosae, Caesalpinioideae) seed ingestion in horses. Equine Vet. J. 2013, 45, 240–244. [Google Scholar] [CrossRef] [PubMed]
  110. Chhapola, V.; Kanwal, S.K.; Sharma, A.G.; Kumar, V. Hepatomyoencephalopathy Secondary to Cassia occidentalis Poisoning: Report of Three Cases from North India. Indian J. Crit. Care Med. 2018, 22, 454–456. [Google Scholar] [CrossRef] [PubMed]
  111. Singh, H.P.; Batish, D.R.; Kaur, S.; Arora, K.; Kohli, R.K. alpha-Pinene inhibits growth and induces oxidative stress in roots. Ann. Bot. 2006, 98, 1261–1269. [Google Scholar] [CrossRef] [PubMed]
  112. Panigrahi, G.K.; Yadav, A.; Yadav, A.; Ansari, K.M.; Chaturvedi, R.K.; Vashistha, V.M.; Raisuddin, S.; Das, M. Hepatic transcriptional analysis in rats treated with Cassia occidentalis seed: Involvement of oxidative stress and impairment in xenobiotic metabolism as a putative mechanism of toxicity. Toxicol. Lett. 2014, 229, 273–283. [Google Scholar] [CrossRef] [PubMed]
  113. Ntchapda, F.; Barama, J.; Kemeta Azambou, D.R.; Etet, P.F.; Dimo, T. Diuretic and antioxidant activities of the aqueous extract of leaves of Cassia occidentalis (Linn.) in rats. Asian Pac. J. Trop. Med. 2015, 8, 685–693. [Google Scholar] [CrossRef] [PubMed]
  114. James, K.D.; Kennett, M.J.; Lambert, J.D. Potential role of the mitochondria as a target for the hepatotoxic effects of (-)-epigallocatechin-3-gallate in mice. Food Chem. Toxicol. 2018, 111, 302–309. [Google Scholar] [CrossRef] [PubMed]
  115. Gu, L.L.; Shen, Z.L.; Li, Y.L.; Bao, Y.Q.; Lu, H. Oxymatrine Causes Hepatotoxicity by Promoting the Phosphorylation of JNK and Induction of Endoplasmic Reticulum Stress Mediated by ROS in LO2 Cells. Mol. Cells 2018, 41, 401–412. [Google Scholar] [PubMed]
  116. Wang, S.; Wang, M.; Wang, M.; Tian, Y.; Sun, X.; Sun, G.; Sun, X. Bavachinin Induces Oxidative Damage in HepaRG Cells through p38/JNK MAPK Pathways. Toxins 2018, 10, 154. [Google Scholar] [CrossRef] [PubMed]
  117. Wang, Z.; Zhang, Y.; Liu, Q.; Sun, L.; Lv, M.; Yu, P.; Chen, X. Investigation of the mechanisms of Genkwa Flos hepatotoxicity by a cell metabolomics strategy combined with serum pharmacology in HL-7702 liver cells. Xenobiotica 2018. [Google Scholar] [CrossRef] [PubMed]
  118. Zheng, J.; Yu, L.; Chen, W.; Lu, X.; Fan, X. Circulating exosomal microRNAs reveal the mechanism of Fructus Meliae Toosendan-induced liver injury in mice. Sci. Rep. 2018, 8, 2832. [Google Scholar] [CrossRef] [PubMed]
  119. Shi, S.; Yao, L.; Guo, K.; Wang, X.; Wang, Q.; Li, W. Hepatocellular toxicity of oxalicumone A via oxidative stress injury and mitochondrial dysfunction in healthy human liver cells. Mol. Med. Rep. 2018, 17, 743–752. [Google Scholar] [CrossRef] [PubMed]
  120. Li, Y.; Zhang, Y.; Gao, Y.; Zhang, W.; Cui, X.; Liu, J.; Wei, Y. Arsenic Induces Thioredoxin 1 and Apoptosis in Human Liver HHL-5 Cells. Biol. Trace Elem. Res. 2018, 181, 234–241. [Google Scholar] [CrossRef] [PubMed]
  121. Guo, S.; Zhang, S.; Liu, L.; Yang, P.; Dang, X.; Wei, H.; Hu, N.; Shi, L.; Zhang, Y. Pinelliae Rhizoma Praeparatum Involved in the Regulation of Bile Acids Metabolism in Hepatic Injury. Biol. Pharm. Bull. 2018, 41, 869–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Xue, Y.; Chen, Q.; Sun, J. Hydroxyapatite nanoparticle-induced mitochondrial energy metabolism impairment in liver cells: In vitro and in vivo studies. J. Appl. Toxicol. 2017, 37, 1004–1016. [Google Scholar] [CrossRef] [PubMed]
  123. Xing, X.; Deng, X.; Shi, J.; Zhang, M.; Sun, G.; Tang, S.; Huang, Q.; Sun, X. The chronic hepatotoxicity assessment of the herbal formula Zishen Yutai pill. Regul. Toxicol. Pharmacol. 2017, 83, 81–88. [Google Scholar] [CrossRef] [PubMed]
  124. Xia, X.H.; Yuan, Y.Y.; Liu, M. The assessment of the chronic hepatotoxicity induced by Polygoni Multiflori Radix in rats: A pilot study by using untargeted metabolomics method. J. Ethnopharmacol. 2017, 203, 182–190. [Google Scholar] [CrossRef] [PubMed]
  125. Oyagbemi, A.A.; Omobowale, T.O.; Asenuga, E.R.; Afolabi, J.M.; Adejumobi, O.A.; Adedapo, A.A.; Yakubu, M.A. Effect of arsenic acid withdrawal on hepatotoxicity and disruption of erythrocyte antioxidant defense system. Toxicol. Rep. 2017, 4, 521–529. [Google Scholar] [CrossRef] [PubMed]
  126. Lee, A.Y.; Jang, Y.; Hong, S.H.; Chang, S.H.; Park, S.; Kim, S.; Kang, K.S.; Kim, J.E.; Cho, M.H. Ephedrine-induced mitophagy via oxidative stress in human hepatic stellate cells. J. Toxicol. Sci. 2017, 42, 461–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Prakash, C.; Kumar, V. Chronic Arsenic Exposure-Induced Oxidative Stress is Mediated by Decreased Mitochondrial Biogenesis in Rat Liver. Biol. Trace Elem. Res. 2016, 173, 87–95. [Google Scholar] [CrossRef] [PubMed]
  128. Wu, Q.; Li, W.K.; Zhou, Z.P.; Li, Y.Y.; Xiong, T.W.; Du, Y.Z.; Wei, L.X.; Liu, J. The Tiβn medicine Zuotai differs from HgCl2 and MeHg in producing liver injury in mice. Regul. Toxicol. Pharmacol. 2016, 78, 1–7. [Google Scholar] [CrossRef] [PubMed]
  129. Lu, H.; Zhang, L.; Gu, L.L.; Hou, B.Y.; Du, G.H. Oxymatrine Induces Liver Injury through JNK Signalling Pathway Mediated by TNF-alpha In Vivo. Basic Clin. Pharmacol. Toxicol. 2016, 119, 405–411. [Google Scholar] [CrossRef] [PubMed]
  130. Li, X.Y.; Sun, R. [Study on efficacy and accompanying toxic and side effects of volatile oil of Evodia Fructus based on stomach cold syndrome model]. Zhongguo Zhong Yao Za Zhi 2015, 40, 3838–3844. [Google Scholar] [PubMed]
  131. Zheng, J.; Ji, C.; Lu, X.; Tong, W.; Fan, X.; Gao, Y. Integrated expression profiles of mRNA and microRNA in the liver of Fructus Meliae Toosendan water extract injured mice. Front. Pharmacol. 2015, 6, 236. [Google Scholar] [CrossRef] [PubMed]
  132. Patel, D.N.; Ho, H.K.; Tan, L.L.; Tan, M.M.; Zhang, Q.; Low, M.Y.; Chan, C.L.; Koh, H.L. Hepatotoxic potential of asarones: In vitro evaluation of hepatotoxicity and quantitative determination in herbal products. Front. Pharmacol. 2015, 6, 25. [Google Scholar] [CrossRef] [PubMed]
  133. Wu, Z.T.; Qi, X.M.; Sheng, J.J.; Ma, L.L.; Ni, X.; Ren, J.; Huang, C.G.; Pan, G.Y. Timosaponin A3 induces hepatotoxicity in rats through inducing oxidative stress and down-regulating bile acid transporters. Acta Pharmacol. Sin. 2014, 35, 1188–1198. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, L.; Li, M.D.; Cao, P.P.; Zhang, C.F.; Huang, F.; Xu, X.H.; Liu, B.L.; Zhang, M. Astin B, a cyclic pentapeptide from Aster tataricus, induces apoptosis and autophagy in human hepatic L-02 cells. Chem. Biol. Interact. 2014, 223, 1–9. [Google Scholar] [CrossRef] [PubMed]
  135. Xiao, R.M.; Wang, J.J.; Chen, J.Y.; Sun, L.J.; Chen, Y. Effects of arecoline on hepatic cytochrome P450 activity and oxidative stress. J. Toxicol. Sci. 2014, 39, 609–614. [Google Scholar]
  136. Ma, Y.; Niu, C.; Wang, J.; Ji, L.; Wang, Z. Diosbulbin B-induced liver injury in mice and its mechanism. Hum. Exp. Toxicol. 2014, 33, 729–736. [Google Scholar] [CrossRef] [PubMed]
  137. Wang, K.; Jin, R.M.; Chen, C.X. [Comparative study on hepatic toxicity of gardeniae fructus and Huanglian Jiedu decoction]. Zhongguo Zhong Yao Za Zhi 2013, 38, 2365–2369. [Google Scholar] [PubMed]
  138. Emoto, Y.; Yoshizawa, K.; Kinoshita, Y.; Yuki, M.; Yuri, T.; Yoshikawa, Y.; Sayama, K.; Tsubura, A. Green Tea Extract-induced Acute Hepatotoxicity in Rats. J. Toxicol. Pathol. 2014, 27, 163–174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Amin, K.A.; Hashem, K.S.; Al-Muzafar, H.M.; Taha, E.M. Oxidative hepatotoxicity effects of monocrotaline and its amelioration by lipoic acid, S-adenosyl methionine and vitamin E. J. Complement. Integr. Med. 2014, 11, 35–41. [Google Scholar] [CrossRef] [PubMed]
  140. Hong, M.; Li, S.; Wang, N.; Tan, H.Y.; Cheung, F.; Feng, Y. A Biomedical Investigation of the Hepatoprotective Effect of Radix salviae miltiorrhizae and Network Pharmacology-Based Prediction of the Active Compounds and Molecular Targets. Int. J. Mol. Sci. 2017, 18, 620. [Google Scholar] [CrossRef] [PubMed]
  141. Hong, M.; Zhang, Y.; Li, S.; Tan, H.Y.; Wang, N.; Mu, S.; Hao, X.; Feng, Y. A Network Pharmacology-Based Study on the Hepatoprotective Effect of Fructus Schisandrae. Molecules 2017, 22, 1617. [Google Scholar] [CrossRef] [PubMed]
  142. Stojanovic, M.; Todorovic, D.; Scepanovic, L.; Mitrovic, D.; Borozan, S.; Dragutinovic, V.; Labudovic-Borovic, M.; Krstic, D.; Colovic, M.; Djuric, D. Subchronic methionine load induces oxidative stress and provokes biochemical and histological changes in the rat liver tissue. Mol. Cell. Biochem. 2018. [Google Scholar] [CrossRef] [PubMed]
  143. Boelsterli, U.A.; Hsiao, C.J. The heterozygous Sod2(+/−) mouse: Modeling the mitochondrial role in drug toxicity. Drug Discov. Today 2008, 13, 982–988. [Google Scholar] [CrossRef] [PubMed]
  144. Osman, H.G.; Gabr, O.M.; Lotfy, S.; Gabr, S. Serum levels of bcl-2 and cellular oxidative stress in patients with viral hepatitis. Indian J. Med. Microbiol. 2007, 25, 323–329. [Google Scholar] [PubMed]
  145. Wu, Y.; Zhou, F.; Jiang, H.; Wang, Z.; Hua, C.; Zhang, Y. Chicory (Cichorium intybus L.) polysaccharides attenuate high-fat diet induced non-alcoholic fatty liver disease via AMPK activation. Int. J. Biol. Macromol. 2018, 118, 886–895. [Google Scholar] [CrossRef] [PubMed]
  146. 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]
  147. Sabado, J.; Casanovas, A.; Tarabal, O.; Hereu, M.; Piedrafita, L.; Caldero, J.; Esquerda, J.E. Accumulation of misfolded SOD1 in dorsal root ganglion degenerating proprioceptive sensory neurons of transgenic mice with amyotrophic lateral sclerosis. Biomed. Res. Int. 2014, 2014, 852163–852176. [Google Scholar] [CrossRef] [PubMed]
  148. Tsikas, D. Assessment of lipid peroxidation by measuring malondialdehyde (MDA) and relatives in biological samples: Analytical and biological challenges. Anal. Biochem. 2017, 524, 13–30. [Google Scholar] [CrossRef] [PubMed]
  149. Bhandari, S.; Agarwal, M.P.; Dwivedi, S.; Banerjee, B.D. Monitoring oxidative stress across worsening Child Pugh class of cirrhosis. Indian J. Med. Sci. 2008, 62, 444–451. [Google Scholar] [CrossRef] [PubMed]
  150. Stewart, R.K.; Dangi, A.; Huang, C.; Murase, N.; Kimura, S.; Stolz, D.B.; Wilson, G.C.; Lentsch, A.B.; Gandhi, C.R. A novel mouse model of depletion of stellate cells clarifies their role in ischemia/reperfusion- and endotoxin-induced acute liver injury. J. Hepatol. 2014, 60, 298–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Li, J.; Wu, D.D.; Zhang, J.X.; Wang, J.; Ma, J.J.; Hu, X.; Dong, W.G. Mitochondrial pathway mediated by reactive oxygen species involvement in alpha-hederin-induced apoptosis in hepatocellular carcinoma cells. World J. Gastroenterol. 2018, 24, 1901–1910. [Google Scholar] [CrossRef] [PubMed]
  152. Bertero, E.; Maack, C. Calcium Signaling and Reactive Oxygen Species in Mitochondria. Circ. Res. 2018, 122, 1460–1478. [Google Scholar] [CrossRef] [PubMed]
  153. Cadenas, S. Mitochondrial uncoupling, ROS generation and cardioprotection. Biochim. Biophys. Acta 2018, 1859, 940–950. [Google Scholar] [CrossRef] [PubMed]
  154. Yang, Y.; Zhou, Y.; Cheng, S.; Sun, J.L.; Yao, H.; Ma, L. Effect of uric acid on mitochondrial function and oxidative stress in hepatocytes. Genet. Mol. Res. 2016, 15, 15028644–15028655. [Google Scholar] [CrossRef] [PubMed]
  155. Tan, B.L.; Norhaizan, M.E.; Chan, L.C. ROS-Mediated Mitochondrial Pathway is Required for Manilkara Zapota (L.) P. Royen Leaf Methanol Extract Inducing Apoptosis in the Modulation of Caspase Activation and EGFR/NF-kappaB Activities of HeLa Human Cervical Cancer Cells. Evid. Based Complement. Altern. Med. 2018, 2018, 6578648–6578667. [Google Scholar] [CrossRef] [PubMed]
  156. Wakeyama, H.; Akiyama, T.; Takahashi, K.; Amano, H.; Kadono, Y.; Nakamura, M.; Oshima, Y.; Itabe, H.; Nakayama, K.I.; Nakayama, K.; et al. Negative feedback loop in the Bim-caspase-3 axis regulating apoptosis and activity of osteoclasts. J. Bone Miner. Res. 2007, 22, 1631–1639. [Google Scholar] [CrossRef] [PubMed]
  157. Qiu, Z.; Zhou, J.; Zhang, C.; Cheng, Y.; Hu, J.; Zheng, G. Antiproliferative effect of urolithin A, the ellagic acid-derived colonic metabolite, on hepatocellular carcinoma HepG2.2.15 cells by targeting Lin28a/let-7a axis. Braz. J. Med. Biol. Res. 2018, 51, e7220. [Google Scholar] [CrossRef] [PubMed]
  158. Zou, W.; Ma, X.; Yang, H.; Hua, W.; Chen, B.; Cai, G. Hepatitis B X-interacting protein promotes cisplatin resistance and regulates CD147 via Sp1 in ovarian cancer. Exp. Biol. Med. 2017, 242, 497–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Xiao, J.; Zhang, R.; Huang, F.; Liu, L.; Deng, Y.; Ma, Y.; Wei, Z.; Tang, X.; Zhang, Y.; Zhang, M. Lychee (Litchi chinensis Sonn.) Pulp Phenolic Extract Confers a Protective Activity against Alcoholic Liver Disease in Mice by Alleviating Mitochondrial Dysfunction. J. Agric. Food Chem. 2017, 65, 5000–5009. [Google Scholar] [CrossRef] [PubMed]
  160. Nishina, T.; Deguchi, Y.; Miura, R.; Yamazaki, S.; Shinkai, Y.; Kojima, Y.; Okumura, K.; Kumagai, Y.; Nakano, H. Critical Contribution of Nuclear Factor Erythroid 2-related Factor 2 (NRF2) to Electrophile-induced Interleukin-11 Production. J. Biol. Chem. 2017, 292, 205–216. [Google Scholar] [CrossRef] [PubMed]
  161. Sharma, R.S.; Harrison, D.J.; Kisielewski, D.; Cassidy, D.M.; McNeilly, A.D.; Gallagher, J.R.; Walsh, S.V.; Honda, T.; McCrimmon, R.J.; Dinkova-Kostova, A.T.; et al. Experimental Nonalcoholic Steatohepatitis and Liver Fibrosis Are Ameliorated by Pharmacologic Activation of Nrf2 (NF-E2 p45-Related Factor 2). Cell. Mol. Gastroenterol. Hepatol. 2018, 5, 367–398. [Google Scholar] [CrossRef] [PubMed]
  162. Akino, N.; Wada-Hiraike, O.; Terao, H.; Honjoh, H.; Isono, W.; Fu, H.; Hirano, M.; Miyamoto, Y.; Tanikawa, M.; Harada, M.; et al. Activation of Nrf2 might reduce oxidative stress in human granulosa cells. Mol. Cell. Endocrinol. 2018, 470, 96–104. [Google Scholar] [CrossRef] [PubMed]
  163. Qaisiya, M.; Coda Zabetta, C.D.; Bellarosa, C.; Tiribelli, C. Bilirubin mediated oxidative stress involves antioxidant response activation via Nrf2 pathway. Cell. Signal. 2014, 26, 512–520. [Google Scholar] [CrossRef] [PubMed]
  164. Ramezani, A.; Nahad, M.P.; Faghihloo, E. The role of Nrf2 transcription factor in viral infection. J. Cell. Biochem. 2018, 119, 6366–6382. [Google Scholar] [CrossRef] [PubMed]
  165. Xie, X.; Chen, Q.; Tao, J. Astaxanthin Promotes Nrf2/ARE Signaling to Inhibit HG-Induced Renal Fibrosis in GMCs. Mar. Drugs 2018, 16, 117. [Google Scholar] [Green Version]
  166. Tian, B.; Lu, Z.N.; Guo, X.L. Regulation and role of nuclear factor-E2-related factor 2 (Nrf2) in multidrug resistance of hepatocellular carcinoma. Chem. Biol. Interact. 2018, 280, 70–76. [Google Scholar] [CrossRef] [PubMed]
  167. Guo, Y.; Li, J.X.; Wang, Y.L.; Mao, T.Y.; Chen, C.; Xie, T.H.; Han, Y.F.; Tan, X.; Han, H.X. Yinchen Linggui Zhugan Decoction Ameliorates Nonalcoholic Fatty Liver Disease in Rats by Regulating the Nrf2/ARE Signaling Pathway. Evid. Based Complement. Altern. Med. 2017, 2017, 6178358–6178369. [Google Scholar] [CrossRef] [PubMed]
  168. Lian, Y.; Xia, X.; Zhao, H.; Zhu, Y. The potential of chrysophanol in protecting against high fat-induced cardiac injury through Nrf2-regulated anti-inflammation, anti-oxidant and anti-fibrosis in Nrf2 knockout mice. Biomed. Pharmacother. 2017, 93, 1175–1189. [Google Scholar] [CrossRef] [PubMed]
  169. Liu, S.; Premont, R.T.; Rockey, D.C. Endothelial nitric-oxide synthase (eNOS) is activated through G-protein-coupled receptor kinase-interacting protein 1 (GIT1) tyrosine phosphorylation and Src protein. J. Biol. Chem. 2014, 289, 18163–18174. [Google Scholar] [CrossRef] [PubMed]
  170. Stoessel, A.; Paliege, A.; Theilig, F.; Addabbo, F.; Ratliff, B.; Waschke, J.; Patschan, D.; Goligorsky, M.S.; Bachmann, S. Indolent course of tubulointerstitial disease in a mouse model of subpressor, low-dose nitric oxide synthase inhibition. Am. J. Physiol. Renal Physiol. 2008, 295, F717–F725. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Okuyama, T.; Nakatake, R.; Kaibori, M.; Okumura, T.; Kon, M.; Nishizawa, M. A sense oligonucleotide to inducible nitric oxide synthase mRNA increases the survival rate of rats in septic shock. Nitric Oxide 2018, 72, 32–40. [Google Scholar] [CrossRef] [PubMed]
  172. Gazdic, M.; Simovic Markovic, B.; Vucicevic, L.; Nikolic, T.; Djonov, V.; Arsenijevic, N.; Trajkovic, V.; Lukic, M.L.; Volarevic, V. Mesenchymal stem cells protect from acute liver injury by attenuating hepatotoxicity of liver natural killer T cells in an inducible nitric oxide synthase- and indoleamine 2,3-dioxygenase-dependent manner. J. Tissue Eng. Regen. Med. 2018, 12, e1173–e1185. [Google Scholar] [CrossRef] [PubMed]
  173. Freitas de Lima, F.; Lescano, C.H.; Arrigo, J.D.S.; Cardoso, C.A.L.; Coutinho, J.P.; Moslaves, I.S.B.; Ximenes, T.; Kadri, M.C.T.; Weber, S.S.; Perdomo, R.T.; et al. Anti-inflammatory, antiproliferative and cytoprotective potential of the Attalea phalerata Mart. ex Spreng. pulp oil. PLoS ONE 2018, 13, e0195678. [Google Scholar] [CrossRef] [PubMed]
  174. Kleniewska, P.; Goraca, A. Influence of endothelin 1 receptor blockers and a nitric oxide synthase inhibitor on reactive oxygen species formation in rat lungs. Physiol. Res. 2016, 65, 789–798. [Google Scholar] [PubMed]
  175. Depinay, N.; Franetich, J.F.; Gruner, A.C.; Mauduit, M.; Chavatte, J.M.; Luty, A.J.; van Gemert, G.J.; Sauerwein, R.W.; Siksik, J.M.; Hannoun, L.; et al. Inhibitory effect of TNF-alpha on malaria pre-erythrocytic stage development: Influence of host hepatocyte/parasite combinations. PLoS ONE 2011, 6, e17464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Khan, M.W.; Priyamvada, S.; Khan, S.A.; Khan, S.; Gangopadhyay, A.; Yusufi, A.N. Fish/flaxseed oil protect against nitric oxide-induced hepatotoxicity and cell death in the rat liver. Hum. Exp. Toxicol. 2016, 35, 302–311. [Google Scholar] [CrossRef] [PubMed]
  177. Sharifi-Rigi, A.; Heidarian, E.; Amini, S.A. Protective and anti-inflammatory effects of hydroalcoholic leaf extract of Origanum vulgare on oxidative stress, TNF-alpha gene expression and liver histological changes in paraquat-induced hepatotoxicity in rats. Arch. Physiol. Biochem. 2018, 9, 1–8. [Google Scholar] [CrossRef] [PubMed]
  178. Huang Da, W.; Sherman, B.T.; Lempicki, R.A. Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009, 37, 1–13. [Google Scholar] [CrossRef] [PubMed]
  179. Liu, Z.; He, X.; Wang, L.; Zhang, Y.; Hai, Y.; Gao, R. Chinese Herbal Medicine Hepatotoxicity: The Evaluation and Recognization Based on Large-scale Evidence Ddatabase. Curr. Drug Metab. 2018. [Google Scholar] [CrossRef] [PubMed]
  180. Teschke, R.; Eickhoff, A. Herbal hepatotoxicity in traditional and modern medicine: actual key issues and new encouraging steps. Front. Pharmacol. 2015, 6, 72. [Google Scholar] [CrossRef] [PubMed]
  181. Danan, G.; Teschke, R. RUCAM in Drug and Herb Induced Liver Injury: The Update. Int. J. Mol. Sci. 2015, 17, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Danan, G.; Teschke, R. Drug-Induced Liver Injury: Why is the Roussel Uclaf Causality Assessment Method (RUCAM) Still Used 25 Years After Its Launch? Drug Saf. 2018, 41, 735–743. [Google Scholar] [CrossRef] [PubMed]
  183. Yu, Y.C.; Mao, Y.M.; Chen, C.W.; Chen, J.J.; Chen, J.; Cong, W.M.; Ding, Y.; Duan, Z.P.; Fu, Q.C.; Guo, X.Y.; et al. CSH guidelines for the diagnosis and treatment of drug-induced liver injury. Hepatol. Int. 2017, 11, 221–241. [Google Scholar] [CrossRef] [PubMed]
  184. Frenzel, C.; Teschke, R. Herbal Hepatotoxicity: Clinical Characteristics and Listing Compilation. Int. J. Mol. Sci. 2016, 17, 588. [Google Scholar] [CrossRef] [PubMed]
  185. Melchart, D.; Hager, S.; Albrecht, S.; Dai, J.; Weidenhammer, W.; Teschke, R. Herbal Traditional Chinese Medicine and suspected liver injury: A prospective study. World J. Hepatol. 2017, 9, 1141–1157. [Google Scholar] [CrossRef] [PubMed]
  186. Teschke, R.; Glass, X.; Schulze, J. Herbal hepatotoxicity by Greater Celandine (Chelidonium majus): Causality assessment of 22 spontaneous reports. Regul. Toxicol. Pharmacol. 2011, 61, 282–291. [Google Scholar] [CrossRef] [PubMed]
  187. Teschke, R.; Frenzel, C.; Glass, X.; Schulze, J.; Eickhoff, A. Greater Celandine hepatotoxicity: A clinical review. Ann. Hepatol. 2012, 11, 838–848. [Google Scholar] [PubMed]
  188. Teschke, R.; Glass, X.; Schulze, J.; Eickhoff, A. Suspected Greater Celandine hepatotoxicity: Liver-specific causality evaluation of published case reports from Europe. Eur. J. Gastroenterol. Hepatol. 2012, 24, 270–280. [Google Scholar] [CrossRef] [PubMed]
  189. Teschke, R.; Schwarzenboeck, A.; Hennermann, K.H. Kava hepatotoxicity: Clinical survey and critical analysis of 26 suspected cases. Eur. J. Gastroenterol. Hepatol. 2008, 20, 1182–1193. [Google Scholar] [CrossRef] [PubMed]
  190. Hao, K.; Yu, Y.; He, C.; Wang, M.; Wang, S.; Li, X. RUCAM scale-based diagnosis, clinical features and prognosis of 140 cases of drug-induced liver injury. Zhonghua Gan Zang Bing Za Zhi 2014, 22, 938–941. [Google Scholar] [PubMed]
  191. Zhang, P.; Ye, Y.; Yang, X.; Jiao, Y. Systematic Review on Chinese Herbal Medicine Induced Liver Injury. Evid. Based Complement. Altern. Med. 2016, 2016, 3560812–3560827. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic mechanisms of redox biology and traditional Chinese medicine (TCM)-induced oxidative stress in hepatocytes.
Figure 1. Schematic mechanisms of redox biology and traditional Chinese medicine (TCM)-induced oxidative stress in hepatocytes.
Ijms 19 02745 g001
Figure 2. Network pharmacology-based target identification of TCM-derived natural compounds or extracts for the generation of oxidative hepatotoxicity.
Figure 2. Network pharmacology-based target identification of TCM-derived natural compounds or extracts for the generation of oxidative hepatotoxicity.
Ijms 19 02745 g002
Figure 3. Target mining for TCM-caused oxidative hepatotoxicity by network pharmacology and bioinformatics enrichment analysis. (A) Scatter plot of enriched KEGG pathways statistics. The gene ratio illustrates the significantly expressed gene number to the total gene number in a certain pathway. (B) Identification of TCM-modulated targets for oxidative liver injury by network pharmacology. (C) GO analysis on the involvement of principal biological process. (*): −log10 (p) > 1.3; (**): −log10 (p) > 2, and (***): −log10 (p) > 3 versus background genes.
Figure 3. Target mining for TCM-caused oxidative hepatotoxicity by network pharmacology and bioinformatics enrichment analysis. (A) Scatter plot of enriched KEGG pathways statistics. The gene ratio illustrates the significantly expressed gene number to the total gene number in a certain pathway. (B) Identification of TCM-modulated targets for oxidative liver injury by network pharmacology. (C) GO analysis on the involvement of principal biological process. (*): −log10 (p) > 1.3; (**): −log10 (p) > 2, and (***): −log10 (p) > 3 versus background genes.
Ijms 19 02745 g003
Table 1. Summary on the properties of TCM-induced hepatotoxicity in the recent 5 years.
Table 1. Summary on the properties of TCM-induced hepatotoxicity in the recent 5 years.
Natural CompoundSources of Chinese MedicineStudy TypeCell or AnimalBiochemical Markers of HepatotoxicityType of InjuryReporting DateRef.
VincristineCatharanthus roseusIn vivoWistar rat ALT, AST, IL-12. IL-4, p53, cleaved caspase-3, Bax↑; Bcl-2↓Hepatitis2018[66]
Epigallocatechin-3-gallateGreen teaIn vivoC57BL/6 mouseSOD, GPx, respiratory complex-I -III, sirtuin 3, FOXO3, Nrf2↓Hepatitis and hemorrhage2018[114]
OxymatrineSophora flavescensIn vitroL-02 cellsPro-caspase-3 -4 -8 -9, GRP78, CHOP, p-JNK, IREI, ATF6, PERK, Bax, MDA, ROS↑; SOD, Bcl-2↓Cell apoptosis2018[115]
BavachininFructus psoraleaeIn vitroHepaRG cellsJNK, p-p38, ROS, MAPK, MDA↑; SOD, GSH, CAT↓Cell necrosis2018[116]
Genkwa Flos extractGenkwa flosIn vitro & in vivoHL-7702 cells; SD ratALT, AST, MDA↑; CAT, GSH, SOD, NO, NOS↓Metabolism Dysregulation 2018[117]
Fructus Meliae Toosendan extractFructus meliae toosendanIn vivoBALB/c mouseALT, AST, MDA, p53, p21, Cyclin E, Bax, CytC, caspase-3 -9, CDK2, ROS↑; Bcl-2, Nrf2, miR-370-3p↓Cell apoptosis2018[118]
Oxalicumone APenicillium oxalicumIn vitroL-02 cellsALT, AST, ROS, Caspase 3, MDA, NO, Fas, Bax, LDH, CytC↑; Bcl-2, GSH, SOD↓Cell apoptosis2018[119]
Arsenic extractArsenicIn vitroHHL-5 cellThioredoxin 1 (Trx1), TrxR1, ROS↑; Bax, CytC, Bcl-2↓Cell apoptosis2018[120]
Pinelliae Rhizoma Praeparatum Pinelliae rhizomaIn vivoICR mouseALT, AST, ALP, bile acid, Mrp3, MDA↑; SOD, GSH, GPx, Bsep, Mrp2, Nrf2↓Metabolism dysregulation 2018[121]
Hydroxyapatitenanoparticles extractHydroxyapatite nanoparticlesIn vitro & In vivoBRL cells;
SD rat
TNF-α, NO, MDA, ROS↑; respiratory complex-I, -II, -III, GSH, SOD↓Metabolism dysregulation2018[122]
Zishen Yutai pill extractZishen yutai pillIn vivoSD ratAST, ALP, ALT, MDA, LDH, PDGF, Cholestasis, Bile acid↑; SOD, GPx↓Cell necrosis2017[123]
Polygoni Multiflori Radix extractPolygonum multiflorum thunbIn vivoSD ratALT, AST, ALP, LDH, bilirubin, creatinine↑SOD↓Metabolism dysregulation2017[70] [124]
Arsenic acidArsenicIn vivoWistar ratMDA, NO↑; SOD, GSH, GST, GPx↓Metabolism dysregulation2017[125]
SaikosaponinsRadix bupleuriIn vitro & In vivoHepG2 cells; Kunming mouseCYP2E1, AST, ALT, LDH, ROS, iNOS↑; GSH↓Metabolism dysregulation2017[62]
EphedrineEphedra sinicaIn vitroLX-2 cellsParkin, SOD2, ROS, Cox IV, p62, LC3 I, LC3 II↑Excessive Mitophagy2017[126]
Arsenic extractArsenicIn vivoWister ratBax, caspase-3↑, CytC, SOD, complexes I, COX-I-IV, NRF-1-2, PGC-1α, Tfam↓Metabolism dysregulation2016[127]
Dioscorea Bulbifera saponins Dioscorea bulbiferaIn vitro & In vivoL-02 cells;
Wister rat
ALT, AST, cytochromes P450, cholestasis↑; SOD, GPx, GST, GR, GCL↓Metabolism dysregulation2016[43]
Zuotai extractZuotaiIn vivoKunming mouseALT, AST, HgS, MeHg, metallothionein-1, heme oxygenase-1 (HO-1), Egr1, Gst-mu, mKC, MIP-2, NAD(P)H, Nqo1, Gclc↑Cell inflammation2016[128]
Oxymatrine Sophora flavescensIn vivoICR mouseALT, AST, ALP, TNF-α, caspase-9, -8, -3, TRADD, p-SAPK, p-JNK↑Cell apoptosis2016[129]
Evodia Fructus volatile oilEvodia fructusIn vivoKunming mouseALT, AST, PGE2, MDA, NO, NOS↑; SOD, GSH, GPx↓Metabolism dysregulation2015[130]
Fructus Meliae Toosendan extract Fructus meliae
toosendan
In vivoBALB/c mouseALT, AST, ALP, bilirubin, cholesterol↑; Nrf2↓Cell necrosis2015[131]
TriptolideTripterygium wilfordiiIn vivoKunmingmouseALT, AST, blood urea nitrogen (BUN), CREA↑; GSH↓Acute hepatic necrosis2015[77]
AsaronesAsarumIn vitroTHLE-2 cellsCaspase-3 -7, MDA↑; GSH, GSSG↓Cell apoptosis2015[132]
Timosaponin A3Anemarrhena asphodeloidesIn vivoSD ratBile acid, ROS, HO-1↑; Ntcp, Bsep, Mrp2, Cyp7a1, F-actin↓ Metabolism dysregulation2014[133]
Astin BAster tataricusIn vitroL-02 cellsROS, JNK, CytC, Bax, caspases-9, -3, LC3-II↑; GSH, Bcl-2, p62↓Cell apoptosis and inflammation 2014[134]
Cassia Occidentalis extractCassia occidentalisIn vivoWister ratTGF-β, JNK, Bax, MDA↑; Akt, CREB1, CYP1A1, CYP2B1, CAT, SOD1, IL-6, SOD, GR↓Metabolism dysregulation and apoptosis2014[112]
Arecoline Hydrobromide Areca catechuIn vivoWister ratALT, AST, MDA, CYP2B, CYP2E1↑; SOD, CAT, GPx, GSH↓Liver cirrhosis and HCC2014[135]
Diosbulbin BDioscorea bulbiferaIn vivoICR mouseALT, AST, ALP, MDA↑; GPx, GST, SOD, CAT↓Metabolism dysregulation2014[136]
Evodiae Fructus extractEvodiae fructusIn vivoSD ratMDA, CytC, AST, ALT, NO, NOS↑; SOD, GSH, GPx↓Cell necrosis2014[94] [95]
Gardeniae Fructus extractGardeniae fructusIn vivoSD ratALT, AST, ALP, bile acid, MDA, TNF-α, Bax↑; SOD, GPx, Bcl-2↓Cell inflammation, necrosis and apoptosis2014[137]
Green tea extractGreen teaIn vivoSD ratALT, AST, ALP, TBil, bilirubin, caspase-3, MDA, TG, GST-P↑Metabolism dysregulation and apoptosis2014[138]
Monocrotaline RattlebushIn vivoSD ratGSH, GR, GPx, GST↓Metabolism dysregulation2014[139]
In the Table 1, the symbols of “↑” and “↓” respectively represent for upregulated (“↑”) or downregulated (“↓”) biochemical markers by traditional Chinese medicine (TCM) treatment.

Share and Cite

MDPI and ACS Style

Zhang, C.; Wang, N.; Xu, Y.; Tan, H.-Y.; Li, S.; Feng, Y. Molecular Mechanisms Involved in Oxidative Stress-Associated Liver Injury Induced by Chinese Herbal Medicine: An Experimental Evidence-Based Literature Review and Network Pharmacology Study. Int. J. Mol. Sci. 2018, 19, 2745. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19092745

AMA Style

Zhang C, Wang N, Xu Y, Tan H-Y, Li S, Feng Y. Molecular Mechanisms Involved in Oxidative Stress-Associated Liver Injury Induced by Chinese Herbal Medicine: An Experimental Evidence-Based Literature Review and Network Pharmacology Study. International Journal of Molecular Sciences. 2018; 19(9):2745. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19092745

Chicago/Turabian Style

Zhang, Cheng, Ning Wang, Yu Xu, Hor-Yue Tan, Sha Li, and Yibin Feng. 2018. "Molecular Mechanisms Involved in Oxidative Stress-Associated Liver Injury Induced by Chinese Herbal Medicine: An Experimental Evidence-Based Literature Review and Network Pharmacology Study" International Journal of Molecular Sciences 19, no. 9: 2745. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19092745

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop