Pharmacophore an International Research Journal
Pharmacophore
Submit Manuscript
Open Access | Published: 2020 - Issue 5

A Review of The Protective Effects of Natural Compounds Against Chemical-Induced Hepatotoxicity

Nadia Salem Alrawaiq1, Azman Abdullah2*, Huda Alrawiq3, Nasser Alrawiq3

 

  1. Department of Pharmacology, Faculty of Pharmacy, Sebha University, Sebha, Libya.
  2. Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia.
  3. Department of Botany, Faculty of Sciences, Sebha University, Sebha, Libya.

ABSTRACT

Globally, liver injuries are a serious public health problem for people. Recent investigations have demonstrated that plants and their important active ingredients exert protective effects against toxic agents that may cause liver injuries, such as pollutants, drugs, and alcohol, which consequently develop into the liver failure, cirrhosis, fibrosis, hepatitis, fatty liver, and even carcinoma. Several natural products and their bioactive compounds are protective against liver injuries. The underlying mechanisms mainly include lipid metabolism regulation, DNA damage repair, anti-necrosis, anti-apoptosis, anti-inflammation, and anti-oxidation. In this review, the protective effects and especially mechanism of action of natural products and bioactive components on liver damages caused by chemicals has been summarized. This current information will aid in the prevention and treatment of liver-related diseases, especially those induced by chemicals.

Keywords:  Liver injury, liver disease, hepatotoxicity, hepatoprotective, natural compounds, natural products.


Introduction

Oxidative stress is a biochemical condition that occurs in the body, producing several types of reactive species, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS) [1]. ROS are very reactive against macromolecules such as DNA, proteins, and membrane lipids in living cells. Oxidative stress-induced changes play a key role in the etiology and pathogenesis of many diseases, such as hepatotoxicity, nephrotoxicity, diabetes mellitus, and cancers [2]. A variety of risk factors, such as drugs, alcohol, irradiation, and environmental pollutants, can induce oxidative stress in the liver and result in the development of severe liver diseases [3].

Liver is the largest internal organ in the human body that plays a significant role in the metabolism and detoxification of various chemicals, drugs, and other toxic compounds [4]. Liver diseases caused by oxidative stress facilitate the pathogenesis of hepatic fibrosis, liver cirrhosis, and even hepatocellular carcinoma [5,6]. Oxidative stress is regarded as one of the pathological mechanisms that cause the initiation and progression of liver damage through inducing irreversible alterations in lipid membranes, proteins, and DNA and, more importantly, through modulating pathways that control biological function [7,8].

The human body has natural defense mechanisms that protect against free radicals and oxidative damage. These mechanisms are both enzymatic, such as reactions catalyzed by catalase (CAT), superoxide dismutase (SOD), and the glutathione system (glutathione (GSH), glutathione reductase, peroxidase, and transferase), and non-enzymatic, such as the pathways driven by low molecular weight antioxidants such as vitamins A, E, and C [9,10]. However, the need for antioxidant supplements to combat oxidative damage becomes essential if the balance between antioxidant defense system and free radicals is compromised [11].

Antioxidants are free radical scavengers that can neutralize free radicals before attacking cells, preventing damage to proteins, enzymes, lipids, carbohydrates, and DNA [12]. For treating liver toxicity, A wide range of antioxidants from natural and synthetic sources have been proposed [13]. The good candidates to protect against toxicities-induced by chemicals are natural compounds that inhibit enzymes that activate free radicals. In this review, we summarized in vivo and in vitro investigations related to the hepatoprotective effects of plants and their active ingredients against toxic chemical agents.

 

Liver Diseases

Liver disorders are among the major global health problems and are the 5th most prevalent cause of death worldwide [14,15]. Changes in liver anatomy or liver function are characterized as liver disease. Liver diseases may be classified as cirrhosis (degenerative disorder leading to liver fibrosis), chronic or acute hepatitis (inflammatory liver diseases), and hepatosis (noninflammatory diseases). Additionally, the liver is the first organ involved in counteracting the effects of toxic compounds and metabolites capable of inducing damage, namely, cellular necrosis, GSH depletion, and increased lipid peroxidation associated with increased liver markers, including bilirubin, alkaline phosphatase (ALP), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) [16].

After chronic liver injury, liver disorders lead to various pathological changes like fatty liver, increased ROS or oxidative stress, liver cell necrosis, hepatitis, steatosis, cholestasis, veno-occlusive diseases vascular lesions, granuloma, and, increased inflammatory markers, cirrhosis, fibrosis, and hepatocellular carcinoma, which further promote portal hypertension and organ failure [17]. Exposure to xenobiotics and several environmental pollutants, including acetaminophen (or N-acetyl-para-aminophenol, APAP), antibiotics, chemotherapeutic agents, carbon tetrachloride (CCl4), thioacetamide (TAA), and chronic alcoholism, is responsible for the progression of liver disorders, as liver damage is caused by elevated levels of ROS in liver tissues [18,19]. Unfortunately, since the conventional or synthetic drugs used to treat these diseases are insufficient and sometimes cause serious side effects, the preferred treatments for liver diseases are controversial [20].

Recent investigations have focused on drugs derived from medicinal plants due to high levels of flavonoid and polyphenolic compounds and presumably minimal treatment side effects as well as relatively low cost; the protection provided by medicinal plant-based drugs against various drug- and chemical-induced hepatotoxicity has been extensively studied in vivo and in vitro, and this research is urgently needed [21,22]. To date, hundreds of plants have been examined in a wide spectrum of liver diseases [23,24].

 

Protection against CCl4-induced hepatotoxicity

CCl4 is a hepatotoxic agent that is extensively used in animal models to induce chronic and acute liver injury [25] and to evalauate the effectiveness of many hepatoprotective drugs [6]. CCl4 is known to cause liver damage and hepatocyte apoptosis/necrosis in vitro and in vivo [26-29]. The damage responses induced by injection of CCl4 in a rat model are similar to human liver cirrhosis [30]. CCl4 is converted by hepatic cytochrome P450 2E1 (cytochrome P450, family 2, subfamily E, polypeptide 1; CYP2E1) into highly reactive radicals, such as trichloromethyl (CCl3) free radical and trichloromethyl peroxy radical (CCl3OO) [31]. Then, these radicals attack cellular macromolecules and cause lipid peroxidation, protein degradation, and DNA damage. The process is followed by the release of hepatic inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), which subsequently leads to damage, including hepatocellular necrosis [26, 27, 31].

In numerous studies, the results have revealed that, by inhibiting lipid peroxidation and enhancing antioxidant enzyme activity, plant extracts with antioxidant activities protect against CCl4-induced hepatotoxicity [15, 32-38]. In vitro and in vivo experimental animal model studies suggested that a variety of natural product extracts or their bioactive components exert potent hepatoprotective activities against CCl4-induced hepatotoxicity (Table 1).

 

Protection against Acetaminophen-Induced Hepatotoxicity

Acetaminophen (APAP), which is also known as paracetamol, is a well-known antipyretic and analgesic drug worldwide [39]. APAP overdose leads to severe centrilobular hepatic necrosis in experimental animals and humans [40]. The metabolic activation of APAP produces N-acetyl-p-benzoquinone imine (NAPQI) and ROS. NAPQI can bind to sulfhydryl groups, spontaneously reacting with GSH and binding to hepatic proteins, leading to the critical event that initiates cell death in APAP-induced liver injury and GSH depletion [41]. However, due to the prolonged activation of cJun-N-terminal kinase (JNK), mitochondrial dysfunction, hepatocyte necrosis, and hepatotoxicity occur with APAP overdose or GSH shortage [42,43]. A recent study demonstrated that PARP1 is involved in this process by activating PXR and promoting toxic P450 enzymes [44].

Several investigations have demonstrated that medicinal plants can enhance liver function against paracetamol-induced toxicity. In in-vivo and in vitro studies, results have revealed that the plasma concentrations of NAPQI in rats significantly enhanced the absorption of paracetamol and was reduced by chrysin, a plant flavonoid, which may be due to inhibition of CYP2E1 and other cytochrome p450 (CYP) enzymes (CYP3A4 and CYP1A2). Additionally, chrysin significantly reduced hepatic injury markers such as serum glutamic oxaloacetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT), ALP, total protein (TP) and total bilirubin (TBL); ameliorated histopathological alterations; reduced oxidative stress damage; and inhibited microscopic changes in liver tissue with paracetamol-induced oxidative stress [45]. Table 2 shows the phytochemicals derived from different plant sources and their hepatoprotective activity against APAP-induced hepatotoxicity.

 

Protection against D-Galactosamine and Lipopolysaccharide-Induced Hepatotoxicity

D-galactosamine, as an amino sugar, is a selective hepatotoxin that induces liver damage resembling viral hepatitis [46]. D-galactosamine causes necrotic and apoptotic cell death in the liver by inducing oxidative stress. The increase in plasma ALT in rats and induction of both oncotic necrosis and hepatocellular caspase-dependent apoptosis can be caused by a single high dose of D-galactosamine [47]. D-galactosamine is metabolized exclusively in the liver through the galactose pathway, leading to the deoxynucleotidyl transferase-mediated depletion of the uracil nucleotide 2′‐deoxyuridine 5′‐triphosphate (dUTP), thus inhibiting hepatic transcription [48].

Numerous reports indicate that, by using free radical scavengers and/or naturally occurring antioxidants, such as spirulina platensis [49], pentoxifylline and caffeic acid phenethyl ester [50], catechin [51], biochanin A [52], p-coumaric acid and kaempferol [53], and quercetin [54], D-galactosamine-induced liver damage is attenuated.

An in vivo study showed that the treatment of ICR mice with flavonoid and phenolic acid fractions isolated from Lolium multiflorum Lam. reduced the D-galactosamine-induced increases in serum ALP, AST, ALT, and lactate dehydrogenase (LDH) levels [55]. The combined treatment of phenolic acid and flavonoid fractions isolated from L. multiflorum Lam. Inhibited the D-galactosamine-mediated increases in histological damage, serum enzyme levels, and hepatic malondialdehyde (MDA) levels, while restoring the activities of hepatic antioxidant enzymes to control levels and augmenting the D-galactosamine-induced expression of nuclear factor erythroid-2-related factor 2 (Nrf2) and Heme oxygenase 1 (HO-1) in the liver [55].

A recent study demonstrated that a nanoformulation of ganoderic acid (bioactive component in Ganoderma lucidum) significantly attenuated markers of hepatic damage in rats with D-galactosamine-induced hepatotoxicity [56]. Also, a methanolic extract of Flacourtia sepiaria exhibited significant hepatoprotective activity and afforded protection from galactosamine-induced liver damage, which could be at least partly attributed to the free radical scavenging activity of the tannins and antioxidants in the extract [57].

Lipopolysaccharide (LPS), a key cell wall component in gram-negative bacteria, can induce severe inflammation leading to hepatic injury [58]. D-galactosamine suppresses the synthesis of different RNAs and increases susceptibility to LPS-induced hepatotoxicity [59]. TNF-α is the main mediator during GalN/LPS-evoked apoptotic liver injury [60]. Also, excess ROS and hepatic GSH depletion are involved in the pathogenesis of GalN/LPS-induced apoptotic liver injury [61].

Amygdalin (D-mandelonitrile-β-D-glucoside-6-β-D-glucoside), also known as laetrile or vitamin B17, is distributed universally in the seeds of almonds, peaches, apricots, and other rosaceous stone fruits and exhibited a protective effect against LPS/GalN-induced acute liver injury that may be associated with the suppression of inflammatory cytokine production, the inhibition of the NLRP3 inflammasome and nuclear factor kappa B (NF-κB) activation, and the upregulation of the Nrf2/quinone oxidoreductase (NQO1) signaling pathway [62].

Also, rosmarinic acid, extracted from Rosmarinus officinalis Linn, can protect C57BL/6 mice from LPS/D-GalN-induced acute liver injury, as indicated not only by the decrease in serum AST and ALT but also in the amelioration of increases in NF-κB and extracellular signal-regulated kinase (ERK1/2) phosphorylation, p38 protein expression and tissue myeloperoxidase (MPO) content. Moreover, rosmarinic acid can increase the glutathione-dependent peroxidase (GPX) level. Furthermore, rosmarinic acid promoted the Nrf2 transport into the nucleus and then upregulated HO-1, glutamate-cysteine ligase catalytic subunit (GCLC), glutamate-cysteine ligase modifier (GCLM), and NQO1 [63].

Recently, forsythiaside (major active component in Forsythia suspense) supplementation alleviated the inflammatory response induced by Escherichia coli–derived LPS in broiler chickens by inhibiting the production of TP, NO, LITAF, IL-1β, IL-17, and IL-6 and by downregulating the mRNA expression of pro-inflammatory cytokines and inducible nitric oxide synthase (iNOS) [64].

Geraniol (GOH) is acyclic monoterpene alcohol isolated from the essential oils of orange, ginger, rose, or lemon [65, 66]. The results showed that LPS/D-GalN-induced fulminant hepatic failure was protected effectively by GOH, as evidenced by the attenuation of pathological liver injury, MPO activity, MDA level, and serum AST and ALT levels. GOH reduced liver TNF-α and IL-1β levels by inhibiting the activation of the NF-κB signaling pathway [67]. Furthermore, GOH increased PPARγ expression in fulminant hepatic failure induced by LPS/D-GalN. This study demonstrated that GOH protects against LPS/D-GalN-induced fulminant hepatic failure by inhibiting the inflammatory response and increasing PPARγ expression [67].

Tectorigenin, a component of Belamcanda adams, has antioxidant, anti‐inflammatory, and antiproliferation activities [68]. In vivo and in vitro studies concluded that pretreatment with tectorigenin ameliorated liver injury, attenuated hepatic inflammation, and decreased mortality in mice with fulminant hepatic failure by inhibiting inflammation through the TLR4/mitogen-activated protein kinase (MAPK) and TLR4/NF‐κB pathways and promoting autophagy [69].

 

Protection against Thioacetamide-Induced Hepatotoxicity

TAA is an organosulfur, white crystalline compound with liver damage and carcinogenic activity caused by cytomegaly [70]. After the oxidative bioactivation of TAA by CYP enzymes and flavin adenine dinucleotide-containing monooxygenase, TAA metabolites react to cellular proteins and amine lipids, preventing hepatocyte dysfunction and cytotoxicity [71] in addition to dramatically increasing ROS production [72], increasing serum transaminase levels, and remarkably decreasing hepatic GSH levels [73].

Dendropanax morbifera significantly prevents hepatic fibrosis by TGF-β1/Smads signaling pathways and inhibiting oxidative stress in rats treated with TAA [74]. The administration of an ethanol extract of Andrographis paniculata at different doses of (200 and 250 mg/kg bw) to albino rats with TAA-induced hepatotoxicity significantly decreased the levels of serum bilirubin, ALP, SGPT, and SGOT but increased the level of protein in the liver [75].

 

Protection against Alcohol-Induced Hepatotoxicity

Alcoholic liver disease is a major global health problem. The alcoholic liver disease typically progresses through the stages of alcoholic steatosis, hepatitis, and cirrhosis [76, 77]. Frequent and excessive alcohol consumption leads to about 2.5million deaths annually [78]. A previous research report linked chronic alcohol consumption to various pathological conditions from simple intoxication to severe life-threatening pathological states [79]. The pathological condition of alcohol-induced liver disease is characterized by a range of morphological changes from minimal injury to advanced liver damage [80]. Chronic ethanol ingestion causes fibrosis, inflammation, hepatomegaly, fatty liver, and cirrhosis, stimulates hepatic oxygen consumption, and induces free radical production.

Fatty liver is present in more than 90% of people with chronic alcoholism, while approximately 10–20% of people who drink heavily progress to alcoholic hepatitis and cirrhosis, indicating that factors such as viral infection (HBV), nutrition, genetic background, and chronic exposure to/intake of xenobiotics, heavy metals, aflatoxins, and paracetamol interact to influence the progression of liver disease [81].

Chronic alcohol consumption has been reported to induce oxidative stress via multiple mechanisms [82, 83]. 3 mechanisms suggested to cause alcoholic liver injury include: (1) acetaldehyde toxicity [84]; (2) exposure to oxidative stress or metabolic generation of ROS [85-88]; and (3) promotion of an immune response that causes oxidative stress in hepatocytes [89-91].

Ethanol is primarily metabolized by alcohol dehydrogenase and aldehyde dehydrogenase enzyme systems that generate acetaldehyde and acetate as products [87]. Acetaldehyde forms adducts with the vital biomolecules DNA and proteins and these adducts are responsible for the impaired structure and function of the liver. Another metabolic pathway that plays a significant role in alcohol toxicity is the CYP2E1 pathway, which comprises a microsomal ethanol oxidation system [83] that is involved in the generation of ROS [92-94].

ROS formation leads to collagen generation in hematopoietic stem cells (HSCs) as acetaldehyde directly upregulates collagen I transcription and indirectly upregulates the synthesis of TGF‐β1 through initiating phagocytosis by macrophages [95]; thus, this process sensitizes hepatocytes to TGF-β-induced apoptosis [96]. In a defensive mechanism, Kupffer cells, along with infiltrating neutrophils and macrophages, potentiate the release of several pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6 [97].

Herbal drugs play a role in the treatment and management of various liver disorders [98]. The oral administration of 250 and 500mg/kg of an aqueous extract of Boswellia serrata offered significant (P < 0.01) dose-dependent protection against hepatotoxicity induced by ethanol [99]. In animal models, by decreasing CYP2E1 activity quercetin, increasing GSH levels, and inducing antioxidant enzymes, the quercetin ameliorated lipid metabolism and ethanol-induced liver damage [87,100]. The activity and expression of CYP2E1 in human hepatocytes is also inhibited by Quercetin [87, 101]. Concerning hepatoprotective effects, in vitro investigations have reported that quercetin ameliorated and AST and LDH release from hepatocytes, GSH depletion, lipid peroxidation, and ethanol-induced liver cell injury, while upregulating hemeoxygenase-1 via the MAPK/Nrf2 pathways in human hepatocytes [102, 103]. Notably, the induction of HO-1 expression utilizing natural compounds helps protect liver in different experimental models. Quercetin was found to induce HO-1 in the livers of normal ICR mice [104, 105].

Following ethanol intoxication, treatment with quercetin also reversed the ethanol-induced increases in serum aminotransferase levels and liver tissue lipid peroxides and hydroperoxides levels and restored the levels of GSH, SOD, GPX, and GR in rats [106]. The antioxidant effect of quercetin is due to the presence of a rich source of OH groups and conjugating π bond orbital that act as potent H2O2 scavengers and proton or electron donors [107].

Silymarin, a traditional herbal medicine extracted from milk thistle fruit (Silybum marianum L. Gaertn), is used to protect the liver [108]. Concerning hepatoprotective effects, silymarin has been shown to possess protective effects in both acute and chronic models of ethanol toxicity [109-111]. Tea extract is rich in catechins that can reduce the CYP2E1 expression and hepatic lesions induced by paracetamol injection [112]; moreover, a diet that contained epigallocatechin gallate (EGCG) improved hepatic injury but did not reduce hepatic CYP2E1 levels [113]. Catechins have previously been reported to stimulate the 63 kDa laminin receptor [114-117], which may initiate ROS production [118]. The development of an ethanol­induced acute gastric lesion was prevented by the ingestion of a strawberry extract rich in anthocyanins before ethanol treatment via the induction of gastric antioxidant enzymes [119].

Pretreatment with Changkil saponins extracted from the root of Platycodon grandiflorum before ethanol administration significantly decreases hepatic triglyceride level, hepatic lipid peroxidation, hepatic TNF-α level, serum and ALT activity [120]. The liver histopathological studies have indicated that changkil saponins prevents ethanol-induced necrosis and steatosis. Additionally, Changkil saponins are protected against depletion of hepatic GSH levels induced by ethanol. CYP2E1 can contribute to ethanol-induced oxidative stress and liver injury. The concurrent administration of Changkil saponins more efficaciously abrogated CYP2E1 induction and the CYP2E1-dependent hydroxylation of aniline than the administration of individual treatments at high doses.

Curcumin has been reported to be the principal active component of Cucuma longa; several recent studies showed the strong antioxidant action of curcumin [121]. Molecular investigations have proven that the administration of curcumin to rats with alcohol-induced liver disease suppresses the expression of iNOS, cyclooxygenase (COX)-2, chemokines, and cytokines in Kupffer cells of the liver and prevents the activation of NF-κB [122]. Previous studies have shown the protective impact of curcumin against alcohol-induced hepatoxicity; curcumin mitigats oxidative stress and prevents liver cell damage in experimental animals [123-125]. In vitro studies with liver slice culture, the results have shown that lipid peroxidation was decreased by curcumin. Curcuin reduced the release of LDH, and attenuated the antioxidant enzymes GPX, CAT, and  SOD [123].

A recent study demonstrated that alcohol-induced hepatic damage in rats was effectively prevented by the extracts of Radix fici Hirtae. Moreover, the possible mechanisms of this hepatoprotective effect may be attributed to free radical scavenging and decreased lipid peroxidation [126]. Also, triterpenoids, major components of G. lucidum, protected from alcohol-induced liver injury by inhibiting lipid peroxidation, increasing antioxidant enzyme activities, and suppressing apoptosis and the immune-inflammatory response [127].

 

Protection against Rifampicin- and Isoniazid-Induced Hepatotoxicity

Tuberculosis (TB) has recently become a major health risk along with the increasing HIV epidemic [128]. Isoniazid (INH) and rifampicin (RIF) are the first-line antituberculosis drugs recommended by the World Health Organization. These two drugs are metabolized in the liver, and both INH and RIF cause hepatotoxicity [129]. The mechanism behind RIF- and INH-induced toxicity is a drug-drug interaction in which the induction of hepatic microsomal enzymes by RIF results in the increased production of a hepatotoxic metabolic product of INH [130]. Irreversible damage, including steatosis, inflammation, cell necrosis, and liver failure can be caused by INH/RIF [131-133], which are mainly associated with oxidative stress [134]; Via oxidative stress, INH induces apoptosis and prevents Nrf2 translocation into the nucleus, thereby inhibiting the cytoprotective effect of Nrf2 [135].

By suppressing hepatocyte apoptosis and oxidative stress, plant extracts alleviate liver injury [136]. The naphthoquinones lawsone (2-hydroxy-1,4-naphthoquinone) is the major bioactive component found in the leaves of the henna plant, Lawsonia inermis L. (Lythraceae). Lawsone at very low concentrations, significantly restores the cells' viability (7.5 μM), reduces transaminase secretion, and decreases MDA levels in RIF-INH-treated HepG2 cells [137]. Additionally, the administration of lawsone to the RIF-INH-treated animals significantly decreases the levels of serum transaminases, improves albumin/globulin ratio, and decreases the level of bilirubin [137].

A fruit pulp extract of Telfairia occidentalis significantly (P < 0.001) reduced the elevated biochemical enzyme markers (ALP, ALT, and AST), lipid profile, and MDA in RIF- and INH-induced oxidative stress and improved the antioxidant potential in rats [138]. A recent study demonstrated that a hot aqueous leaf extract of Lasianthera africana attenuated RIF- and INH-induced hepatotoxicity and the major mechanisms of action responsible for this hepatoprotective effect of L. africana depend on antioxidant effects [139]. Tamarix gallica leaf extract possesses promising hepatoprotective activity against RIF- and INH-induced liver injury in experimental rats [140].

 

Protection against Non-Steroidal Anti-Inflammatory Drugs (Nsaids)-Induced Hepatotoxicity

10% of the total cases of drug-induced hepatotoxicity are caused by Non-steroidal anti-inflammatory drugs (NSAIDs) [141]. NSAIDs induce liver damage due to ROS production and increase lipid peroxidation by decreasing the GSH levels that induce hepatotoxicity due to the generation of free radicals [142, 143]. Several enzymes found in the cytosol are released into the blood in response to the disturbance of hepatocyte transport function, resulting in increased enzyme levels in the blood serum, which indicate hepatocellular damage [144].

Diclofenac is an extensively circulated drug used for the treatment of pain, musculoskeletal diseases, and inflammation in animals and humans due to anti-inflammatory, analgesic, and antipyretic properties; however, severe pathologic conditions like renal papillary necrosis, hepatotoxicity, gastrointestinal bleeding, peptic ulceration, and renal failure could be as a result of long-term administration of this drug [145-148]. Diclofenac was found to cause a rare but fatal hepatotoxic disorder that may be related to the formation of reactive metabolites (5-hydroxy diclofenac and N,5-dihydroxy diclofenac) [149-151] that may act as ROS/RNS, further increasing the accumulation of endogenous ROS and promoting oxidative stress in animal tissues, which consequently leads lipids, protein, and nucleic acid oxidation [152].

A recent study concluded that ethanol extraction of moringa leaves exhibited in vivo antioxidant activity, suggesting a beneficial effect against the oxidative liver damage induced by diclofenac sodium. Also, the bioactive phenolics in ethanol extracts of moringa leaves protected the plasma membrane and increased the reparative and regenerative capacity of the liver [153]. Compared with the control, pretreatment of diclofenac-intoxicated rats with an aqueous ethanol extract of Dialium guineense leaf significantly (p <0.01) reduced the levels of ALT, AST, gamma-glutamyl transferase (GGT), MDA, creatinine, and urea and significantly (p < 0.01) increased the levels of SOD, CAT, GPX, glutathione S-transferase (GST), GSH, and glucose 6-phosphatase (G6Pase) [154].

 

Protection against Chemotherapeutic Agent-Induced Hepatotoxicity

Carboplatin is a second-generation platinum group-based chemotherapeutic drug [155] that is commonly used for ovarian, head-neck, and small cell lung cancer and is preferred over other platinum (cisplatin, etc.) group-based drugs due to few adverse effects and high tolerance [156, 157]. It interacts with nucleophilic molecules including RNA and DNA as it does with all other members of the platinum group when entering the cell and causes various lesions on the purine base after binding to DNA [158].

Cisplatin, as a chemotherapeutic drug, is used to treatm different types of malignancies, including head, neck, ovarian, and testicular cancers [158]. Despite the well-documented efficacy of cisplatin in the treatment of a variety of tumors, the clinical use of cisplatin is often limited because of cellular resistance and severe side effects in normal tissues [159-161]. Also, the non-specific action of cisplatin is detrimental to the morphology and functions of vital tissues, which primarily occur in kidneys [162,163], testes [164-166], and liver [167,168]. The exact biochemical and molecular mechanism of cisplatin-induced tissue injury has not been well defined, although many hypotheses propose that cisplatin toxicity is related to the generation of RNS and ROS, which results in cell injury and mitochondrial dysfunction [169, 170]. The key mechanisms involved in cisplatin toxicity include inflammation, oxidative stress, and apoptosis in tissue [171,172].

Pro-apoptotic proteins are activated to induce BCL2-associated X (Bax) translocation to the mitochondrial outer membrane, which causes the release of cytochrome c into the cytosol when cisplatin induces ROS generation [173]. Caspase-9 is initiated and activates downstream caspase-3, which causes apoptosis upon receiving an apoptosis signal from Bax [174]. Also, numerous studies have shown that activated NF-κB may activate many pro-inflammatory cytokines, such as IL-1β and TNF-α, and induce inflammatory mediators, such as COX-2 and iNOS [175,176].

Medicinal plants have been recognized as a major source of dietary phytochemicals with strong antioxidant activity, which, when coadministered with chemotherapeutic agents, provide better efficacy than chemotherapeutic drugs alone and attenuate vital tissue toxicity [177-179]. Three limonoids, deacetoxy-7R-hydroxygedunin, 7-deacetoxy-7-oxogedunin, and 17-epi-methyl-6-hydroxylangolensate, were isolated from Khaya grandifoliola and found to protect the normal human liver cell line L-02 against cisplatin-induced hepatotoxicity mainly through the induction of mitogen-activated protein kinase phosphatase-1, an endogenous inhibitor of JNK phosphorylation, and through the nuclear translocation of Nrf2 [180]. Nigella sativa, known as black cumin, has been used as a traditional medicine to prevent a wide range of diseases in different parts of the world, particularly in Arab countries. A recent study has shown that N. sativa seed oil has a hepatoprotective effect that decreases the oxidative stress markers and histological damage in the liver tissue of carboplatin-treated rats [181]. Several plant extracts have hepatoprotective effects against cisplatin-induced hepatotoxicity (Table 3).

 

Protection against Lead-Induced Hepatotoxicity

Heavy metals (lead, arsenic, mercury, etc.) are severe menaces to human health due to their toxic effects on various vital organs, including the liver. Lead is a potential environmental pollutant that is toxic to most body organs, including organs in the nervous, digestive, and cardiovascular systems as well as the bone, kidney, liver, and blood [182-184]. Long-term exposure to lead even at a low concentrations has been reported to be a potential factor that induces nephrotoxicity, hepatotoxicity, behavioral dysfunction, reproductive dysfunctions, and so on [185-187]. In the organs of animals and humans affected with lead poisoning, the liver is considered the second most common organ for lead storage, followed by the kidney [188]. Also, some research has shown that lead causes liver lipid peroxidation [189,190], GSH reduction [191], and antioxidant defense system destruction [192], which is the main reason for lead-induced liver injury [193, 194].

C. longa (turmeric), native to tropical South Asia, belongs to the Zingiberaceae family [195]. Investigations have reported that the liver was protected against lead acetate by C. longa at a dose of 500mg/kg body weight [196] and curcumin or nano curcumin at doses of 15mg/kg through decreasing lipid peroxidation, liver enzymes, and oxidative stress, while increasing antioxidant content, such as SOD [197]. Salidroside is a secondary metabolite of Rhodiola rosea L. can decrease the lead content in the body, inhibit oxidative stress, increase antioxidant activity, and inhibit liver CYP2E1/NOX2 expression, thus improving the liver tissue structure [198].

 

Conclusion

Due to universal alcohol overconsumption, medicine administration, and unavoidable environmental pollutants that exist worldwide, chemical-induced liver injuries are very common. The cascades of pathological processes can be initiated by liver injuries, which goes beyond the liver and worsen overall health. Many natural products have been investigated, and their potent hepatoprotective effects have been demonstrated through mechanisms that reduce oxidative stress, decrease inflammation, inhibit apoptosis, ameliorate necrosis, repair DNA damage, and modulate lipid metabolism. These capabilities to restore the structure and function of hepatocytes enable natural products to be considered promising alternatives in the selection of a healthy diet and potential candidates for the development of functional pharmaceuticals and foods. As most of the current evidence is derived from animal-based studies, human trials are needed in the future, and the bioavailability of natural products should be taken into consideration. Moreover, additional natural products should be evaluated for hepatoprotective effects, and the active components of natural products need to be identified and isolated to describe additional active chemicals. Various mechanisms must also be further explored and clarified. This review provides current information regarding the use of natural products in the treatment and prevention of chemical-induced liver injuries. Using this review, people can be advised by physicians and nutritionists to consume some foods and also, medicinal herbs to protect themselves against  alcohol, drugs, and environmental pollutants which cause liver injuries.

 

Acknowledgements

The authors would like to thank Sebha University and Universiti Kebangsaan Malaysia for their support in conducting hepatoxicity research.

 

Table 1: Natural Products that Display Hepatoprotective Effects against Carbon Tetrachloride Toxicity

Experimental

Model

Natural Products

Reference

Rats

Matricaria chamomilla L. extract

[199]

Rats

Olive oil and N. sativa oil

[200]

Rats

Fresh juice of the bark from tender stems of Azadirachta indica

[201]

Rats

Extracts of Zapoteca portoricensis leaves

[202]

Mice

Apple polyphenols (AP, Appjfnol)

[203]

Mice

Rosmarinic acid

[204]

Rats

Aerial and root extracts of Gentiana asclepiadea L.

[205]

Mice

Oleuropein (leaves and fruits of olive, Olea europaea L.)

[206]

Rats

Methanol extract of Solanum xanthocarpum leaves

[207]

Rats

Methanol extracts of different parts of Tamarindus indica Linn.

[208]

Rats

Citrullus lanatus extract

[209]

Mice

Ethanol extracts of Lophatherum gracile leaves

[210]

Rats

Extract of Vetiveria zizanioides roots

[211]

Rats

Ethyl acetate fraction of L. inermis fruit extract

[212]

Rats

Silene villosa (Caryophyllaceae) methanol extract (aerial parts)

[213]

Rabbits

Parthenium hysterophorus crude extract

[214]

Mice

Ethyl acetate fraction of Dracocephalum rupestre Hance

[215]

Mice

Ethanol extract of Syringa oblata Lindl. leaves

[216]

Mice

Spondias mombin leaf and stem extracts

[217]

Rats

Hydroalcoholic extract of Urena lobata leaves

[218]

Rats

Whole plant methanol extracts of Sauropus bacciformis

[219]

Mice

Capparis spinosa seeds (extracted with methanol)

[220]

Rats

Olea europaea L. dry leaf extract

[221]

Rats

Methanol extract of Anacardium occidentale bark

[222]

Rats

Aqueous extract of Allium cepa

[223]

Rabbits

Aqueous extract of Argyrolobium roseum (whole plant)

[224]

Rats

Methanol extract of Kyllinga brevifolia (whole plant)

[225]

Rats

Origanum majorana root extract

[226]

Rats

Ethanol extract of Nerium indicum Mill. leaves

[227]

Rats

Black mulberry (Morus nigra) extract

[228]

Rats

Aqueous extract of Atriplex halimus leaves

[229]

Rats

Methanol extract of Syzygium samarangense leaves

[230]

Rats

Hydroethanolic extract of the stem bark of Oroxylum indicum

[231]

Rats

Linum grandiflorum Desf Seed oil.

[232]

Rats

Hydroalcoholic extract of Cichorium intybus leaves

[233]

Rats

Ethanol extract of Mentha arvensis Leaves

[234]

Rats

Aqueous-methanol extract of Cichorium intybus seeds

[235]

Rats

Mentha piperita L. essential oil

[236]

Rats

Rumex tingitanus leaf extracts

[237]

Rats

S. marianum or milk thistle seed extract

[238]

 

Table 2: Plants That Have Hepatoprotective Effects against Apap-Induced Hepatotoxicity

Experimental

Model

Natural Products

Reference

Rats

Curcuma xanthorrhiza Roxb extracts

[239]

Rats

Psidium guajava (guava) fruit polysaccharide

[240]

Rats

Opuntia robusta and Opuntia streptacantha fruit extracts

[241]

Mice

Methanol extract of Costus Speciosus (Koen. ex. Retz.)

[242]

Rats

Aqueous extract of Casuarina equisetifolia

[243]

Rats

Chloroform extract of Indigofera barberi (whole plant)

[244]

Rats

Leaf extract of Aegle marmelos (L.) Corrêa, Rutaceae

[245]

Rats

Aqueous extract of Celosia argentea L.

[246]

Rats

Ethanol extract of Pavetta indica Linn leaves

[247]

Mice

Ethanol and ethyl acetate extracts of Copaifera multijuga bark

[248]

Mice

Polysaccharides isolated from Dendrobium officinale

[249]

Rats

Ethanol extract of Centella asiatica Linn.

[250]

Rats

Saponarin, isolated from Gypsophila trichotoma Wend.

[251]

Rats

N. sativa Linn. oil

[252]

Rabbit

Aqueous ethanol extract of Alhagi maurorum Boiss.

[253]

Rats

Ethanol extract of Moringa oleifera bark

[254]

Rats

Ethanol extract of Trichosanthes lobata

[255]

Rats

Leaf extract of Eclipta alba (L.) Hassk.

[256]

Rats

Aqueous and ethanolic extracts of Helicanthus elastica

[257]

Rats

Zerumbone isolated from the rhizomes of Zingiber zerumbet

[258]

Rats

Ethanol and aqueous extracts of Aloe vera Linn. stems

[259]

Rats

Ethanolic root extract of Taraxacum syriacum Boiss

[260]

Rats

Leaf extract of Pterospermum acerifolium

[261]

Mice

Diallyl sulfide (DAS)

[262]

Rats

Myristica fragrans (nutmeg) kernel extract

[263]

 

Table 3: Plants that Have Hepatoprotective Effects against Cisplatin-Induced Hepatotoxicity

Experimental

Model

Natural Products

Reference

Mice

Mice

Methanol extract of Capparis spinosa seeds

N. sativa

[220]

[264]

Mice

Cuminum cyminum

[264]

Rats

Hydro-methanol extract of Cornus mas fruit

[265]

Rats

Aegle marmelos fruit

[266]

References

  1. Ermak G, Davies KJ. Calcium and oxidative stress: from cell signaling to cell death. Mol Immunol 2002;38(10):713-72
  2. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med 2010;49(11):1603-1616.
  3. Jagtap P, Szabó C. Poly (ADP-ribose) polymerase and the therapeutic effects of its inhibitors. Nat Rev Drug Discov 2005;4(5):421.
  4. Forouzandeh H, Azemi ME, Rashidi I, Goudarzi M, Kalantari H. Study of the protective effect of Teucrium polium L. extract on acetaminophen-induced hepatotoxicity in mice. Iran J Pharm Res 2013;12(1):123.
  5. Yang J, Zhu D, Ju B, Jiang X, Hu J. Hepatoprotective effects of Gentianella turkestanerum extracts on acute liver injury induced by carbon tetrachloride in mice. Am J Transl Res 2017;9(2):569.
  6. Recknagel RO, Glende Jr EA, Dolak JA, Waller RL. Mechanisms of carbon tetrachloride toxicity. Pharmac Ther 1989;43(1):139-154.
  7. Feng Y, Wang N, Ye X, Li H, Feng Y, Cheung F, Nagamatsu T. Hepatoprotective effect and its possible mechanism of Coptidis rhizoma aqueous extract on carbon tetrachloride-induced chronic liver hepatotoxicity in rats. J Ethnopharmacol 2011;138(3):683-690.
  8. Cichoż-Lach H, Michalak A. Oxidative stress as a crucial factor in liver diseases. World J Gastroenterol 2014;20(25):8082.
  9. Evans MD, Dizdaroglu M, Cooke MS. Oxidative DNA damage and disease: induction, repair, and significance. Mutat Res 2004;567(1):1-61.
  10. Manda G, Nechifor MT, Neagu TM. Reactive oxygen species, cancer, and anti-cancer therapies. Curr Chem Biol 2009;3(1):22-46.
  11. Shukla S, Mehta A, Bajpai VK, Shukla S. In vitro antioxidant activity and total phenolic content of ethanolic leaf extract of Stevia rebaudiana Bert. Food Chem Toxicol 2009;47(9):2338-2343.
  12. Fang YZ, Yang S, Wu G. Free radicals, antioxidants, and nutrition. Nutr 2002;18(10):872-879.
  13. Cuzzocrea S, Riley DP, Caputi AP, Salvemini D. Antioxidant therapy: a new pharmacological approach in shock, inflammation, and ischemia/reperfusion injury. Pharmacol Rev 2001;53(1):135-159.
  14. Arbab AH, Parves MK, Al-Dosari MS, Al-Rehaily AJ, Ibrahim KE, Alam P, Alsaid MS, Rafatullah S. Therapeutic efficacy of ethanolic extract of Aerva javanica aerial parts in the amelioration of CCl4-induced hepatotoxicity and oxidative damage in rats. Food Nutr Res 2016;60(1):30864.
  15. Eidi A, Mortazavi P, Moghadam JZ, Mardani PM. Hepatoprotective effects of Portulaca oleracea extract against CCl4-induced damage in rats. Pharm Biol 2015;53(7):1042-1051.
  16. Ganaie MA, Khan TH, Siddiqui NA, Ansari MN. Ameliorative effect of methanol extract of Rumex vesicarius on CCl4-induced liver damage in Wistar albino rats. Pharm Biol 2015;53(8);1163-1167.
  17. Singh A, Bhat T, Sharma O. Clinical Biochemistry of Hepatotoxicity. J Clin Toxicol 2011;4(0001):1-19.
  18. Sreelatha S, Padma P, Umadevi M. Protective effects of Coriandrum sativum extracts on carbon tetrachloride-induced hepatotoxicity in rats. Food Chem Toxicol 2009;47(4):702-708.
  19. Roy A, Bhoumik D, Sahu RK, Dwivedi J. Medicinal plants used in liver protection: A review. UK J Pharm Biosci 2014;2(1);23-33.
  20. Kumar CH, Ramesh A, Suresh Kumar JN, Ishaq BM. A review on hepatoprotective activity of medicinal plants. Int J Pharm Sci Res 2011;2(3):501.
  21. Pereira C, Barros L, Ferreira IC. Extraction, identification, fractionation, and isolation of phenolic compounds in plants with hepatoprotective effects. J Sci Food Agr 2016;96(4):1068-1084.
  22. Levy C, Seeff LD, Lindor KD. Use of herbal supplements for chronic liver disease. Clin Gastroenterol Hepatol 2004;2(11):947-956.
  23. Asadi-Samani M, Rafieian-Kopaei M, Azimi N. Gundelia: a systematic review of medicinal and molecular perspective. Pak J Biol Sci 2013;16(21):1238-1247.
  24. Asadi-Samani M, Bahmani M, Rafieian-Kopaei M. The chemical composition, botanical characteristic and biological activities of Borago officinalis: a review. Asian Pac J Trop Med 2014;7(Suppl 1):S22-S28.
  25. Lee HY, Kim SW, Lee GH, Choi MK, Jung HW, Kim YJ, Kwon HJ, Chae HJ. Turmeric extract and its active compound, curcumin, protect against chronic CCl 4-induced liver damage by enhancing antioxidation. BMC Complem Altern M 2016;16(1):316.
  26. Zhang W, Dong Z, Chang X, Zhang C, Rong G, Gao X, Zeng Z, Wang C, Chen Y, Rong Y, Qu J, Lui Z, Lu Y. Protective effect of the total flavonoids from Apocynum venetum L. on carbon tetrachloride-induced hepatotoxicity in vitro and in vivo. J Physiol Biochem 2018;74(2):301-312.
  27. Sadek K, Saleh E, Nasr S. Molecular hepatoprotective effects of lipoic acid against carbon tetrachloride-induced liver fibrosis in rats: Hepatoprotection at molecular level. Hum Exp Toxicol 2018;37(2):142-154.
  28. Yang C, Li L, Ma Z, Zhong Y, Pang W, Xiong M, Fang S, Li Y. Hepatoprotective effect of methyl ferulic acid against carbon tetrachloride-induced acute liver injury in rats. Exp Ther Med 2018;15(3):2228-2238.
  29. Peng X, Dai C, Lui Q, Li J, Qui J. Curcumin attenuates on carbon tetrachloride-induced acute liver injury in mice via modulation of the Nrf2/HO-1 and TGF-β1/Smad3 pathway. Molecules 2018;23(1):215.
  30. Weiler-Normann C, Herkel J, Lohse A. Mouse models of liver fibrosis. Z Gastroenterol 2007;45(1):43-50.
  31. Weber LW, Boll M, Stampfl A. Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol 2003;33(2):105-136.
  32. Dhiman A, Nanda A, Ahmad S. A recent update in research on the antihepatotoxic potential of medicinal plants. Chin J Integr Med 2012;10(2):117-127.
  33. Duh PD, Lin SL, Wu SC. Hepatoprotection of Graptopetalum paraguayense E. Walther on CCl4-induced liver damage and inflammation. J Ethnopharmacol 2011;134(2):379-385.
  34. Kuo DH, Kang WH, Shieh PC, Chen FA, Chang CD, Tsai ML, Cheng AN, Ho CT, Pan MH. Protective effect of Pracparatum mungo extract on carbon tetrachloride-induced hepatotoxicity in rats. Food Chem 2010;123(4):1007-1012.
  35. Quan J, Yin X, Xu H. Boschniakia rossica prevents the carbon tetrachloride-induced hepatotoxicity in rats. Exp Toxicol Pathol 2011;63(1-2):53-59.
  36. Shahjahan M, Sabitha KE, Jainu M, Shyamala Devi CS. Effect of Solanum trilobatum against carbon tetrachloride-induced hepatic damage in albino rats. Indian J Med Res 2004;120(3):194-198.
  37. Vuda M, D’souza R, Upadhya S, Kumar V, Rao N, Kumar V, Boillat C, Mungli P. Hepatoprotective and antioxidant activity of aqueous extract of Hybanthus enneaspermus against CCl4-induced liver injury in rats. Exp Toxicol Pathol 2012;64(7-8):855-859.
  38. Yang L, Wang CZ, Ye JZ, Li HT. Hepatoprotective effects of polyprenols from Ginkgo biloba L. leaves on CCl4-induced hepatotoxicity in rats. Fitoterapia 2011;82(6):834-840.
  39. Qinna NA, Ghanim BY. Chemical induction of hepatic apoptosis in rodents. J Appl Toxicol 2019;39(2):178-190.
  40. Pandit A, Sachdeva T, Bafna P. Drug-induced hepatotoxicity: a review. J Appl Pharm Sci 2012;2(5):233-43.
  41. Abdel-Daim M, Abushouk AI, Reggi R, Yarla NA, Palmery M, Peluso I. Association of antioxidant nutraceuticals and acetaminophen (paracetamol): Friend or foe? J Food Drug Anal 2018;26(2):78-87.
  42. McGill MR, Jaeschke H. Metabolism and disposition of acetaminophen: recent advances in relation to hepatotoxicity and diagnosis. Pharm Res 2013;30(9):2174-2187.
  43. Yoon E, Babar A, Choudhary M, Kutner M, Pyrsopoulos N. Acetaminophen-induced hepatotoxicity: a comprehensive update. J Clin Transl Hepatol 2016;4(2):131.
  44. Wang C, Xu W, Zhang Y, Huang D, Huang K. Poly (ADP-ribosyl) ated PXR is a critical regulator of acetaminophen-induced hepatotoxicity. Cell Death Dis 2018;9(8):819.
  45. Pingili RB, Pawar AK, Challa SR. Effect of chrysin on the formation of N-acetyl-p-benzoquinone imine, a toxic metabolite of paracetamol in rats and isolated rat hepatocytes. Chem Biol Interact 2019(Apr 1);302:123-134.
  46. Wang Y, Wan Y, Ye G, Wang P, Xue X, Wu G, Ye B. Hepatoprotective effects of AdipoRon against d-galactosamine-induced liver injury in mice. Eur J Pharm Sci 2016;93(Oct 10):123-131.
  47. Gujral JS, Farhood A, Jaeschke H. Oncotic necrosis and caspase-dependent apoptosis during galactosamine-induced liver injury in rats. Toxicol Appl Pharm 2003;190(1):37-46.
  48. Malhi H, Gores GJ, Lemasters JJ. Apoptosis and necrosis in the liver: a tale of two deaths? Hepatology 2006;43(2 Suppl 1):S31-S44.
  49. Lu J, Ren DF, Wang JZ, Sanada H, Egashira Y. Protection by dietary Spirulina platensis against d-galactosamine-and acetaminophen-induced liver injuries. Brit J Nutr 2010;103(11):1573-1576.
  50. Taslidere E, Vardi N, Esrefoglu M, Ates B, Taskapan C, Yologlu S. The effects of pentoxifylline and caffeic acid phenethyl ester in the treatment of d-galactosamine-induced acute hepatitis in rats. Hum Exp Toxicol 2016;35(4):353-365.
  51. Raj PV, Nitesh K, Gang SS, Jagani VH, Chandrashekhar HR, Rao JV, Rao CM, Udupa N. Protective role of catechin on d-galactosamine induced hepatotoxicity through a p53 dependent pathway. Ind J Clin Biochem 2010;25(4):349-356.
  52. Liu X, Wang T, Liu X, Cai L, Qi J, Zhang P, Li Y. Biochanin A protects lipopolysaccharide/D-galactosamine-induced acute liver injury in mice by activating the Nrf2 pathway and inhibiting NLRP3 inflammasome activation. Int Immunopharmacol 2016;38(23 Jun):324-331.
  53. Matsuda H, Ninomiya K, Shimoda H, Yoshikawa M. Hepatoprotective principles from the flowers of Tilia argentea (linden): structure requirements of tiliroside and mechanisms of action. Bioorg Med Chem 2002;10(3):707-712.
  54. Lekić N, Canová NK, Hořínek A, Farghali H. The involvement of heme oxygenase 1 but not nitric oxide synthase 2 in a hepatoprotective action of quercetin in lipopolysaccharide-induced hepatotoxicity of D-galactosamine sensitized rats. Fitoterapia 2013;87(Jun):20-26.
  55. Son YO, Hwang JM, Choi KC, Lee JC. A Phenolic Acid and Flavonoid Fraction Isolated from Lolium multiflorum Lam. Prevents d-Galactosamine-Induced Liver Damages through the Augmentation of Nrf2 Expression. Ind J Clin Biochem 2019;34(1):68-75.
  56. Shafique H, Ahad A, Khan W, Want MY, Bhatt PC, Ahmad S, Panda BP, Mujeeb M. Ganoderic acid-loaded solid lipid nanoparticles ameliorate d-galactosamine induced hepatotoxicity in Wistar rats. J Drug Deliv Sci Tech 2019;50(April):48-
  57. Sreejith M, Marathakam A, Kannapan N, Harikrishnan V. Hepatoprotective Activity of Methanolic Extract of Flacourtia sepiaria ROXB against D-galactosamine Induced Oxidative Stress in Rats. Int J Pharma Res Health Sci 2018;6(1):2143-2147.
  58. Yang SQ, Lin HZ, Lane MD, Clemens M, Diehl AM. Obesity increases sensitivity to endotoxin liver injury: implications for the pathogenesis of steatohepatitis. Proc Natl Acad Sci 1997;94(6):2557-2562.
  59. Shang Y, Liu Y, Du L, Wang Y, Cheng X, Xiao W, Wang X, Jin H, Yang X, Liu S, Chen Q. Targeted expression of uncoupling protein 2 to mouse liver increases the susceptibility to lipopolysaccharide/galactosamine‐induced acute liver injury. Hepatology 2009;50(4):1204-1216.
  60. Tiegs G, Wolter M, Wendel A. Tumor necrosis factor is a terminal mediator in galactosamine/endotoxin-induced hepatitis in mice. Biochem Pharmacol 1989;38(4):627-631.
  61. Wang H, Xu DX, Lu JW, Zhao L, Zhang C, Wei W. N‐Acetylcysteine attenuates lipopolysaccharide‐induced apoptotic liver damage in D‐galactosamine‐sensitized mice 1. Acta Pharmacol Sin 2007;28(11):1803-1809.
  62. Tang F, Fan K, Wang K, Bian C. Amygdalin attenuates acute liver injury induced by D-galactosamine and lipopolysaccharide by regulating the NLRP3, NF-κB, and Nrf2/NQO1 signaling pathways. Biomed Pharmacother 2019;111(March):527-536.
  63. Li Z, Feng H, Wang Y, Shen B, Tian Y, Wu L, Zhang Q, Jin M, Liu G. Rosmarinic acid protects mice from lipopolysaccharide/d-galactosamine-induced acute liver injury by inhibiting MAPKs/NF-κB and activating Nrf2/HO-1 signaling pathways. Int Immunopharmacol 2019;67(Feb):465-472.
  64. Bai J, Wang X, Hao M, Li H, Cheng G, Liu D, Yang Y, Li Y. Forsythiaside attenuates Escherichia coli lipopolysaccharide-induced liver acute inflammatory response in chicken. Eur J Inflamm 2019;17(12):1-7.
  65. Vinothkumar V, Monoharan S, Sindhu G, Nirmal MR, Vetrichelvi V. Geraniol modulates cell proliferation, apoptosis, inflammation, and angiogenesis during 7, 12-dimethylbenz [a] anthracene-induced hamster buccal pouch carcinogenesis. Mol Cell Biochem 2012;369(1-2):17-25.
  66. Jiang K, Zhang T, Yin N, Ma X, Zhao G, Wu H, Qiu C, Deng G. Geraniol alleviates LPS-induced acute lung injury in mice via inhibiting inflammation and apoptosis. Oncotarget 2017;8(41):71038.
  67. Li Y, Wang N, Jiang Y. Geraniol protects against lipopolysaccharide and D-galactosamine-induced fulminant hepatic failure by activating PPARγ. Microb Pathog 2019;128(March):7-12.
  68. Wang CL, Li D, Wang CD, Xiao F, Zhu JF, Shen C, Zuo B, Cui YM, Wang H, Gao Y, Hu GL, Zhang XL, Chen XD. Anti-inflammatory and anti-osteoarthritis effects of tectorigenin. Biol Open 2017;6(8):1130-1136.
  69. Zhang L, Zhao Y, Fan L, Xu K, Ji F, Xei Z, Ouyang X, Wu D, Li L. Tectorigenin protects against experimental fulminant hepatic failure by regulating the TLR4/mitogen‐activated protein kinase and TLR4/nuclear factor‐κB pathways and autophagy. Phytother Res 2019;33(4):1055-1064.
  70. Ichimura R, Mizukami S, Takahashi M, Taniai E, Kemmochi S, Mitsumori K, Shibutani M. Disruption of Smad-dependent signaling for growth of GST-P-positive lesions from the early stage in a rat two-stage hepatocarcinogenesis model. Toxicol Appl Pharmacol 2010;246(3):128-140.
  71. Zargar S, Wani TA, Alamro AA, Ganaie MA. Amelioration of thioacetamide-induced liver toxicity in Wistar rats by rutin. Int J Immunopathol Pharmacol 2017;30(3):207-214.
  72. Koblihová E, Mrázová I, Vernerová Z, Ryska M. Acute liver failure induced by thioacetamide: selection of optimal dosage in Wistar and Lewis rats. Physiol Res 2014;63(4):491-503.
  73. Shirai M, Matsuoka M, Makino T, Kai K, Teranishi M, Takasaki W. Hepatic glutathione contributes to attenuation of thioacetamide-induced hepatic necrosis due to suppression of oxidative stress in diet-induced obese mice. J Toxicol Sci 2015;40(4):509-521.
  74. Yang HY, Kim KS, Lee YH, Park JH, Kim JH, Lee SY, Kim YM, Kim IS, Kacew S, Lee BM, Kwak JH, Yoon K, Kim HS. Dendropanax morbifera ameliorates thioacetamide-induced hepatic fibrosis via TGF-β1/Smads pathways. Int J Biol Sci 2019;15(4):800.
  75. Salunkhe A, Patil R. Hepatoprotective effect of ethanolic extract of Andrographis paniculata against thioacetamide-induced toxicity in the liver of albino rats. Res J Life Sci Bioinform Pharm Chem Sci 2018;4(6):549-559.
  76. Kerr WC, Fillmore KM, Marvy P. Beverage‐specific alcohol consumption and cirrhosis mortality in a group of English‐speaking beer‐drinking countries. Addiction 2000;95(3):339-346.
  77. Seth D, Hogg PJ, Gorrell MD, McCoughan GW, Haber PS. Direct effects of alcohol on hepatic fibrinolytic balance: implications for alcoholic liver disease. J Hepatol 2008;48(4):614-627.
  78. Stickel F, Moreno C, Hampe J, Morgan MY. The genetics of alcohol dependence and alcohol-related liver disease. J Hepatol 2017;66(1):195-211.
  79. Sid B, Verrax J, Calderon P. Role of oxidative stress in the pathogenesis of alcohol-induced liver disease. Free Radic Res 2013;47(11):894-904.
  80. Gao B, Bataller R. Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology 2011;141(5):1572-1585.
  81. Mandayam S, Jamal MM, Morgan TR. Epidemiology of alcoholic liver disease. Semin Liver Dis 2004;24(3):217-232.
  82. Guo R, Xu X, Babcock SA, Zhang Y, Ren J. Aldehyde dedydrogenase-2 plays a beneficial role in ameliorating chronic alcohol-induced hepatic steatosis and inflammation through regulation of autophagy. J Hepatol 2015;62(3):647-656.
  83. Beier JI, McClain CJ. Mechanisms and cell signaling in alcoholic liver disease. Biol Chem 2010;391(11):1249-1264.
  84. Guo R, Zhong L, Ren J. Overexpression of aldehyde dehydrogenase‐2 attenuates chronic alcohol exposure‐induced apoptosis, change in Akt and Pim signaling in liver. Clin Exp Pharm Physiol 2009;36(5-6):463-468.
  85. Powell CL, Bradford BU, Craig CP, Tsuchiya M, Uehara T, O’Connell TM, Pogribny IP, Melnyk S, Koop DR, Bleyle L, Threadgill DW, Rusyn I. Mechanism for prevention of alcohol-induced liver injury by dietary methyl donors. Toxicol Sci 2010;115(1):131-139.
  86. Yin HQ, Kim YC, Chung YS, Kim YC, Shin YK, Lee BH. Honokiol reverses alcoholic fatty liver by inhibiting the maturation of sterol regulatory element-binding protein-1c and the expression of its downstream lipogenesis genes. Toxicol Appl Pharmacol 2009;236(1):124-130.
  87. Tang Y, Li Y, Yu H, Gao C, Liu L, Xing M, Liu L, Yao P. Quercetin attenuates chronic ethanol hepatotoxicity: implication of “free” iron uptake and release. Food Chem Toxicol 2014;67(May):131-138.
  88. Surapaneni K, Jainu M. Comparative effect of pioglitazone, quercetin, and hydroxy citric acid on the status of lipid peroxidation and antioxidants in experimental non-alcoholic steatohepatitis. J Physiol Pharmacol 2014;65(1):67-74.
  89. Liu J, Wang X, Liu R, Liu Y, Zhang T, Fu H, Hai C. Oleanolic acid co-administration alleviates ethanol-induced hepatic injury via Nrf-2 and ethanol-metabolizing modulating in rats. Chem Biol Interact 2014;221(Aug):88-98.
  90. Kao ES, Hsu JD, Wang CJ, Yang SH, Cheng SY, Lee HJ. Polyphenols extracted from Hibiscus sabdariffa L. inhibited lipopolysaccharide-induced inflammation by improving antioxidative conditions and regulating cyclooxygenase-2 expression. Biosci Biotechnol Biochem 2009;73(2):385-3
  91. Park HY, Choi HD, Ecom H, Choi I. Enzymatic modification enhances the protective activity of citrus flavonoids against alcohol-induced liver disease. Food Chem 2013;139(1-4):231-240.
  92. Cederbaum AI. Molecular mechanisms of the microsomal mixed-function oxidases and biological and pathological implications. Redox Biol 2015;4(Nov):60-73.
  93. Das S, Seth RK, Kumar A, Kadiiska MB, Michelotti G, Diehl AM, Chatterjee S. Purinergic receptor X7 is a key modulator of metabolic oxidative stress-mediated autophagy and inflammation in experimental nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2013;305(12):G950-G963.
  94. Abdelmegeed MA, Banerjee A, Jang S, Yoo SH, Yun JW, Gonzalez FJ, Keshavarzian A, Song BJ.  CYP2E1 potentiates binge alcohol-induced gut leakiness, steatohepatitis, and apoptosis. Free Radic Biol Med 2013;65(Sept):1238-1245.
  95. Purohit V, Brenner DA. Mechanisms of alcohol‐induced hepatic fibrosis: a summary of the Ron Thurman Symposium. Hepatology 2006;43(4):872-878.
  96. Gaitantzi H, Meyer C, Rakoczy P, Thomas M, Wahl K, Wandrer F, Bantel H, Alborzinia H, Wölfl S, Ehnert S, Nüssler A, Bergheim I, Ciuclan L, Ebert M, Breitkopf-Heinlein K, Dooley S. Ethanol sensitizes hepatocytes for TGF-β-triggered apoptosis. Cell Death Dis 2018;9(2):51.
  97. Roberts RA, Ganey PE, Kamendulis LM, Rusyn I, Klaunig JE. Role of the Kupffer cell in mediating hepatic toxicity and carcinogenesis. Toxicol Sci 2006;96(1):2-15.
  98. Malaguarnera G, Cataudella E, Giordano M, Nunnari G, Chisari G, Malaguarnera M. Toxic hepatitis in occupational exposure to solvents. World J Gastroenterol 2012;18(22):2756.
  99. Subramaniyan V, Middha A. Chronic ethanol consumption-induced hepatotoxicity and protective effect of Boswellia serrata. Nat J Physiol Pharm Pharmacol 2016;6(2):170.
  100. Tang Y, Gao C, Xing M, Li Y, Zhu L, Wang D, Yang X, Liu L, Yao P. Quercetin prevents ethanol-induced dyslipidemia and mitochondrial oxidative damage. Food Chem Toxicol 2012;50(5):1194-1200.
  101. Yao P, Hao L, Nussler N, Lehmann A, Song F, Zhao J, Neuhaus P, Liu L, Nussler A. The protective role of HO-1 and its generated products (CO, bilirubin, and Fe) in ethanol-induced human hepatocyte damage. Am J Physiol Gastrointest Liver Physiol 2009;296(6):G1318-G1323.
  102. Yao P, Nussler A, Liu L, Hao L, Song F, Schireier A, Nussler N. Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. J Hepatol 2007;47(2):253-261.
  103. Liu S, Hou W, Yao P, Zhang B, Sun S, Nussler AK, Liu L. Quercetin protects against ethanol-induced oxidative damage in rat primary hepatocytes. Toxicol In Vitro 2010;24(2):516-522.
  104. Abdullah A, Alrawaiq NS, Atia A. Heme oxygenase 1 gene expression in mice liver is differently affected by equal doses of phytochemicals. Pharmacophore 2018;9(5):13-17.
  105. Abdullah A, Alrawaiq NA, Atia A. The effect of administration of an equal dose of different classes of phytochemicals on heme oxygenase-1 gene and protein expression in mice liver. Asian J Pharm Clin Res 2019;12(3):256-260.
  106. Molina MF, Sanchez-Reus I, Iglesias I, Benedi J. Quercetin, a flavonoid antioxidant, prevents and protects against ethanol-induced oxidative stress in mouse liver. Biol Pharm Bull 2003;26(10):1398-1402.
  107. Alrawaiq NS, Abdullah  A. A review of flavonoid quercetin: metabolism, bioactivity, and antioxidant properties. Int J Pharm Tech Res 2014;6(3):933–941.
  108. Hackett E, Twedt D, Gustafson D. Milk thistle and its derivative compounds: a review of opportunities for treatment of liver disease. J Vet Intern Med 2013;27(1):10-16.
  109. Song Z, Deaciuc I, Song M, Lee DY, Liu Y, Ji X, McClain C. Silymarin protects against acute ethanol‐induced hepatotoxicity in mice. Alcohol Clin Exp Res 2006;30(3):407-413.
  110. Habib-ur-Rehman M, Mahmood T, Salim T, Afzal N, Ali N, Iqbal J, Tahir M, Khan A. Effect of silymarin on serum levels of ALT and GGT in ethanol-induced hepatotoxicity in albino rats. J Ayub Med Coll Abbottabad 2009;21(4):73-75.
  111. Lieber CS, Leo MA, Cao Q, Ren C, DeCarli LM. Silymarin retards the progression of alcohol-induced hepatic fibrosis in baboons. J Clin Gastroenterol 2003;37(4):336-339.
  112. Chen X, Sun CK, Han GZ, Peng JY, Li Y, Liu YX, Lv YY, Liu KX, Zhou Q, Sun HJ. Protective effect of tea polyphenols against paracetamol-induced hepatotoxicity in mice is significantly correlated with cytochrome P450 suppression. World J Gastroenterol 2009;15(15):1829.
  113. Yun JW, Kim YK, Lee BS, Kim CW. Hyun JS, Baik JH, Kim JJ, Kim BH. Effect of dietary epigallocatechin-3-gallate on cytochrome P450 2E1-dependent alcoholic liver damage: enhancement of fatty acid oxidation. Biosci Biotechnol Biochem 2007;71(12):2999-3006.
  114. Byun EH, Omura T, Yamada K, Tachibana H. Green tea polyphenol epigallocatechin‐3‐gallate inhibits TLR2 signaling induced by peptidoglycan through the polyphenol sensing molecule 67‐kDa laminin receptor. FEBS Lett 2011;585(5):814-820.
  115. Byun EB, Choi HG, Sung NY, Byun EH. Green tea polyphenol epigallocatechin-3-gallate inhibits TLR4 signaling through the 67-kDa laminin receptor on lipopolysaccharide-stimulated dendritic cells. Biochem Biophys Res Comm 2012;426(4):480-485.
  116. Li J, Ye L, Wang X, Liu J, Wang Y, Zhou Y, Ho W. (−)-Epigallocatechin gallate inhibits endotoxin-induced expression of inflammatory cytokines in human cerebral microvascular endothelial cells. J Neuroinflammation 2012;9(1):161.
  117. Tsukamoto S, Huang Y, Umeda D, Yamada S, Yamashita S, Kumazoe M, Kim Y, Murata M, Yamada K, Tachibana H. 67-kDa laminin receptor-dependent protein phosphatase 2A (PP2A) activation elicits melanoma-specific antitumor activity overcoming drug resistance. J Biol Chem 2014;289(47):32671-32681.
  118. Gundimeda U, McNeill TH, Fan TK, Deng R, Rayudu D, Chen Z, Cadenas E, Gopalakrishna R. Green tea catechins potentiate the neuritogenic action of brain-derived neurotrophic factor: role of 67-kDa laminin receptor and hydrogen peroxide. Biochem Biophys Res Commun 2014;445(1):218-224.
  119. Alvarez-Suarez JM, Dekanski D, Ristić S, Radonjić NV, Petronijević ND, Giampieri F, Astolfi P, González-Paramás AM, Santos-Buelga C, Tulipani S, Quiles JL, Mezzetti B, Battino M. Strawberry polyphenols attenuate ethanol-induced gastric lesions in rats by activation of antioxidant enzymes and attenuation of MDA increase. PLoS One 2011;6(10):e25878.
  120. Khanal T, Choi JH, Hwang YP, Chung YC, Jeong HG. Saponins isolated from the root of Platycodon grandiflorum protect against acute ethanol-induced hepatotoxicity in mice. Food Chem Toxicol 2009;47(3):530-535.
  121. Zheng J, Cheng J, Zheng S, Feng Q, Xiao X. Curcumin, a polyphenolic curcuminoid with its protective effects and molecular mechanisms in diabetes and diabetic cardiomyopathy. Front Pharmacol 2018;9(May):472.
  122. Nanji AA, Jokelainen K, Tipoe GL, Rahemtulla A, Thomas P, Dannenberg AJ. Curcumin prevents alcohol-induced liver disease in rats by inhibiting the expression of NF-κB-dependent genes. Am J Physiol Gastrointest Liver Physiol 2003;284(2):G321-G327.
  123. Naik R, Mujumdar A, Ghaskadbi S. Protection of liver cells from ethanol cytotoxicity by curcumin in liver slice culture in vitro. J Ethnopharmacol 2004;95:31-37.
  124. Bao W, Li K, Rong S, Yao P, Hao L, Ying C, Zhang X, Nussler A, Liu L.  Curcumin alleviates ethanol-induced hepatocytes oxidative damage involving heme oxygenase-1 induction. J Ethnopharmacol 2010;128(2):549-553.
  125. Rukkumani R, Aruna K, Varma PS, Rajasekaran KN, Menon VP. Comparative effects of curcumin and an analog of curcumin on alcohol and PUFA induced oxidative stress. J Pharm Pharm Sci 2004;7(2):274-283.
  126. Feng X, Li K, Tan F, Zhu M, Zhou J, Lai Y, Zeng L, Ye Y, Huang J, Wu X, Li S. Assessment of hepatoprotective potential of Radix Fici Hirtae on alcohol-induced liver injury in Kunming mice. Biochem Biophys Rep 2018;16(Dec):69-73.
  127. Zhao C, Fan J, Liu Y, Guo W, Cao H, Xiao J, Wang Y, Liu B. Hepatoprotective activity of Ganoderma lucidum triterpenoids in alcohol-induced liver injury in mice, an iTRAQ-based proteomic analysis. Food Chem 2019;271(Jan):148-156.
  128. Baskaran UL, Sabina EP. Clinical and experimental research in antituberculosis drug-induced hepatotoxicity: a review. J Integr Med 2017;15(1):27-36.
  129. Ramanathan R, Sivanesan K. Evaluation of ameliorative ability of Silibinin against zidovudine and isoniazid-induced hepatotoxicity and hyperlipidaemia in rats: Role of Silibinin in Phase I and II drug metabolism. Chem Biol Interact 2017;273(Aug):142-153.
  130. Steele MA, Burk RF, DesPrez RM. Toxic hepatitis with isoniazid and rifampin: a meta-analysis. Chest 1991;99(2):465-471.
  131. Harrington T, Manangan L, Jerab J, Navin T, Powell K. Severe isoniazid-associated liver injuries among persons being treated for latent tuberculosis infection-United States, 2004-2008. MMWR 2010;59(8):224.
  132. Westphal J, Vetter D, Brogard J. Hepatic side-effects of antibiotics. J Antimicrob Chemother 1994;33(3):387-401.
  133. Sarich TC, Youssefi M, Zhou T, Adams SP, Wall RA, Wright JM. Role of hydrazine in the mechanism of isoniazid hepatotoxicity in rabbits. Arch Toxicol 1996;70(12):835-840.
  134. Bhadauria S, Singh G, Sinha N, Srivastava S. Isoniazid induces oxidative stress, mitochondrial dysfunction, and apoptosis in Hep G2 cells. Cell Mol Biol (Noisy-le-Grand) 2007;53(1):102-114.
  135. Verma AK, Yadav A, Dewangan J, Singh SV, Mishra M, Singh PK, Rath SK. Isoniazid prevents Nrf2 translocation by inhibiting ERK1 phosphorylation and induces oxidative stress and apoptosis. Redox Biol 2015(Dec);6:80-92.
  136. Wang C, Fan RQ, Zhang YX, Nie H, Li K. Naringenin protects against isoniazid-and rifampicin-induced apoptosis in hepatic injury. World J Gastroenterol 2016;22(44):9775.
  137. Darvin SS, Esakkimuthu S, Toppo E, Balakrishna K, Paulraj MG, Pandikumar P, Ignacimuthu S, Al-Dhabi NA. Hepatoprotective effect of lawsone on rifampicin-isoniazid induced hepatotoxicity in in-vitro and in vivo models. Environ Toxicol Pharmacol 2018;61(July):87-94.
  138. Nwidu LL, Oboma YI. Telfairia occidentalis (Cucurbitaceae) pulp extract mitigates rifampicin-isoniazid-induced hepatotoxicity in an in vivo rat model of oxidative stress. J Integr Med 2019;17(1):46-56.
  139. Nwidu LL, Teme RE. Hot aqueous leaf extract of Lasianthera africana (Icacinaceae) attenuates rifampicin-isoniazid-induced hepatotoxicity. J Integr Med 2018;16(4):263-272.
  140. Urfi MK, Mujahid M, Rahman MA, Rahman MA. The role of Tamarix gallica leaves extract in liver injury induced by rifampicin plus isoniazid in Sprague Dawley rats. J Diet Suppl 2018;15(1):24-33.
  141. Agúndez JA, Lucena MI, Martínez C, Andrade RJ, Blanca M, Ayuso P, García-Martín E. Assessment of nonsteroidal anti-inflammatory drug-induced hepatotoxicity. Expert Opin. Drug Metab Toxicol 2011;7(7):817-828.
  142. Maity T, Ahmad A, Pahari N, Ganguli S. Hepatoprotective activity of mikania scandens (L.) wild. against diclofenac sodium-induced liver toxicity in rats. Asian J Pharm Clin Res 2012;5(2):185-189.
  143. Husna M, Sumera S, Laiba S, Anam A. The effect of crude Nigella sativa oil against the acute toxicity of diclofenac sodium and ibuprofen on the livers of albino mice. Slov Vet Res 2017;54(1):21-27
  144. Sadasivan S, Latha PG, Sasikumar JM, Rajashekaran S, Shyamal S, Shine VJ. Hepatoprotective studies on Hedyotis corymbosa (L.) Lam. J Ethnopharmacol 2006;106(2):245-249.
  145. Gomaa S. Adverse effects induced by diclofenac, ibuprofen, and paracetamol toxicity on immunological and biochemical parameters in Swiss albino mice. J Basic Appl Zool 2018;79(1):5.
  146. Aprioku J, Uche F. Renal effects of non-steroidal anti-inflammatory drugs in albino rats. Br J Pharm Res 2013;3(3):314-325.
  147. Fries J. Assessing and understanding patient risk. Scand J Rheumatol 1992;21(Suppl 92):21-24.
  148. Orinya OA, Adenkola AY, Ogbe RJ. Haematological and biochemical studies on the effect of diclofenac sodium on Wistar Rattus norvegicus. Int J Biol Chem Sci 2016;10(5):2231-2242.
  149. Oaks JL, Meteyer CU, Rideout BA, Shivaprasad HL, Gilbert M, Virani M, Watson RT, Khan AA. Diagnostic investigation of vulture mortality: the anti-inflammatory drug diclofenac is associated with visceral gout. Raptors worldwide World Working Group on Birds of Prey and Owls, Berlin, Germany. Blaine, WA: Hancock House Publishers 2004;241-243.
  150. Bhogaraju A, Nazeer S, Al-Baghdadi Y, Rahman M, Wrestler F, Patel N. Diclofenac-associated hepatitis. South Med J 1999;92(7):711-713.
  151. Aydin G, Gökçimen A, Öncü M, Çicek E, Karahan N, Gökalp O. Histopathologic changes in liver and renal tissues induced by different doses of diclofenac sodium in rats. Turk J Vet Anim Sci 2003;27(5):1131-1140.
  152. Bort R, Ponsoda X, Jover R, Gómez-Lechón MJ, Castell JV. Diclofenac toxicity to hepatocytes: a role for drug metabolism in cell toxicity. J Pharmacol Exp Ther 1999;288(1):65-72.
  153. El‐Hadary AE, Ramadan MF. Antioxidant traits and protective impact of Moringa oleifera leaf extract against diclofenac sodium‐induced liver toxicity in rats. J Food Biochem 2018; 43(2):e12704.
  154. Ogbe RJ, Luka CD, Adoga GI. Effect of aqueous ethanol extract of Dialium guineense leaf on diclofenac-induced oxidative stress and hepatorenal injuries in Wistar rats. Comp Clin Path 2019;28(1):241-248.
  155. Tanley SW, Diederichs K, Kroon-Batenburg LM, Levy C, Schreurs AM, Helliwell JR. Carboplatin binding to histidine. Acta Crystallogr F Struct Biol Commun 2014;70(9):1135-1142.
  156. Chen X, Wang J, Fu Z, Zhu B, Wang J, Guan S, Hua Z. Curcumin activates DNA repair pathway in bone marrow to improve carboplatin-induced myelosuppression. Sci Rep 2017;7(1):17724.
  157. Rajeswaran A, Trojan A, Burnand B, Giannelli M. Efficacy and side effects of cisplatin-and carboplatin-based doublet chemotherapeutic regimens versus non-platinum-based doublet chemotherapeutic regimens as first-line treatment of metastatic non-small cell lung carcinoma: a systematic review of randomized controlled trials. Lung Cancer 2008;59(1):1-11.
  158. Rabik CA, Dolan ME. Molecular mechanisms of resistance and toxicity associated with platinating agents. Cancer Treat Rev 2007;33(1):9-23.
  159. Brock PR, Knight KR, Freyer DR, Campbell KC, Steyger PS, Blakley BW, Rassekh SR, Chang KW, Fligor BJ, Rajput K, Sullivan M, Neuwelt EA. Platinum-induced ototoxicity in children: a consensus review on mechanisms, predisposition, and protection, including a new International Society of Pediatric Oncology Boston ototoxicity scale. J Clin Oncol 2012;30(19):2408.
  160. Shen DW, Pouliot LM, Hall MD, Gottesman MM. Cisplatin resistance: a cellular self-defense mechanism resulting from multiple epigenetic and genetic changes. Pharmacol Rev 2012;64(3):706-721.
  161. McWhinney SR, Goldberg RM, McLeod HL. Platinum neurotoxicity pharmacogenetics. Mol Cancer Ther 2009;8(1):10-16.
  162. dos Santos NA, Carvalho Rodrigues MA, Martins NM, dos Santos AC. Cisplatin-induced nephrotoxicity and targets of nephroprotection: an update. Arch Toxicol 2012;86(8):1233-1250.
  163. Purena R, Seth R, Bhatt R. Protective role of Emblica Officinalis hydro-ethanolic leaf extract in cisplatin-induced nephrotoxicity in Rats. Toxicol Rep 2018:5(Feb):270-277.
  164. Zhang X, Yamamoto N, Soramoto S, Takenaka I. Cisplatin-induced germ cell apoptosis in mouse testes. Arch Androl 2001;46(1):43-49.
  165. Sherif IO, Abdel‐Aziz A, Sarhan OM. Cisplatin‐induced testicular toxicity in rats: the protective effect of arjunolic acid. J Biochem Mol Toxicol 2014;28(11):515-521.
  166. El-Amir YO, Yahia D, Yousef MS. Protective effect of avenanthramides against cisplatin-induced testicular degeneration in rats. J Adv Vet Res 2019;9(1):14-22.
  167. Waseem M, Bhardwaj M, Tabassum H, Raisuddin S, Parvez S. Cisplatin hepatotoxicity mediated by mitochondrial stress. Drug Chem Toxicol 2015;38(4):452-459.
  168. Omar HA, Mohamed WR, Arafa el-SA, Shehata BA, El Sherbiny GA, Arab HH, Elgendy AN. Hesperidin alleviates cisplatin-induced hepatotoxicity in rats without inhibiting its antitumor activity. Pharmacol Rep 2016;68(2):349-356.
  169. Pabla N, Dong Z. Cisplatin nephrotoxicity: mechanisms and renoprotective strategies. Kidney Int 2008;73(9):994-1007.
  170. Guo X, Bai X, Li L, Li J, Wang H. Forskolin protects against cisplatin-induced ototoxicity by inhibiting apoptosis and ROS production. Biomed Pharmacother 2018;99(March):530-536.
  171. Neamatallah T, El-Shitany NA, Abbas AT, Ali SS, Eid BG. Honey protects against cisplatin-induced hepatic and renal toxicity through inhibition of NF-κB-mediated COX-2 expression and the oxidative stress-dependent BAX/Bcl-2/caspase-3 apoptotic pathway. Food Funct 2018;9(7):3743-3754.
  172. El-Kashef DH, Sharawy MH. Venlafaxine mitigates cisplatin-induced nephrotoxicity via down-regulating apoptotic pathway in rats. Chem Biol Interact 2018;290(June):110-118.
  173. Liu X, Huang Z, Zou X, Yang Y, Qiu Y, Wen Y. Panax notoginseng saponins attenuate cisplatin-induced nephrotoxicity via inhibiting the mitochondrial pathway of apoptosis. Int J Clin Exp Pathol 2014;7(12):8391.
  174. Song J, Liu D, Feng L, Zhang Z, Jia X, Xiao W. Protective effect of standardized extract of Ginkgo biloba against cisplatin-induced nephrotoxicity. Evid Based Complement Alternat Med 2013; 2013:846126. doi: 10.1155/2013/846126.
  175. Kulms D, Schwarz T. NF‐κB, and Cytokines. Vitam Horm 2006;74(Feb):283-300.
  176. Honma S, Takahashi N, Shinohara M, Nakamura K, Mitazaki S, Abe S, Yoshida M. Amelioration of cisplatin-induced mouse renal lesions by a cyclooxygenase (COX)-2 selective inhibitor. Eur J Pharmacol 2013;715(1-3):181-188.
  177. Yadav YC. Hepatoprotective effect of Ficus religiosa latex on cisplatin-induced liver injury in Wistar rats. Rev Bras Farmacogn 2015;25(3):278-283.
  178. Singh TD, Meitei HT, Sharma AL, Robinson A, Singh LS, Singh TR. Anticancer properties and enhancement of therapeutic potential of cisplatin by leaf extract of Zanthoxylum armatum DC. Biol Res 2015;48(1):46.
  179. Moon JH, Shin JS, Kim JB, Baek NI, Cho YW, Lee YS, Kay HY, Kim SD, Lee KT. Protective effects of 6-hydroxy-1-methylindole-3-acetonitrile on cisplatin-induced oxidative nephrotoxicity via Nrf2 inactivation. Food Chem Toxicol 2013;62(Dec):159-166.
  180. Kouam AF, Njayou FN, Yuan F, Oladejo BO, Mkounga P, Song F, Moundipa PF. Protective Mechanisms of Limonoids from Khaya grandifoliola against Cisplatin-Toxicity in L-02 Hepatocytes: Targeting JNK Activation and Nuclear Translocation of Nrf2. Invest Med Chem Pharmacol 2019;2(Jan):24.
  181. Erisgin Z, Atasever M, Cetinkaya K, Akarca Dizakar SÖ, Omeroglu S, Sahin H. Protective effects of Nigella sativa oil against carboplatin-induced liver damage in rats. Biomed Pharmacother 2019;110(Feb):742-747.
  182. Goyer RA. Lead toxicity: current concerns. Environ Health Perspect 1993;100(Apr):177-187.
  183. Obeng-Gyasi E, Armijos RX, Weigel MM, Filippelli GM, Sayegh MA. Cardiovascular-related outcomes in US adults exposed to lead. Int J Environ Res Public Health 2018;15(4):759.
  184. Qi S, Zheng H, Chen C, Jiang H. Du-Zhong (Eucommia ulmoides Oliv.) cortex extract alleviates Lead acetate-induced bone loss in rats. Biol Trace Elem Res 2019;187(1):172-180.
  185. Sharma A, Sharma V, Kansal L. Amelioration of lead-induced hepatotoxicity by Allium sativum extracts in Swiss albino mice. Libyan J Med 2010;5(1):4621.
  186. Ommati MM, Jamshidzadeh A, Heidari R, Sun Z, Zamiri MJ, Khodaei F, Mousapour S, Ahmadi F, Javanmard N, Yeganeh BS. Carnosine and histidine supplementation blunt lead-induced reproductive toxicity through antioxidative and mitochondria-dependent mechanisms. Biol Trace Elem Res 2019;187(1):151-162.
  187. Kwon SY, Chung JH. Erythrophagocytosis of lead-exposed erythrocytes by renal tubular cells may contribute to lead-induced nephrotoxicity. Environ Health Perspect 2015;123(2):120-7.
  188. Berrahal AA, Lasram M, El Elj N, Kerkeni A, Gharbi N, El-Fazâa S. Effect of age‐dependent exposure to lead on hepatotoxicity and nephrotoxicity in male rats. Environ Toxicol 2011;26(1):68-78.
  189. Flora G, Gupta D, Tiwari A. Toxicity of lead: a review with recent updates. Interdiscip Toxicol 2012;5(2):47-58.
  190. Hasanein P, Kazemian-Mahtaj A, Khodadadi I. Bioactive peptide carnosin protects against lead acetate-induced hepatotoxicity by abrogation of oxidative stress in rats. Pharm Biol 2016;54(8):1458-1464.
  191. Hsu PC, Guo YL. Antioxidant nutrients and lead toxicity. Toxicology 2002;180(1):33-44.
  192. Omobowale TO, Oyagbemi AA, Akinrinde AS, Saba AB, Daramola OT, Ogunpolu BS, Olopade JO. Failure of recovery from lead-induced hepatoxicity and disruption of erythrocyte antioxidant defense system in Wistar rats. Environ Toxicol Pharmacol 2014;37(3):1202-1211.
  193. Liu B, Jiang H, Lu J, Baiyun R, Li S, Lv Y, Li D, Wu H, Zhang Z. Grape seed procyanidin extract ameliorates lead-induced liver injury via miRNA153 and AKT/GSK-3β/Fyn-mediated Nrf2 activation. J Nutr Biochem 2018;52(Feb):115-123.
  194. Dobrakowski M, Pawlas N, Kasperczyk A, Kozłowska A, Olewińska E, Machoń-Grecka A, Kasperczyk S. Oxidative DNA damage and oxidative stress in lead-exposed workers. Hum Exp Toxicol 2017;36(7):744-754.
  195. Alrawaiq NS, Abdullah A. A review of antioxidant polyphenol curcumin and its role in detoxification. Int J Pharmtech Res 2014;6(1):280-289.
  196. Baxla S, Gora RH, Kerketta P, Kumar N, Roy BK, Patra PH. Hepatoprotective effect of Curcuma longa against lead-induced toxicity in Wistar rats. Vet World 2013;6(9):664-667.
  197. Flora G, Gupta D, Tiwari A. Preventive efficacy of bulk and nano curcumin against lead-induced oxidative stress in mice. Biol Trace Elem Res 2013;152(1):1-40.
  198. Chen C, Lin B, Qi S, He J, Zheng H. Protective effects of salidroside on lead acetate-induced oxidative stress and hepatotoxicity in Sprague-Dawley rats. Biol Trace Elem Res 2019;191(2):426-434.
  199. Aksoy L, Sözbilir NB. Effects of Matricaria chamomilla L. on lipid peroxidation, antioxidant enzyme systems, and key liver enzymes in CCl4-treated rats. Environ Toxicol Chem 2012;94(9):1780-1788.
  200. Al-Seeni MN, El Rabey HA, Zamzami MA, Alnefayee AM. The hepatoprotective activity of olive oil and Nigella sativa oil against CCl 4 induced hepatotoxicity in male rats. BMC Complement Altern Med 2016;16(1):438.
  201. Gomase PV, Rangari VD, Verma P. Phytochemical evaluation, and hepatoprotective activity of fresh juice of young stem (tender) bark of Azadirachta indica A. Juss. J Pharm Pharm Sci 2011;3(2): 55-59.
  202. Agbafor KN, Ogbanshi ME, Akubugwo EI. Phytochemical screening, hepatoprotective and antioxidant effects of leaf extracts of Zapoteca portoricensis. Adv Biol Chem 2014;4(1):35-39.
  203. Yang J,  Li Y, Wang F,  Wu C. Hepatoprotective effects of apple polyphenols on CCl4-induced acute liver damage in mice. J Agric Food Chem 2010;58(10):6525-6531.
  204. Domitrović R, Škoda M, Marchesi VV, Cvijanović O, Pugel EP, Štefan MB. Rosmarinic acid ameliorates acute liver damage and fibrogenesis in carbon tetrachloride-intoxicated mice. Food Chem Toxicol 2013;51(Jan):370-378.
  205. Mihailović V, Katanić J, Mišić D, Stanković V, Mihailović M, Uskoković A, Arambašić J, Solujić S, Mladenović M, Stanković N. Hepatoprotective effects of secoiridoid-rich extracts from Gentiana cruciata L. against carbon tetrachloride-induced liver damage in rats. Food Funct 2014;5(8):1795-1803.
  206. Domitrović R, Jakovac H, Marchesi VV, Šain I, Romić Ž, Rahelić D. Preventive and therapeutic effects of oleuropein against carbon tetrachloride-induced liver damage in mice. Pharmacol Res 2012;65(4):451-464.
  207. Jalali Ghassam B, Ghaffari H, Prakash HS, Kini KR. Antioxidant and hepatoprotective effects of Solanum xanthocarpum leaf extracts against CCl4-induced liver injury in rats. Pharm Biol 2014;52(8):1060-1068.
  208. Atawodi SE, Liman ML, Onyike EO. Antioxidant effects of Tamarindus indica following acute and chronic carbon tetrachloride-induced liver injury. Int J Agric Biol 2013;15(3):410-418.
  209. Adebayo HA, Song FH, Liu XT, Dai QH, Huang P, Zhang JY, Zhang LX. Citrullus Ianatus Extract Reverses Oxidative and Haematological Dysfunction in Carbon Tetrachloride Induced Liver Damaged Rats. Int J Pharmacol 2014;10(4):218-224.
  210. He Q, Li Y, Liu J, Zhang P, Yan S, He X, Zhang A. Hepatoprotective activity of Lophatherum gracile leaves of ethanol extracts against carbon tetrachloride-induced liver damage in mice. Int J Pharmacol 2016;12(4):387-393.
  211. Parmar M, Shah P, Thakkar V, Al-Rejaie S, Gandhi T. Hepatoprotective potential of methanolic extract of vetiveria zizanioides roots against carbon tetrachloride-induced acute liver damage in rats. Dig J Nanomater Bios 2013;8(2):835-844
  212. Hsouna AB, Mongi S, Culioli G, Blache Y, Ghlissi Z, Chaabane R, El Feki A, Jaoua S, Trigui M. Protective effects of ethyl acetate fraction of Lawsonia inermis fruits extract against carbon tetrachloride-induced oxidative damage in rat liver. Toxicol Ind Health 2016;32(4):694-706.
  213. Yusufoglu HS, Soliman GA, Foudah AI, Abdelkader MS, Alam A, Salkini MA. Anti-inflammatory and hepatoprotective potentials of the aerial parts of Silene villosa Caryophyllaceae methanol extract in rats. Trop J Pharm Res 2018;17(1):117-125.
  214. Saleem M, Naureen H, Khan A, Noreen F, Ali F, Al Darmahi A. Protective effect of Parthenium hysterophorus against carbon tetrachloride-and paracetamol-induced hepatotoxicity in rabbits. Trop J Pharm Res 2018;17(3):467-473.
  215. Zhu CS, Liu K, Wang JL, Li JF, Liu MF, Hao N, Lin YX, Xiao ZF. Antioxidant activities and hepatoprotective potential of Dracocephalum rupestre Hance extract against CCl4‐induced hepatotoxicity in Kunming mice. J Food Biochem 2018;42(2):e12484.
  216. Li Y, Li Z, Li C, Ma X, Chang Y, Shi C, He J, Li R, Muhammad I, Liu F. Evaluation of hepatoprotective activity of Syringa oblata leaves ethanol extract with the indicator of glutathione S-transferase A1. Rev Bras Farmacogn 2018;28(4):489-494.
  217. Nwidu LL, Elmorsy E, Oboma YI, Carter WG. Hepatoprotective and antioxidant activities of Spondias mombin leaf and stem extracts against carbon tetrachloride-induced hepatotoxicity. J Taibah Univ Sci 2018;13(3):262-271.
  218. Ali SL, Uma C, Sai VB, Jyothi G, Krishna V, Ashajyothi U, Sri S. Hepatoprotective Activity of Hydroalcoholic Extract of Leaves of Urena Lobata Plant on Carbon Tetrachloride Induced Hepatotoxicity in Albino Rats. Int J Pharma Res Health Sci 2018;6(5):2792-2795.
  219. Sheela D, Udhayakumari F. Evaluation of Hepatoprotective and antioxidant activity of Sauropus bacciformis whole plant methanolic extracts against CCL4: Induced liver injury in rats. Int J Pharm Pharm Sci 2018;3(1):51-55.
  220. Tir M, Feriani A, Labidi A, Mufti A, Saadaoui E, Nasri N, Khaldi A, El Cafsi M, Tlili N. Protective effects of phytochemicals of Capparis spinosa seeds with cisplatin and CCl4 toxicity in mice. Food Biosci 2019;28(Apr): 42-48.
  221. Vidičević S, Tošić J, Stanojević Ž, Isaković A, Mitić D, Ristić D, Dekanski D. Standardized Olea europaea L. leaf extract exhibits protective activity in carbon tetrachloride-induced acute liver injury in rats: the insight into potential mechanisms. Arch Physiol Biochem 2019; 11(Jan):1-9.
  222. Suurbaar J, Donkor AM, Donkor M, Saeed M, Alimatu-Sadia F, Samuel DF. Effect of methanol extract of anarcardium occidentale (cashew) stem bark on some biochemical parameters of carbon tetrachloride-induced hepatotoxicity in rats. Int J Pharm Sci Rev Res 2018;9(9):3689-3695.
  223. Maurya H. Pharmacological evaluation of Allium cepa extract as hepatoprotective potential in albino rats. Int J Health Clin Res 2018;1(2):7-13.
  224. Rashid N, Siddique W, Zaheer Z. Evaluation of effect of Argyrolobiumroseum Aqueous Extract on Carbon Tetrachloride induced liver injury in rabbits. Pak J Med Health Sci 2018;12(1):161-165.
  225. Ahmed SA, Chakravarthy K. Simultaneous evaluation, estimation of hepatoprotective effects of kyllinga brevifolia, and antiepileptic activity of rorippa sarmentosa extracts in rats. Glob J Res Anal 2018;6(6):15-19.
  226. Kamble V, Rao BG. Effect of roots of origanum Majorana in methanolic and ethyl acetate extracts on ccl4-induced hepatotoxicity in rats. Int J Pharm Sci Drug Res 2018;10(2):51-56.
  227. Ayyanna C, Khatoon S, Reddy S. Evaluation of the hepatoprotective and antioxidant activity of ethanolic extract of Nerium indicum Mill. leaves against carbon tetrachloride-induced hepatotoxicity in albino rats. Int J Res Pharm Sci 2018;9(4):1088-1095.
  228. Deniz GY, Laloglu E, Koc K, Nadaroglu H, Geyikoglu F. The effect of black mulberry (Morus nigra) extract on carbon tetrachloride-induced liver damage. Arch Biol Sci 2018;70(2):371-378.
  229. Slama K, Boumendjel M, Taibi F, Boumendjel A, Messarah M. Atriplex halimus aqueous extract abrogates carbon tetrachloride-induced hepatotoxicity by modulating biochemical and histological changes in rats. Arch Physiol Biochem 2020;126(Feb):49-60.
  230. Sobeh M, Youssef FS, Esmat A, Petruk G, El-Khatib AH, Monti DM, Ashour ML, Wink M. High-resolution UPLC-MS/MS profiling of polyphenolics in the methanol extract of Syzygium samarangense leaves and its hepatoprotective activity in rats with CCl4-induced hepatic damage. Food Chem Toxicol 2018;113(Mar):145-153.
  231. Mohapatra SS, Roy RK, Mohan P, Upadhyaya TN, Sarma J. Phytochemical Analysis and Hepatoprotective Effect of Hydroethanolic Extract of Stem Bark of Oroxylum indicum. Int J Curr Microbiol App Sci 2018;7(1):1000-1006.
  232. Mohammed MM, Ibrahim NA, Ali SA, Hamed MA, El‐Rigal NS. Triacylglycerols of the seed oil of Linum grandiflorum Desf.: Their composition, cytotoxicity, and hepatoprotective activity. J Food Biochem 2018; 42(1-2):e12525.
  233. Heibatollah S, Reza NM, Izadpanah G, Sohailla S. Hepatoprotective effect of Cichorium intybus on CCl4-induced liver damage in rats. Afr J Biochem Res 2008;2(6):141-144.
  234. Rajesh K, Swamy V, Shivakumar S, Inamdar JV, Kurnool NA. Hepatoprotective and antioxidant activity of ethanol extract of Mentha arvensis leaves against carbon tetrachloride-induced hepatic damage in rats. Int J Pharmtech Res 2013;5(2):426-430.
  235. Khalid A, Shahid S, Khan SA, Kanwal S, Yaqoob A, Rasool ZG, Rizwan K. Antioxidant activity and hepatoprotective effect of Cichorium intybus (Kasni) seed extract against carbon tetrachloride-induced liver toxicity in rats. Trop J Pharm Res 2018;17(8):1531-1538.
  236. Ogaly HA, Eltablawy NA, Abd-Elsalam RM. Antifibrogenic Influence of Mentha piperita L. Essential Oil against CCl4-Induced Liver Fibrosis in Rats. Oxid Med Cell Longev 2018 (Apr):4039753. doi: 10.1155/2018/4039753.
  237. Mhalla D, Zouari Bouassida K, Chawech R, Bouaziz A, Makni S, Jlaiel L, Tounsi S, Mezghani Jarraya R, Trigui M. Antioxidant, Hepatoprotective, and Antidepression Effects of Rumex tinnitus Extracts and Identification of a Novel Bioactive Compound. Biomed Res Int 2018; Article ID 7295848, 10 pages, 2018. https://doi.org/10.1155/2018/7295848
  238. Kumar S, Khanna R. IDDF2018-ABS-0265 Beneficial effects of Silybum marianum seed extract against hepatic fibrosis induced by carbon tetrachloride in rats. Br Med J 2018:67(Suppl 2):A26.
  239. Pramono S, Arifah FH, Pribadi FH, Nugroho AE. Hepatoprotective Activity of Curcuma xanthorrhiza Roxb. Paracetamol-induced Liver Damage in Rats and Correlation with Their Chemical Compounds. Thai J Pharm Sci (TJPS) 2018;42(4):188-195.
  240. Alias A, Othman F, Li AR, Kamaruddin A, Yusof R, Hussan F. Supplementation of Psidium guajava (Guava) fruit polysaccharide attenuates paracetamol-induced liver injury by enhancing the endogenous antioxidant activity. Sains Malays 2015;44(8):1129-1136.
  241. González-Ponce HA, Martínez-Saldaña MC, Rincón-Sánchez AR, Sumaya-Martínez MT, Buist-Homan M, Faber KN, Moshage H, Jaramillo-Juárez F. Hepatoprotective effect of Opuntia robusta and Opuntia streptacantha fruits against acetaminophen-induced acute liver damage. Nutrients 2016;8(10):607.
  242. AlSaadi BH, AlHarbi SH, Ibrahim SR, El-Kholy AA, El-Agamy DS, Mohamed GA. Hepatoprotective activity of Costus speciosus (Koen. Ex. Retz.) Against paracetamol-induced liver injury in mice. Afr J Tradit Complement Altern Med 2018;15(2):35-41.
  243. Rao OU, Eswaraiah MC, Prabhakar MC, Santhikrupa D. Hepatoprotective activity of aqueous extract from inflorescence and pollen grains of Casuarina equisetifolia against paracetamol-induced hepatotoxicity in Wistar rats. Int J Pharm Sci Res 2018;9(2):743-747.
  244. Rao AL, Aminabee S, Eswariah MC. Evaluation of Hepatoprotective Activity of Indigofera barberi in Rats against Paracetamol Induced Hepatic Injury. Adv Inv Pha The Medic 2018;1(Jan):1-9.
  245. Rathee D, Kamboj A, Sachdev RK, Sidhu S. Hepatoprotective effect of Aegle marmelos augmented with piperine co-administration in paracetamol model. Rev Bras Farmacogn 2018;28(1):65-72.
  246. Dokunmu TM, Oyelade IF, Ogunlana OO, Bello OA, Ezekiel OM, Oladele FW. Hepatoprotective Potential and Histological Studies of Effects of Celosia Argentea L. on Paracetamol-Induced Liver Damage. Covenant Journal of Physical & Life Sciences (CJPL) 2018;6(1):8-19.
  247. Singh MP, Valte V, Raleng I, RK SL. Hepatoprotective activity of ethanol extract of Pavetta Indica Linn leaves. Indian J Pharm Pharmacol 2018;5(1):37-39.
  248. Pereira DL, Cunha APSD, Cardoso CRP, Rocha CQD, Vilegas W, Sinhorin AP, Sinhorin VDG. Antioxidant and hepatoprotective effects of ethanolic and ethyl acetate stem bark extracts of Copaifera multijuga (Fabaceae) in mice. Acta Amazon 2018;48(4):347-357.
  249. Lin G, Luo D, Liu J, Wu X, Chen J, Huang Q, Su L, Zeng L, Wang H, Su Z. Hepatoprotective Effect of Polysaccharides Isolated from Dendrobium officinale against Acetaminophen-Induced Liver Injury in Mice via Regulation of the Nrf2-Keap1 Signaling Pathway. Oxid. Med Cell Longev 2018;2018:6962439. doi: 10.1155/2018/6962439. 
  250. Sivakumar V, Sadiq AM, Bharathi SD. Hepatoprotective activity of Centella asiatica Linn. against paracetamol-induced liver damage in experimental animals. Emergent Life Sci Res 2018;4(1):19-26.
  251. Simeonova R, Vitcheva V, Kondeva-Burdina M, Krasteva I, Manov V, Mitcheva M. Hepatoprotective and antioxidant effects of saponarin, isolated from Gypsophila trichotoma Wend. on paracetamol-induced liver damage in rats. Biomed Res Int 2013; 2013:757126. doi: 10.1155/2013/757126.
  252. Pal DR, Nahar S, Roy K, Talukder SA, Hossain MM, Paul PC, Eva EO. Hepatoprotective Effect of Nigella Sativa Linn (Kalajira) On Paracetamol-induced Liver Damage. Bangladesh Med J 2011;40(3):52-54.
  253. Rehman JU, Aktar N, Khan MY, Ahmad K, Ahmad M, Sultana S, Asif HM. Phytochemical screening and hepatoprotective effect of Alhagi maurorum boiss (Leguminosae) against paracetamol-induced hepatotoxicity in rabbits. Trop J Pharm Res 2015;14(6):1029-1034.
  254. Islam R, Alam MJ. Evaluation of liver protective activity of Moringa oleifera bark extract in paracetamol-induced hepatotoxicity in rats. BioRxiv 2019; https://doi.org/10.1101/513002.
  255. Rajasekaran A, Periyasamy M. Hepatoprotective effect of ethanolic extract of Trichosanthes lobata on paracetamol-induced liver toxicity in rats. Chin Med 2012;7(1):12.
  256. Prabu K, Kanchana N, Sadiq AM. Hepatoprotective effect of Eclipta alba on paracetamol-induced liver toxicity in rats. J Microbiol Biotechn Res 2017;1(3):75-79.
  257. Kumar KS, Rajakrishnan R, Thomas J, Reddy GA. Hepatoprotective effect of Helicanthus elastica. Bangladesh J Pharmacol 2016;11(2):525-30.
  258. Hamid A, Lee LS, Karim SR, Jufri NF. Hepatoprotective Effects of Zerumbone against Paracetamol-Induced Acute Hepatotoxicity in Rats. Malays J Med Sci 2018;25(2):64-71.
  259. Hena PT, Srivastava M, Ghoshal S. Hepatoprotective, and histopathological activity of ethanol and aqueous extracts of stem of Aloe vera Linn. (Ghee gang war) against paracetamol-induced liver damage in rats. Int J Pharm Bio Sci 2016;3(1):1-7.
  260. Nazari A, Fanaei H, Dehpour AR, Hassanzadeh GRE, Jafari M, Salehi M, Mohammadi M. Chemical composition and hepatoprotective activity of ethanolic root extract of Taraxacum Syriacum Boiss against acetaminophen intoxication in rats. Bratisl Lek Listy 2015;116(1):41-46.
  261. George M,  Joseph L, Deshwal N, Joseph J. Hepatoprotective activity of different extracts of Pterospermum acerifolium against paracetamol-induced hepatotoxicity in albino rats. Pharm Innov J 2016;5(3):32-36.
  262. Li M, Wang S, Li X, Kou R, Wang Q, Wang X, Zhao N, Zeng T, Xie K. Diallyl sulfide treatment protects against acetaminophen-/carbon tetrachloride-induced acute liver injury by inhibiting oxidative stress, inflammation, and apoptosis in mice. Toxicol Res 2019;8(1):67-76.
  263. Dkhil MA, Abdel Moneim AE, Hafez TA, Mubaraki MA, Mohamed WF, Thagfan FA, Al-Quraishy S. Myristica fragrans Kernels Prevent Paracetamol-Induced Hepatotoxicity by Inducing Anti-Apoptotic Genes and Nrf2/HO-1 Pathway. Int J Mol Sci 2019;20(4):993.
  264. Abbas N, Alyousef L, Agabien EM, Ahmed ES, Ahmed AS, Begum A. Comparative study of hepatoprotective effect produced by Cuminum cyminum and Nigella sativa against cisplatin-induced hepatotoxicity; with histopathological studies. Int J Pharm Sci Res 2018;9:393-401.
  265. Abbasi MM, Hassanalilou T, Khordadmehr M, Vardin AM, Kohlan AB, Khalili L. Effects of Cornus mas Fruit Hydro-Methanolic Extract on Liver Antioxidants and Histopathologic Changes Induced by Cisplatin in Rats. Indian J Clin Biochem 2019; 35(1):218-224.
  266. Chandel SS, Shirsat M, Sahu RK, Nayak SS. Modulatory Effect of Dietary Inclusion of Aegle marmelos Fruits against Cisplatin-Induced Hepatotoxicity In Wistar Rats. Ann Hepatol 2018;17(3):482-489.
QR code:

Short Link:
Quick Access

Pharmacophore
ISSN: 2229-5402

Pharmacophore
© 2024 All rights reserved
Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.