Skip to main content
Heliyon logoLink to Heliyon
. 2024 Jun 25;10(13):e33631. doi: 10.1016/j.heliyon.2024.e33631

Advances in the mechanism of emodin-induced hepatotoxicity

Yupeng Wang a,1, Mengchao Zhao b,1, Bo Li a, Xingchao Geng a,
PMCID: PMC11255441  PMID: 39027614

Abstract

Emodin is a naturally occurring anthraquinone derivative and serves as an active component in various traditional Chinese herbal medicines. It is widely known for its broad pharmacological effects, including anti-inflammatory, antioxidant, and anticancer properties. However, high doses and long-term use of emodin can also lead to liver toxicity. Nevertheless, the mechanism of emodin-induced liver toxicity remains unclear at present. This article aims to summarize the toxicological research progress on emodin, with a particular focus on elucidating the mechanisms underlying emodin-induced hepatocyte injury. By providing essential information, the study intends to facilitate further research and safe usage of emodin for researchers and clinical practitioners.

Keywords: Emodin, Frangulic acid, Hepatocyte injury, Liver injury, Hepatotoxicity

Highlights

  • The mechanism of emodin induces hepatocytotoxicity may involve the concurrent inhibition of CDK2, CDK4, and CDK6.

  • Hepatocyte autophagy induced by emodin may also be a mechanism for inducing hepatotoxicity.

  • Interference of emodin with fatty acid metabolism in hepatocytes may be one of the mechanisms inducing hepatotoxicity.

  • Differences in the timing of emodin administration, dosage, and the species on which it acts can affect its effects.

1. Introduction

Emodin (C15H10O5), whose chemical structure is illustrated in Fig. 1, is a naturally occurring anthraquinone derivative. It serves as a vital constituent in various traditional Chinese herbal remedies, including Polygonum multiflorum, Rheum palmatum, and Cassia seeds. Emodin is prevalent in more than 800 types of Chinese medicinal formulations [1](as shown in Table 1). A multitude of prior investigations have ascertained that emodin exhibits a diverse array of pharmacological effects, encompassing its roles as an anticancer agent [2], an anti-inflammatory agent [3], an antiviral agent [4], and an antioxidant [5]. These properties have rendered emodin a valuable ingredient in Chinese medicinal preparations and dietary supplements. However, several experimental and clinical studies have found that high doses and prolonged use of emodin or emodin-containing herbal medicines are associated with hepatocellular damage, and abnormalities in liver function [[6], [7], [8], [9], [10]]. This has triggered the need for evaluation and in-depth studies of the hepatotoxic effects of emodin. Currently, studies on emodin-induced hepatocellular injury have focused on emodin-induced hepatic mitochondrial dysfunction, which leads to oxidative stress and apoptosis, while other aspects, such as emodin-induced endoplasmic reticulum stress, disruption of bile acid metabolism in the hepatocytes, and hepatocellular steatosis, have yet to be investigated in depth. In-depth studies of these aspects may be important for our comprehensive understanding of emodin-induced hepatocyte injury. Table 2 summarizes studies of emodin-induced hepatotoxicity over the past decades.

Fig. 1.

Fig. 1

Chemical structure of emodin.

Table 1.

Chinese herbal medicines containing emodin and related Chinese medicine preparations.

Name of Herbs Number of relevant formulations Examples
Rheum Officinale 2805 Daiokanzoto; Rhubarb draining liver soup
Rhizoma Dioscoreae 269 Rhizoma Dioscoreae Detoxifying Decoction
Polygonum Multiflorum 268 Four substance decoction; Liuwei Dihuang Pill
Lilium Brownie 209 Lily Snow fungus Decoction
Polygonum Cuspidatum 98 Quyu-Huoxue Decoction
Semen Cassia 85 Cassia Seed Xiegan Decoction
Buckwheat 31 Buckwheat Fire Reducing Decoction
Vine of Multiflower Knotweed 24 Shouwu Teng Decoction
Rumex Japonicus Houtt 19 Rumex Japonicus Houtt fire reducing Decoction
Ramie Root 16 Ramie Root Qingre Jiedu Decoction
Artemisia Scoparia 3 Artemisinin Decoction

Table 2.

Studies on the emodin-induced hepatotoxicity.

Reference Cell lines/Animal model Dose of administration duration of administration Pathological characteristic
Yu et al. [11] L02 0–300 μM 48h AST, ALT and ALP abnormal;
Decreased cell survival rate;
Cui et al. [12] L02 0–120 μM 24, 48, 72h Decreased cell survival rate; Apoptosis;
Jiang et al. [13] PHH/Rat 0–120 μM/200 mg/kg 48h/4d Decreased cell survival rate;
AST abnormal;
Inflammatory infiltration;
Steatosis;
Wu et al. [14] HepG2/Rat 0–100 μM/150 mg/kg 48h/28d AST, ALT abnormal;
Decreased cell survival rate;
Inflammatory infiltration;
Apoptosis;
Lin et al. [15] L02 50 μM 24, 48h Increased ROS;
Mitochondrial damage;
Apoptosis;
Wang et al. [16] PMH/Rat/Mice 50μM/150 mg/kg/30, 90, 300 mg/kg 6h/7d/14d Bile acid increased significantly
Hu et al. [17] L02/HepaRG 0–60 μM 48h Apoptosis;
Increased ROS;
Calcium ion overload;
Zhang et al. [18] HepG2 0–70 μM 24h AST, ALT abnormal;
Increased ROS;
Chen et al. [19] HepG2 0–200 μM 48h Decreased cell survival rate;
BUN increased;
Li et al. [20] PRH 10 μM 24h Apoptosis;
Up-regulation on bile acids synthesis
Wang et al. [21] L02/Rat/Mouse, 50μM/20,60,180 mg/kg
/40,120,360 mg/kg
30d/30d Apoptosis;
Increased ROS;
Calcium ion overload;
Dong et al. [22] HepaRG 0–20 μM 8h Increased ROS;
Apoptosis;
Cell cycle arrest
Lin et al. [23] L02/Mouse 0–200μM/25,50 mg/kg 12,24,48h/31d Decreased cell survival rate;
Apoptosis;
MAPK, signal pathway activated
Cui et al. [24] HepG2/L02 0–80 μM 24h Apoptosis;
PI3K/AKT signal pathway inhibited
Ni et al. [25] L02/Zebrafish 50/1.8 μM 48/72h Increased ROS;
Wang et al. [26] HepaRG 100–800 μM 0-7d Block bile acid metabolism pathway;
Chen et al. [27] Rat 1500 mg/kg 28d Steatosis; Cholestasis;
Yang et al. [28] Rat 150,500,1500 mg/kg 28d Inflammatory infiltration;
Steatosis;
Zhang et al. [29] Rat 150 mg/kg 28d Fatty acid β oxidation disorder;
Oxidative stress

PHH: Primary human hepatocytes; ROS: Reactive oxygen species; PMH: Primary human hepatocytes; BUN: Blood urea nitrogen; PRH: Primary rat hepatocytes.

In this review, we conducted a search using “Emodin, Frangulic Acid, Hepatocyte injury, Liver injury, Hepatotoxicity, and Chemical and Drug-induced Liver Injury” as key words in the Pubmed and Embase databases, resulting in the identification of 31 articles. We comprehensively examined the relationship between emodin and hepatotoxicity and discussed its potential mechanisms. Additionally, we examined the influence of factors such as emodin metabolism, drug interactions, dose-response relationships, and individual differences on the hepatotoxic effects of emodin. We aim to provide necessary information to researchers and clinicians by delving into the mechanisms underlying emodin-induced hepatotoxicity, thereby promoting the safe use of emodin and emodin-containing traditional Chinese medicine preparations.

Table 1. Chinese herbal medicines containing emodin and related Chinese medicinal preparations. The Chinese herbal medicines containing emodin and the related Chinese medicinal formulations were searched and compiled from several authoritative databases, such as the Chinese Medicine Pharmacology Database, the Taiwan Chinese Medicine Pharmacology Database, Pharmacopoeia, and China National Knowledge Internet.

2. Mechanism of emodin-induced hepatotoxicity

2.1. Emodin induces hepatocyte apoptosis

2.1.1. Emodin induces mitochondrial damage

Hepatocyte apoptosis refers to an orderly, programmed cell death process, usually achieved through the activation of apoptosis signaling pathways. These pathways include intrinsic and extrinsic pathways. The intrinsic pathway primarily involves a series of proteins and signaling molecules within the cell, such as apoptosis-related proteins associated with mitochondria. The extrinsic pathway typically involves cell death signals triggered by external stimuli, such as cytokines and cell death ligands [30]. Mitochondria are abundant in hepatocytes and are involved in a wide range of cellular physiological activities, including regulation of electrolyte homeostasis, ion transport across membranes, oxygen radical production, and cell signaling. Furthermore, mitochondria provide 80 % of the energy required for cellular life activities, and they are also the main organelles for cellular bio-oxidation and energy conversion [15,22,24,29,31]. Mitochondrial damage may be an obvious trigger for apoptosis.

Research indicates emodin exhibits concentration- and time-dependent cytotoxic effects on normal hepatocytes and hepatocellular carcinoma cells [32,33]. It is noteworthy that the primary mechanism by which emodin exerts hepatocellular cytotoxicity is through inducing apoptosis [[11], [12], [13], [14], [15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26],34]. A study in 2016 found that emodin could increase the ratio of Bax/Bcl2 proteins in HepG2 and primary human hepatocytes, leading to disruption of mitochondrial membrane potential (MMP) and consequent mitochondrial damage. Furthermore, emodin can modulate mitochondria-dependent and death receptor-mediated apoptotic signals by inhibiting the PI3K/AKT pathway and activating the MAPK P38 signaling pathway [24]. A study in 2017 discovered that emodin could accumulate in the mitochondria of HepG2 cells and induce mitochondrial dysfunction by upregulating the mitochondrial matrix protein Cyclophilin D (CypD) [31]. In a study by Ni Jian et al., it was found that emodin could induce apoptosis in HepaRG cells by damaging mitochondria, consistent with the results observed by Liu et al. [22,24]. Additionally, emodin can induce mitochondrial damage and cell apoptosis by inhibiting the oxidative phosphorylation pathway of L02 cells, leading to disruption of adenosine triphosphate (ATP) synthesis [15]. The mechanisms by which emodin induces apoptosis in hepatocytes by damaging mitochondria are illustrated in Fig. 2.

Fig. 2.

Fig. 2

Mechanistic model of emodin-induced apoptosis in hepatocytes. MMP: mitochondrial membrane potential; Cytc: cytochrome C; MPTP: mitochondrial permeability transition pore; PARP: poly (ADP-ribose) polymerase.

2.1.2. Emodin induces DNA damage

DNA damage refers to situations where the structure of DNA molecules is compromised or altered. DNA damage can potentially lead to cellular dysfunction and even cell death. Currently, there are numerous reports about emodin causing DNA damage to hepatocytes, but the underlying mechanisms remain unclear [35]. Research has found that emodin exhibits DNA-damaging properties by stabilizing the TopoⅡ-DNA cleavage complex and inhibiting ATP hydrolysis [36]. Additionally, emodin can significantly upregulate various proteins related to DNA damage detection, DNA damage signal transduction, and DNA damage repair in L02 cells. Excessive reactive oxygen species (ROS) can also cause oxidative DNA damage, leading to cell cycle arrest or delay [37,38]. Similarly, emodin may induce DNA damage in hepatocytes by generating ROS, although this requires further experimental validation. Furthermore, although research has indicated that emodin induces DNA double-strand breaks in mammalian cells in a concentration-dependent manner, whether emodin-induced DNA damage in hepatocytes is correlated with its concentration and duration of exposure remains to be further investigated.

2.1.3. Emodin induces cell cycle arrest

Cell cycle arrest refers to the phenomenon where a cell halts progression to the next phase in its normal lifecycle. Numerous factors contribute to cell cycle arrest, including physiological factors and pathological factors (e.g., DNA damage and abnormal expression of cell cycle regulatory proteins). Fig. 3 depicts the classical regulatory model of the cell cycle: Mitogens initially stimulate the MAPK pathway, leading to the expression of Cyclin D (CycD) and the activation of Cyclin-dependent kinase 4/6 (CDK4/6). Subsequently, the CDK4/6-CycD complex phosphorylates the retinoblastoma protein (Rb1), releasing E2F to promote the transcription of Cyclin E and Cyclin A (CycE, CycA), and activating CDK2. Then, activated CDK2 hyper-phosphorylates Rb1, establishing an efficient and sustained positive feedback loop to ensure the continuous expression of CycE, CycA, and other proteins crucial for the S phase. Upon completion of the S phase, Cyclin A and Cyclin B form complexes with CDK1, driving the progression into mitosis.

Fig. 3.

Fig. 3

Classic model of cell cycle regulation.

A recent study by Sabrina L. has shown that inhibition of CycD -Dependent Kinases 2 (CDK2) alone does not cause cell cycle arrest, as an enhanced CycD -Dependent Kinases 4/6 (CKD4/6) activity allows cells to rapidly overcome the detrimental effects of CDK2 inhibition and complete the cell cycle through a CDK2/4/6-Rb-E2F-dependent mechanism, only the simultaneous inhibition of CDK2 and CDK4/CDK6 can cause cell cycle arrest [39]. Emodin can induce cell cycle arrest at the S and G2/M phases in hepatocytes by damaging DNA and regulating the expression of cell cycle proteins [40]. The previous section has discussed emodin's DNA-damaging effects, so I won't delve into that further here. Studies have shown that emodin induces apoptosis in HepaRG cells while significantly upregulating the expression of Cyclin E and downregulating the expression of Cyclin A and CDK2 [22]. Similarly, emodin induces apoptosis in L02 cells while significantly downregulating the expression of CDK6 protein [15]. Additionally, Manoharan's research group found that emodin can inhibit the expression of CDK4 protein in cells. It is noteworthy that aloe-emodin, which shares a similar chemical structure with emodin, significantly inhibits the expression of CDK4/CDK6 in HepG2 cells [15,22]. Ni Jian et al. discovered that emodin can induce abnormal expression of various proteins in L02 cells that play crucial roles in cell cycle regulation, cell cycle arrest, cell cycle checkpoints, mitotic control, and cell cycle signal transduction [15]. Unfortunately, there is currently no research confirming the concentration-dependent induction of hepatocyte cycle arrest by emodin. However, it has been observed in chronic myelogenous leukemia cells (K562) that emodin increases the proportion of cells in the G0/G1 phase while reducing the proportion of cells in the S and G2/M phases, and this effect is dose-dependent [41].

Based on the evidence presented above, we have reason to believe that the simultaneous inhibition of CDK2, CDK4, and CDK6, along with the modulation of aberrant expression of related cell cycle proteins, may be one of the mechanisms by which emodin induces hepatocyte damage.

2.2. Emodin induces hepatocyte stress injury

2.2.1. Emodin induces oxidative stress

Oxidative stress refers to the phenomenon where the oxidation-reduction balance within cells or tissues is disrupted. The disturbance of the redox state can lead to a series of metabolic disorders, including oxidative stress and disruption of fatty acid oxidation. In recent years, the role of oxidative stress in emodin-induced hepatocyte damage has been confirmed by multiple research groups. For example, the increase in reactive oxygen species (ROS) upregulates the ratio of Bax to Bcl2 in cells, leading to cytochrome C release, which subsequently induces cell death through the mitochondrial apoptotic pathway [[42], [43], [44]]. Additionally, ROS can oxidize two redox-sensitive sites on the mitochondrial permeability transition pore (MPTP), leading to MPTP opening. Subsequently, changes in mitochondrial membrane permeability cause the release of cytochrome C from the mitochondrial matrix, activating related apoptotic factors, specifically inducing cell toxicity and apoptosis [15]. The antioxidant N-acetyl-l-cysteine inhibits cell apoptosis, further demonstrating the damaging effect of ROS on hepatocytes [31]. The nuclear factor erythroid-2 related factor (Nrf2) signaling pathway plays an important regulatory role in cell protection and the expression of antioxidant genes, and is considered a major regulatory mechanism for reducing oxidative stress. Under normal physiological conditions, Nrf2 binds to Kelch-like-ECH-related protein 1 (Keap1), which is a sensor protein against ROS. However, under oxidative stress, Nrf2 dissociates from Keap1 and translocate to the nucleus upon activation. In the nucleus, activated Nrf2 interacts with the muscle aponeurotic fibrosarcoma protein (Maf) and other co-activators, thereby activating genes related to the antioxidant response element (ARE), such as superoxide dismutase (SOD), glutathione S-transferase (GST), heme oxygenase-1 (HO-1), and NAD(P)H quinone dehydrogenase-1 (NQO-1). Studies have shown that high concentrations of emodin can inhibit the nuclear translocation of Nrf2, while low concentrations of emodin have the opposite effect [[45], [46], [47]]. Additionally, similar to acetaminophen, emodin can form complexes with glutathione, leading to glutathione depletion, which may be a key factor in emodin-induced hepatocyte damage [13]. In the latest study, Li et al. reported 1399 genes related to oxidative stress [48]. Building upon their research, we found that 50 μM emodin treatment of L02 cells for 48h resulted in differential expression of 73 proteins related to oxidative stress, further confirming that emodin can induce hepatocyte damage by disrupting the redox balance within hepatocytes.

2.2.2. Emodin induces endoplasmic reticulum stress

Endoplasmic reticulum stress in hepatocytes refers to a state where the endoplasmic reticulum (ER) functions abnormally or becomes overloaded, leading to disruption in protein folding and repair processes within the ER, thereby triggering a cellular stress response. This endoplasmic reticulum dysfunction triggers the activation of a series of cellular stress signaling pathways, including three major sensor proteins: endoplasmic reticulum membrane-associated tyrosine kinase/endoplasmic reticulum membrane-associated ribonucleic acid endonuclease 1 (IRE1), protein kinase RNA-like endonuclease activator (PKR-like endoplasmic reticulum kinase, PERK) and activating transcription factor (ATF). Activation of these sensor proteins triggers a series of responses, including activation of the anti-stress protein CHOP, oxidative stress, inflammation, and regulation of autophagic processes in an attempt to correct endoplasmic reticulum dysfunction and restore cellular homeostasis. However, if endoplasmic reticulum stress persists or is excessively severe, the cell may not be able to respond effectively, leading to cell death or the onset of disease.

Research suggests that the hepatotoxicity of emodin may be associated with the aryl hydrocarbon receptor (AhR) - cytochrome P450 1A1 (CYP1A1) - endoplasmic reticulum (ER) stress pathway. Further investigations have revealed that a toxic metabolite of emodin, 5-hydroxyemodin, can induce hepatocyte damage by activating the PERK-ATF4-CHOP signaling pathway [21]. In addition, Emodin can led to the upregulation of various proteins related to ER stress, such as transmembrane protein 214 (TMEM214) and BCL2 interacting protein 1 (BNIP1) [15]. Additionally, Dong et al. confirmed that a structurally similar compound to emodin, aloe emodin, can induce the upregulation of ER stress-related proteins (Caspase 12 and GRP78), subsequently inducing cell apoptosis [49]. Currently, there is limited research on the induction of ER stress in hepatocytes by emodin, and the mechanisms behind emodin-induced ER stress in hepatocytes require further in-depth investigation.

2.2.3. Emodin induces inflammatory stress

Hepatocellular inflammation stress refers to the cellular stress response triggered by stimuli on hepatocytes. As one of the vital organs of the immune system, the liver exhibits specific responsiveness to inflammation. When the liver is exposed to toxic substances, inflammatory reactions can lead to the production of various inflammatory mediators by hepatocytes. The release of these inflammatory mediators activates a series of cellular signaling pathways. The activation of these pathways induces a cascade of inflammatory responses, including the synthesis of inflammatory mediators, infiltration of inflammatory cells, as well as inflammation-related cellular apoptosis and necrosis.

As is well known, low-dose emodin possesses anti-inflammatory effects [50]. However, there are relatively few reports regarding the effects of high-dose emodin on inflammation. It has been shown that 250 μM emodin can induce activation of the NFκB signaling pathway and upregulation of the IL-6 in L02 cells [51]. In addition, aloe emodin can induce liver toxicity by activating the NFκB inflammatory pathway in zebrafish [52]. Further investigation is needed to understand the dose-response relationship between emodin and inflammatory stress in hepatocytes.

2.3. Emodin induces hepatocyte metabolic disorders

2.3.1. Emodin induces cholestasis

Hepatocellular cholestasis refers to the phenomenon where the normal flow of bile within hepatocytes is obstructed, resulting in the accumulation of bile within the liver. Bile secretion relies on the functionality of the transport systems in hepatocytes and bile duct cells' membranes, as well as the integrity of bile duct tree structure and function [53]. Bile accumulation may cause some degree of damage to hepatocytes, but the exact mechanism remains incompletely understood. Studies have shown that high concentrations of bile acids can activate death receptors, thereby inducing apoptosis in hepatocytes [54,55]. Subsequent research reports have indicated that bile acids under different concentration conditions can induce apoptosis or necrosis in cells [56]. Additionally, recent studies indicate that bile acids can induce inflammatory responses and oxidative stress by activating hepatocytes, thereby leading to liver damage [57,58].

The research conducted by Zhang et al. revealed that emodin inhibits the bile salt export pump (BSEP) potentially by disrupting the crosstalk between AMP-activated protein kinase (AMPK) and Farnesoid X receptor (FXR) in mouse liver, thus indicating a potential pro-cholestasis effect [16]. Multidrug-resistant protein 2 (MRP2), also known as ATP binding cassette subfamily C member 2 (ABCC2), is an ATP-dependent transporter protein that plays a significant role in the transport of various drugs, toxins, and endogenous substances across cell membranes [59]. Studies suggest that MRP2 is crucial for hepatobiliary excretion [60]. Similarly, multidrug-resistant protein 3 (MRP3), or ATP binding cassette subfamily C member 3 (ABCC3), is another ATP-dependent transporter protein that transports various bile salts and provides an alternative pathway for the efflux of bile acids in cholestatic hepatocytes [61]. In in vitro cell experiments, it was found that 50 μM emodin significantly increases the protein expression of MRP2 and MRP3 in HepG2 and L02 cells [14,15]. In in vivo experiments, Tang et al. discovered that continuous oral administration of 150 mg/kg emodin for 28 days induces an increase in both gene expression and protein levels of MRP2 in male rats' livers. On the other hand, Zhang's team found that continuous oral administration of 150 mg/kg emodin for 5 days leads to a decrease in both gene expression and protein levels of MRP2 in the livers of male mice [14,16]. The conflicting results may be attributed to differences in emodin administration duration and animal species.

2.3.2. Emodin induces fatty acid metabolism disorder

Fat acid metabolic disorder refers to abnormalities in processes such as synthesis, oxidation, and transportation of fatty acids, leading to the accumulation of fatty acids in cells. Lipid accumulation resulting from hepatocyte fat acid metabolic disorder can induce lipid peroxidation and ROS generation. Furthermore, lipid accumulation can also trigger endoplasmic reticulum stress and inflammatory responses, causing hepatocyte damage, and even apoptosis. Metabolites produced by abnormal lipid metabolism, such as excess free fatty acids and fatty acid metabolites generated in fatty acid metabolism, exert direct toxic effects on hepatocytes, potentially resulting in cell damage and death.

Currently, research on the induction of hepatocyte fatty acid metabolism disruption by emodin is not common. A few studies suggest that emodin, in combination with dexamethasone (a CYP3A inducer) or BSO (a glutathione depletion agent), can induce hepatic steatosis in rats [13]. Apart from co-administration, emodin alone can also induce hepatic steatosis in rats [14,62]. Additionally, Ni Jian et al.'s research indicates that treatment with 50 μM emodin for 48 h affects the expression of various proteins related to lipid synthesis, lipid transport, lipid storage, and lipid metabolism in L02 cells. This further demonstrates the role of emodin in hepatocyte fat acid metabolic disorder. However, the underlying mechanisms by which emodin induces hepatic steatosis require further investigation.

2.3.3. Emodin induces abnormal expression of hepatocyte metabolic enzymes

The liver is the primary organ for drug metabolism, and metabolic enzymes in hepatocytes play a crucial role in the process. Drug metabolism in the body typically involves two main phases: Phase I metabolism and Phase II metabolism. Phase I metabolism reactions are typically catalyzed by the cytochrome P450 enzyme system in the liver. Phase II reactions typically involve transferases in the liver, such as glucuronyl transferase, sulfotransferase, and methyltransferase. Therefore, abnormalities in the level or activity of hepatic metabolic enzymes may affect cellular metabolic function and trigger cellular damage.

Recent research indicates that the absolute bioavailability of emodin is only 3.2 %. Approximately 56 % of emodin is not absorbed and is primarily excreted in its original form in feces. The absorbed components are mainly metabolized into hydroxylated and glucuronidated metabolites. Enzymes CYP1A2, CYP2E1, UGT1A1, UGT1A9, and UGT2B7 play crucial roles in emodin metabolism [62,63]. Emodin-3-O-β-D-glucuronide is a major glucuronidated metabolite of emodin both in vivo and in vitro and is also a substrate of MRP2. Its toxicity is much lower than that of emodin [14,63]. The study found that emodin can downregulate the gene expression of UGT2B7 in cells, leading to increased exposure of emodin in the body [14]. 5-hydroxyemodin, a hydroxylated metabolite of emodin, has higher toxicity than emodin, and its toxicity mechanism may be related to the activation of the Aryl hydrocarbon receptor (AhR) [21]. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor of the Per-Arnt-Sim (PAS) superfamily of environmental sensors, playing a significant role in cell proliferation, differentiation, and organ homeostasis. Direct activation of AhR can induce the production of CYP1A1 and CYP1A2, thereby increasing the concentration of 5-hydroxy emodin in the body, leading to hepatocyte damage. This suggests that emodin may act as an activator of CYP1A1 and CYP1A2. However, Tang Yun's research group found that emodin can inhibit CYP1A2 [53]. This contradictory conclusion might be attributed to emodin dosage or other factors, requiring further investigation. Furthermore, drugs that interact with emodin and exacerbate liver damage often affect the expression of hepatic metabolic enzymes, leading to abnormalities. For instance, Li et al. found that when hydroxychloroquine is used in combination with emodin, it further exacerbates emodin-induced liver damage by inhibiting UGTs and MRP2 [64,65]. Resveratrol, on the other hand, can increase the accumulation of emodin in hepatocytes by inhibiting CYP2C19 and CYP3A4, thus enhancing its hepatotoxicity. These studies suggest that emodin may induce hepatocyte damage by affecting the level or activity of hepatic metabolic enzymes, thereby influencing the metabolic function of hepatocytes.

2.4. Emodin induces autophagy in hepatocyte

Autophagy is a cellular process of self-degradation and recycling, aimed at degrading and clearing damaged or aging organelles, proteins, and other components within the cell, to maintain cellular homeostasis and biological balance. The regulation of hepatocyte autophagy involves multiple signaling pathways and key proteins, with the most important being the mammalian target of the rapamycin (mTOR) pathway and adenosine monophosphate-activated protein kinase (AMPK). The mTOR pathway acts as a negative regulator, being suppressed under low-energy conditions to promote autophagy initiation, while AMPK activation stimulates the autophagic process. Dysregulation of autophagy regulation can lead to either excessive activation or inhibition of autophagy, thereby significantly impacting the structure and function of the liver.

Previous studies have indicated that emodin induces autophagy in L02 cells by inhibiting the PI3K/AKT/mTOR signaling pathway [66]. Zhang et al. also confirmed in their study that emodin significantly activates AMPK protein [16]. Additionally, Ni et al. found that emodin markedly upregulates autophagy-related protein 7 (ATG7) [15]. Research on emodin's induction of cellular autophagy is limited, and more in-depth investigations are anticipated in the future.

3. Conclusion

As a monomeric component widely found in traditional Chinese medicinal herbs and classic herbal formulations, the potential hepatotoxic risk of emodin cannot be overlooked. This article comprehensively reviews the mechanisms through which emodin may induce hepatocyte damage, including apoptosis, cell cycle, stress, DNA damage, bile stasis, disruption of fatty acid metabolism, abnormal expression of metabolic enzymes, and autophagy. Current research mainly focuses on apoptosis, the cell cycle, and oxidative stress. In comparison, studies on DNA damage, bile stasis, disruption of fatty acid metabolism, ion channel imbalance, abnormal expression of metabolic enzymes, and autophagy are scarce. Additionally, the existing research primarily centers on cellular and organ tissue levels, definitive toxic targets and damage mechanisms have not yet been identified. Future research should delve deeper into the molecular mechanisms by which emodin triggers liver toxicity, providing concrete evidence for understanding toxicity-induced damage and guiding safe clinical drug usage.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Data availability

Data will be made available on request.

CRediT authorship contribution statement

Yupeng Wang: Writing – original draft, Visualization, Supervision. Mengchao Zhao: Visualization, Supervision, Investigation. Bo Li: Writing – review & editing. Xingchao Geng: Writing – review & editing, Funding acquisition, Conceptualization.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

The authors would like to express their sincere gratitude to the reviewers for their helpful comments on this manuscript and to Yikun Zhang, M.S., for his assistance in revising the manuscript.

References

  • 1.Dong X., et al. Emodin: a review of its Pharmacology, toxicity and Pharmacokinetics. Phytother Res. : Ptr. 2016;30:1207–1218. doi: 10.1002/ptr.5631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Fu C., et al. Dahuang Zhechong pill attenuates hepatic sinusoidal capillarization in liver cirrhosis and hepatocellular carcinoma rat model via the MK/integrin signaling pathway. J. Ethnopharmacol. 2023;308 doi: 10.1016/j.jep.2023.116191. [DOI] [PubMed] [Google Scholar]
  • 3.Yang H., et al. Emodin suppresses oxaliplatin-induced neuropathic pain by inhibiting COX2/NF-κB mediated spinal inflammation. J. Biochem. Mol. Toxicol. 2023;37 doi: 10.1002/jbt.23229. [DOI] [PubMed] [Google Scholar]
  • 4.Guo Y., et al. Emodin and rhapontigenin inhibit the replication of African swine fever virus by interfering with virus entry. Vet. Microbiol. 2023;284 doi: 10.1016/j.vetmic.2023.109794. [DOI] [PubMed] [Google Scholar]
  • 5.Liu Y., et al. Emodin protects against homocysteine-induced cardiac dysfunction by inhibiting oxidative stress via MAPK and Akt/eNOS/NO signaling pathways. Eur. J. Pharmacol. 2023;940 doi: 10.1016/j.ejphar.2022.175452. [DOI] [PubMed] [Google Scholar]
  • 6.Kang L., et al. Hepatotoxicity of the major anthraquinones derived from Polygoni multiflori radix based on bile acid homeostasis. Front. Pharmacol. 2022;13 doi: 10.3389/fphar.2022.878817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zhang G., et al. Polygoni multiflori radix exacerbates idiosyncratic inflammatory liver injury through the FXR-SHP pathway and altered pharmacokinetic behavior. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie. 2023;160 doi: 10.1016/j.biopha.2023.114233. [DOI] [PubMed] [Google Scholar]
  • 8.Teka T., et al. Polygonum multiflorum: recent updates on newly isolated compounds, potential hepatotoxic compounds and their mechanisms. J. Ethnopharmacol. 2021;271 doi: 10.1016/j.jep.2021.113864. [DOI] [PubMed] [Google Scholar]
  • 9.Rao T., Liu Y., Zeng X., Li C., Ou-Yang D. The hepatotoxicity of Polygonum multiflorum: the emerging role of the immune-mediated liver injury. Acta Pharmacol. Sin. 2021;42:27–35. doi: 10.1038/s41401-020-0360-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wang Y., et al. Clinicopathological features of He Shou Wu-induced liver injury: this ancient anti-aging therapy is not liver-friendly. Liver Int. 2019;39:389–400. doi: 10.1111/liv.13939. [DOI] [PubMed] [Google Scholar]
  • 11.Yu J., et al. Hepatoxicity of major constituents and extractions of radix Polygoni multiflori and radix Polygoni multiflori Praeparata. J. Ethnopharmacol. 2011;137:1291–1299. doi: 10.1016/j.jep.2011.07.055. [DOI] [PubMed] [Google Scholar]
  • 12.Li C., Ma J., Zheng L., Li H., Li P. Determination of emodin in L-02 cells and cell culture media with liquid chromatography–mass spectrometry: application to a cellular toxicokinetic study. J. Pharm. Biomed. Anal. 2012;71:71–78. doi: 10.1016/j.jpba.2012.07.031. [DOI] [PubMed] [Google Scholar]
  • 13.Jiang L., et al. CYP3A activation and glutathione depletion aggravate emodin-induced liver injury. Chem. Res. Toxicol. 2018;31:1052–1060. doi: 10.1021/acs.chemrestox.8b00117. [DOI] [PubMed] [Google Scholar]
  • 14.Wu L., et al. Emodin-induced hepatotoxicity was exacerbated by probenecid through inhibiting UGTs and MRP2. Toxicol. Appl. Pharmacol. 2018;359:91–101. doi: 10.1016/j.taap.2018.09.029. [DOI] [PubMed] [Google Scholar]
  • 15.Lin L., et al. Inhibition of mitochondrial complex function-the hepatotoxicity mechanism of emodin based on quantitative proteomic analyses. Cells. 2019;8 doi: 10.3390/cells8030263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wang X., et al. Paradoxical effects of emodin on ANIT-induced intrahepatic cholestasis and herb-induced hepatotoxicity in mice. Toxicol. Sci. 2019;168:264–278. doi: 10.1093/toxsci/kfy295. [DOI] [PubMed] [Google Scholar]
  • 17.Hu Y., et al. Exploration of components and mechanisms of Polygoni Multiflori Radix-induced hepatotoxicity using siRNA-mediated CYP3A4 or UGT1A1 knockdown liver cells. J. Ethnopharmacol. 2021;270 doi: 10.1016/j.jep.2021.113845. [DOI] [PubMed] [Google Scholar]
  • 18.Zhang T., et al. Insight into the practical models for predicting the essential role of the cytochrome P450-mediated biotransformation in emodin-associated hepatotoxicity. Toxicology. 2021;462 doi: 10.1016/j.tox.2021.152930. [DOI] [PubMed] [Google Scholar]
  • 19.Chen D., et al. Construction and application of hepatocyte model based on microfluidic chip technique in evaluating emodin. Nutrients. 2022;14 doi: 10.3390/nu14132768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Li D., et al. 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucopyranoside enhances the hepatotoxicity of emodin in vitro and in vivo. Toxicol. Lett. 2022;365:74–85. doi: 10.1016/j.toxlet.2022.06.008. [DOI] [PubMed] [Google Scholar]
  • 21.Wang M., et al. Emodin-induced hepatotoxicity is enhanced by 3-methylcholanthrene through activating aryl hydrocarbon receptor and inducing CYP1A1 in vitro and in vivo. Chem. Biol. Interact. 2022;365 doi: 10.1016/j.cbi.2022.110089. [DOI] [PubMed] [Google Scholar]
  • 22.Dong X., et al. Emodin induces apoptosis in human hepatocellular carcinoma HepaRG cells via the mitochondrial caspase-dependent pathway. Oncol. Rep. 2018;40:1985–1993. doi: 10.3892/or.2018.6620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lin W., et al. Emodin induces hepatocellular carcinoma cell apoptosis through MAPK and PI3K/AKT signaling pathways in vitro and in vivo. Oncol. Rep. 2016;36:961–967. doi: 10.3892/or.2016.4861. [DOI] [PubMed] [Google Scholar]
  • 24.Cui Y., et al. Involvement of PI3K/Akt, ERK and p38 signaling pathways in emodin-mediated extrinsic and intrinsic human hepatoblastoma cell apoptosis. Food Chem. Toxicol. : An International Journal Published for the British Industrial Biological Research Association. 2016;92:26–37. doi: 10.1016/j.fct.2016.03.013. [DOI] [PubMed] [Google Scholar]
  • 25.Ni B., et al. Isoliquiritigenin attenuates emodin-induced hepatotoxicity in vivo and in vitro through Nrf2 pathway. Comparative Biochemistry and Physiology. Toxicology & Pharmacology : Cbp. 2022;261 doi: 10.1016/j.cbpc.2022.109430. [DOI] [PubMed] [Google Scholar]
  • 26.Wang Q., Wen H., Ma S., Zhang Y. Polygonum multiflorum Thunb. Induces hepatotoxicity in SD rats and hepatocyte spheroids by Disrupting the metabolism of bilirubin and bile acid. J. Ethnopharmacol. 2022;296 doi: 10.1016/j.jep.2022.115461. [DOI] [PubMed] [Google Scholar]
  • 27.Chen C., et al. Metabolic profiling of emodin drug-induced liver injury and silybin treatment in rats using ultra-performance liquid chromatography-quadrupole-time-of-flight-mass spectrometry: a metabolomic and mechanistic approach. Biomed. Chromatogr. : Bmc. 2022;36:e5469. doi: 10.1002/bmc.5469. [DOI] [PubMed] [Google Scholar]
  • 28.Yang X., et al. Emodin induces liver injury by inhibiting the key enzymes of FADH/NADPH transport in rat liver. Toxicol. Res. 2018;7:888–896. doi: 10.1039/c7tx00307b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhang Y., et al. Proteomics unravels emodin causes liver oxidative damage elicited by mitochondrial dysfunction. Front. Pharmacol. 2020;11:416. doi: 10.3389/fphar.2020.00416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Giacomini C., Granéli C., Hicks R., Dazzi F. The critical role of apoptosis in mesenchymal stromal cell therapeutics and implications in homeostasis and normal tissue repair. Cell. Mol. Immunol. 2023;20:570–582. doi: 10.1038/s41423-023-01018-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhang L., et al. Emodin targets mitochondrial cyclophilin D to induce apoptosis in HepG2 cells. Biomed. Pharmacother. 2017;90:222–228. doi: 10.1016/j.biopha.2017.03.046. [DOI] [PubMed] [Google Scholar]
  • 32.Jiang L., et al. CYP3A activation and glutathione depletion aggravate emodin-induced liver injury. Chem. Res. Toxicol. 2018;31:1052–1060. doi: 10.1021/acs.chemrestox.8b00117. [DOI] [PubMed] [Google Scholar]
  • 33.Cui Y., et al. Involvement of PI3K/Akt, ERK and p38 signaling pathways in emodin-mediated extrinsic and intrinsic human hepatoblastoma cell apoptosis. Food Chem. Toxicol. 2016;92:26–37. doi: 10.1016/j.fct.2016.03.013. [DOI] [PubMed] [Google Scholar]
  • 34.Liu X., Liu Y., Cheng M., Xiao H. Metabolomic responses of human hepatocytes to emodin, aristolochic acid, and triptolide: chemicals purified from traditional Chinese medicines. J. Biochem. Mol. Toxicol. 2015;29:533–543. doi: 10.1002/jbt.21724. [DOI] [PubMed] [Google Scholar]
  • 35.Trybus W., Trybus E., Król T. Emodin sensitizes cervical cancer cells to vinblastine by inducing apoptosis and mitotic death. Int. J. Mol. Sci. 2022;23 doi: 10.3390/ijms23158510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Li Y., et al. Emodin triggers DNA double-strand breaks by stabilizing topoisomerase II-DNA cleavage complexes and by inhibiting ATP hydrolysis of topoisomerase II. Toxicol. Sci. : An Official Journal of the Society of Toxicology. 2010;118:435–443. doi: 10.1093/toxsci/kfq282. [DOI] [PubMed] [Google Scholar]
  • 37.Lv L., et al. Dioscin, a natural steroid saponin, induces apoptosis and DNA damage through reactive oxygen species: a potential new drug for treatment of glioblastoma multiforme. Food Chem. Toxicol. : An International Journal Published for the British Industrial Biological Research Association. 2013;59:657–669. doi: 10.1016/j.fct.2013.07.012. [DOI] [PubMed] [Google Scholar]
  • 38.Lee H., Lin C., Yang W., Leung W., Chang S. Aloe-emodin induced DNA damage through generation of reactive oxygen species in human lung carcinoma cells. Cancer Lett. 2006;239:55–63. doi: 10.1016/j.canlet.2005.07.036. [DOI] [PubMed] [Google Scholar]
  • 39.Arora M., et al. Rapid adaptation to CDK2 inhibition exposes intrinsic cell-cycle plasticity. Cell. 2023;186:2628–2643. doi: 10.1016/j.cell.2023.05.013. [DOI] [PubMed] [Google Scholar]
  • 40.Qin B., et al. Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin. BMC Cancer. 2022;22:671. doi: 10.1186/s12885-022-09684-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chen P., et al. AZT and emodin exhibit synergistic growth-inhibitory effects on K562/ADM cells by inducing S phase cell cycle arrest and suppressing MDR1 mRNA/p-gp protein expression. Pharm. Biol. 2013;51:1586–1591. doi: 10.3109/13880209.2013.803257. [DOI] [PubMed] [Google Scholar]
  • 42.D'Autréaux B., Toledano M.B. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 2007;8:813–824. doi: 10.1038/nrm2256. [DOI] [PubMed] [Google Scholar]
  • 43.Tomek M., Akiyama T., Dass C.R. Role of Bcl-2 in tumour cell survival and implications for pharmacotherapy. J. Pharm. Pharmacol. 2012;64:1695–1702. doi: 10.1111/j.2042-7158.2012.01526.x. [DOI] [PubMed] [Google Scholar]
  • 44.Krieser R.J., Eastman A. Cleavage and nuclear translocation of the caspase 3 substrate Rho GDP-dissociation inhibitor, D4-GDI, during apoptosis. Cell Death Differ. 1999;6:412–419. doi: 10.1038/sj.cdd.4400515. [DOI] [PubMed] [Google Scholar]
  • 45.Xing M., et al. Emodin disrupts the Notch1/Nrf2/GPX4 antioxidant system and promotes renal cell ferroptosis. J. Appl. Toxicol. : Jat. 2023 doi: 10.1002/jat.4509. [DOI] [PubMed] [Google Scholar]
  • 46.Rossi M., Wen K., Caruso F., Belli S. Emodin scavenging of superoxide radical includes π-π interaction. X-ray crystal structure, hydrodynamic voltammetry and theoretical studies. Antioxidants. 2020;9 doi: 10.3390/antiox9030194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ding Z., et al. Emodin ameliorates antioxidant capacity and exerts neuroprotective effect via PKM2-mediated Nrf2 transactivation. Food Chem. Toxicol. : An International Journal Published for the British Industrial Biological Research Association. 2022;160 doi: 10.1016/j.fct.2021.112790. [DOI] [PubMed] [Google Scholar]
  • 48.Wang H., et al. A four oxidative stress gene prognostic model and integrated immunity-analysis in pancreatic adenocarcinoma. Front. Oncol. 2022;12 doi: 10.3389/fonc.2022.1015042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cheng C., Dong W. Aloe-emodin induces endoplasmic reticulum stress-dependent apoptosis in colorectal cancer cells. Med. Sci. Mon. Int. Med. J. Exp. Clin. Res. : International Medical Journal of Experimental and Clinical Research. 2018;24:6331–6339. doi: 10.12659/MSM.908400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang X., et al. Role of emodin in atherosclerosis and other cardiovascular diseases: pharmacological effects, mechanisms, and potential therapeutic target as a phytochemical. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie. 2023;161 doi: 10.1016/j.biopha.2023.114539. [DOI] [PubMed] [Google Scholar]
  • 51.Zhang Y., Yang X., Dai Y., Xiao H. Effects of emodin on lipid accumulation and inflammation in hepatocytes] Zhongguo Zhong Yao Za Zhi = Zhongguo Zhongyao Zazhi = China Journal of Chinese Materia Medica. 2019;44:2820–2826. doi: 10.19540/j.cnki.cjcmm.20190321.401. [DOI] [PubMed] [Google Scholar]
  • 52.Quan Y., et al. Aloe emodin induces hepatotoxicity by activating NF-κB inflammatory pathway and P53 apoptosis pathway in zebrafish. Toxicol. Lett. 2019;306:66–79. doi: 10.1016/j.toxlet.2019.02.007. [DOI] [PubMed] [Google Scholar]
  • 53.Wang Y., et al. Insights into the molecular mechanisms of Polygonum multiflorum Thunb-induced liver injury: a computational systems toxicology approach. Acta Pharmacol. Sin. 2017;38:719–732. doi: 10.1038/aps.2016.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Malhi H., Guicciardi M.E., Gores G.J. Hepatocyte death: a clear and present danger. Physiol. Rev. 2010;90:1165–1194. doi: 10.1152/physrev.00061.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Faubion W.A., et al. Toxic bile salts induce rodent hepatocyte apoptosis via direct activation of Fas. J. Clin. Investig. 1999;103:137–145. doi: 10.1172/JCI4765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Woolbright B.L., et al. Bile acid-induced necrosis in primary human hepatocytes and in patients with obstructive cholestasis. Toxicol. Appl. Pharmacol. 2015;283:168–177. doi: 10.1016/j.taap.2015.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zhang Y., et al. Effect of bile duct ligation on bile acid composition in mouse serum and liver. Liver Int. : Official Journal of the International Association for the Study of the Liver. 2012;32:58–69. doi: 10.1111/j.1478-3231.2011.02662.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Allen K., Jaeschke H., Copple B.L. Bile acids induce inflammatory genes in hepatocytes: a novel mechanism of inflammation during obstructive cholestasis. Am. J. Pathol. 2011;178:175–186. doi: 10.1016/j.ajpath.2010.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang S., et al. FOXA2 prevents hyperbilirubinaemia in acute liver failure by maintaining apical MRP2 expression. Gut. 2023;72:549–559. doi: 10.1136/gutjnl-2022-326987. [DOI] [PubMed] [Google Scholar]
  • 60.Kamisako T., et al. Transport of monoglucuronosyl and bisglucuronosyl bilirubin by recombinant human and rat multidrug resistance protein 2. Hepatology. 1999;30:485–490. doi: 10.1002/hep.510300220. [DOI] [PubMed] [Google Scholar]
  • 61.Dai Y., et al. Polygoni Multiflori Radix interferes with bile acid metabolism homeostasis by inhibiting Fxr transcription, leading to cholestasis. Front. Pharmacol. 2023;14 doi: 10.3389/fphar.2023.1099935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wu L., et al. Gender differences in the hepatotoxicity and toxicokinetics of emodin: the potential mechanisms mediated by UGT2B7 and MRP2. Mol. Pharm. 2018;15:3931–3945. doi: 10.1021/acs.molpharmaceut.8b00387. [DOI] [PubMed] [Google Scholar]
  • 63.Zhou L., et al. Comprehensive investigation on the metabolism of emodin both in vivo and in vitro. J. Pharm. Biomed. Anal. 2023;223 doi: 10.1016/j.jpba.2022.115122. [DOI] [PubMed] [Google Scholar]
  • 64.Wu L., et al. Emodin-induced hepatotoxicity was exacerbated by probenecid through inhibiting UGTs and MRP2. Toxicol. Appl. Pharmacol. 2018;359:91–101. doi: 10.1016/j.taap.2018.09.029. [DOI] [PubMed] [Google Scholar]
  • 65.Wang X., et al. 2,3,5,4′-Tetrahydroxy stilbene-2-Ο-β-D-glucoside, a mechanism-based inactivator of CYP2C19 and CYP3A4, potentiates hepatic protein adduction and hepatotoxicity induced by emodin in vivo. Chem. Biol. Interact. 2022;368 doi: 10.1016/j.cbi.2022.110234. [DOI] [PubMed] [Google Scholar]
  • 66.Zheng X., et al. Emodin-induced autophagy against cell apoptosis through the PI3K/AKT/mTOR pathway in human hepatocytes. Drug Des. Dev. Ther. 2019;13:3171–3180. doi: 10.2147/DDDT.S204958. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data will be made available on request.


Articles from Heliyon are provided here courtesy of Elsevier

RESOURCES