825
views
0
recommends
+1 Recommend
1 collections
    2
    shares

      Acta Materia Medica now indexed by SCOPUS from May 2024. Interested in becoming an AMM published author?

      • Platinum Open Access with no APCs.
      • Fast peer review/Fast publication online after article acceptance.

      Check out the call for papers on our website https://amm-journal.org/index.php/2023/04/26/acta-materia-medica-call-for-papers-2/

      scite_
      0
      0
      0
      0
      Smart Citations
      0
      0
      0
      0
      Citing PublicationsSupportingMentioningContrasting
      View Citations

      See how this article has been cited at scite.ai

      scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.

       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Hepatotoxic effects of aristolochic acid: mechanisms and implications

      Published
      review-article
      Bookmark

            Abstract

            Herbal plants that contain aristolochic acids (AAs) have been widely used for medicinal purposes for centuries. However, human exposure to AAs via herbal or dietary intake is thought to be a causative factor for aristolochic acid nephropathy (AAN), hepatotoxic effects, and carcinomas. At present, the molecular mechanisms underlying AA-induced hepatotoxicity and carcinogenesis and the corresponding detoxification strategies are unclear. This review summarizes the exposure, absorption, distribution, metabolism, and excretion (ADME) process of AAs. Importantly, to more objectively determine the emerging correlation between AAs and liver cancer, this review summarizes the possible direct and indirect connections between AAs and liver cancer. In brief, this review comprehensively summarizes and analyzes the molecular mechanisms underlying AA-induced hepatotoxicity and carcinogenesis, as well as an assessment of current detoxification strategies. At the same time, a new view on the prevention and detoxification of AA-induced hepatotoxicity is proposed. Chinese medicines that contain AAs might induce liver cancer but this is a controversial notion. This review summarizes relevant views from the past and provides novel insight into AA-induced liver injury or cancer to lay the foundation for AA detoxification.

            Main article text

            1. INTRODUCTION

            Aristolochic acids (AAs) are a mixture of structurally related nitrophenanthrene carboxylic acids that can be extracted from the genus Aristolochia [1, 2] ( Figure 1 ). AAs are composed of two major components, aristolochic acid I (AAI) and aristolochic acid II (AAII), which are also known as 8-methoxy-6-nitro-phenanthro-[3,4-d]-1,3-dioxolo-5-carboxylic acid and 6-nitro-phenanthro-[3,4-d]-1,3-dioxolo-5-carboxylic acid, respectively ( Figure 1B ). The two compounds are structurally different only in the presence or absence of the O-methoxy group at the 8-position [3, 4]. Aristolactams are mainly found in the Aristolochiaceae plants together with the AAs [4, 5]. Aristolactam I (AL-I) and aristolactam II (AL-II) are two main aristolactams that are derivatives or metabolites of AAI and AAII, respectively ( Figure 1 ) [57].

            Next follows the figure caption
            Figure 1 |

            Herbal medicines or foods that contain AAs induce renal damage and liver injury.

            (A) Aristolochia plants and drugs or foods that contain AAs; and (B) chemical structures of aristolochic acid I (AAI) and II (AAII), aristolactam I (AL-I) and II (AL-II); (C) AA-induced renal damage and liver injury.

            Dozens of herbal medicines and relevant remedies that contain AAs have been used in the clinical practice of traditional Chinese (TCM), complementary, and alternative medicine to treat various diseases, including snake bites, upper respiratory tract infections, dysmenorrhea, hepatitis, pneumonia, urinary tract infections, eczema, hypertension, heart failure, and arthritis [2, 811]. Vanherweghem et al. [12, 13] first reported in the 1990s that young women in Belgium who ingested weight loss pills that contain A. fangchi ( Figure 1A ) subsequently had rapidly progressive tubulointerstitial nephritis, ultimately developing renal failure in a phenomenon which they termed Chinese herb nephropathy (CHN). Balkan endemic nephropathy (BEN), which has similar clinical and morphologic etiology to CHN, has been linked to A. clematitis exposure in Europe [14, 15]. Notably, A. clematitis contains high amounts of AAs [14, 15]. Indeed, AA-DNA adducts have been detected in the kidneys of patients with CHN and BEN, suggesting that AAs are the principal causative agent of CHN and BEN [16]. Therefore, this nephropathy has been termed aristolochic acid nephropathy (AAN) [1719]. It has also been shown that AAN patients are at increased risk for developing bladder cancer (BC) and upper tract urothelial cancer (UTUC) [2023], raising widespread concern globally.

            The International Center for Cancer Research (IARC) has classified AAI as a human carcinogen (group I) due to the underlying carcinogenic, mutagenic, and genotoxic mechanisms [2427]. Subsequently, use of herbal drugs or preparations that contain AAs has been banned in multiple countries and regions, including Japan, Canada, the United Kingdom, the United States, Australia, and Europe [9, 28, 29]. Moreover, the Chinese government issued a notice for standards of medicinal materials associated with A. manshuriensis, A. fangchi, and Radix aristolochiae. Some AA-containing herbal drugs were officially prohibited or withdrawn from the market in Taiwan and Hong Kong in 2004 [20]. However, there are still some geographic regions, such as China and the Balkans, where AA-containing herbal remedies are used or where individuals are exposed to AAs in drinking groundwater or contaminated food [19, 21, 3032].

            The international community has acknowledged that AAs can cause nephrotoxicity and urinary tract tumors over the past few decades. However, recent studies have reported that AAs are also associated with liver cancer pathogenesis ( Figure 1C ). On 18 October 2017 an article was published claiming that AAs are widely associated with the occurrence of liver cancer in Taiwan and throughout Asia [33] generating significant concern and controversy. Zeguang et al. [34] confirmed that AAI exposure could enhance the occurrence of liver cancer in CCl4-treated or PTEN-deficient mice. Epidemiologic studies subsequently showed that AA-containing herbal medicines may be an important risk factor for hepatitis B or C virus infection-associated hepatocellular carcinoma (HCC). For example, herbal medicines containing AA are associated with the risk of hepatocellular carcinoma in patients with hepatitis B virus infection [35, 36]. However, whether AAs induce liver cancer is controversial. To more rationally and objectively determine the correlation between AAs and liver cancer, this review presents the absorption, distribution, metabolism, and excretion (ADME) process of AAs with a particular focus on the liver and kidney. We also comprehensively summarize and analyze recent studies on the association between AAs and liver cancer to provide a theoretical basis for future basic and epidemiologic research on AAs as well as the potential application in clinical trials.

            2. AA EXPOSURE AND ADME

            2.1 AA exposure, absorption, and distribution

            The main known routes by which AAs are introduced into the human body are outlined in the following three ways: (1) ethnobotanical use (intentional or accidental) of herbal medicines and preparations that contain AAs [37, 38]; (2) consumption of foods that contain AAs [39]; and (3) AAs that persistently contaminate soil leading to food contamination [31, 40].

            AAs are mainly absorbed through the gastrointestinal tract and kidneys, then distributed throughout the body, including the stomach, intestines, kidneys, liver, bladder, spleen, and lungs [41]. Thus, exposure to AAs causes gastric, skin, kidney, liver, and bladder cancers in rodents [34, 42, 43]. It is worth noting that AA content in rat kidneys has been shown to be more than twice the AA content in the liver and other tissues in rats that were administered AAs orally, suggesting that the distribution of AAs in various organs is not equal [44]. This finding partially explains why the urinary system is particularly susceptible to cancer induced by AAs. In addition, the distribution and excretion of AAs and AA metabolites are affected by a variety of factors, such as mOat1, mOat2, and mOat3 proteins [3].

            2.2 AA metabolism

            Metabolism determines the bioavailability or effective concentration of AAs in individuals. There are two major pathways of AA metabolism: (1) bioactivation of AAs leads to exacerbated toxicity; and (2) detoxification of AAs reduces toxicity ( Figure 2 ). The liver is one of the main organs for metabolizing carcinogens and xenobiotics, including AAs [45]. Several endogenous cytosolic nitroreductases and microsomal enzymes contribute to AA metabolism in the liver and kidneys [46], which include cytosolic nicotinamide adenine dinucleotide phosphate hydrate (NADPH), quinone oxidoreductase 1 (NQO1), microsomal cytochrome P450 (CYP [predominantly CYP1A1 and CYP1A2]), P450 oxidoreductase (POR), and cyclooxygenase (COX) [6, 47]. Regulation of the AA biotransformation pathways in the liver and kidneys is of vital importance for the prevention and treatment of AAN and AA-related diseases.

            Next follows the figure caption
            Figure 2 |

            AAs have two different metabolic pathways (bioactivation and detoxification) that are catalyzed by metabolic enzymes in vivo.

            (1) AAs are catalyzed by metabolic enzymes and converted into aristolactam ions which attack DNA, induce mutations, and cause liver and kidney damage. (2) AAs are catalyzed by metabolic enzymes and bioconverted into some less toxic metabolites (AAIa and AIacIa), thereby reducing the side effects caused by AAs. Aristolochic acid I; AAII, aristolochic acid II; NQO1, NAD(P)H:quinone oxidoreductase; CYP1A1/2, cytochrome P450 1A1 and 1A2; POR, P450 oxidoreductase; COX, cyclooxygenase; UGT, UDP glucuronosyltransferase; SULTs, sulfotransferases; dA-AA 7-(deoxyadenosin-N6-yl) aristolactam; AAN, aristolochic acid nephropathy; UTUC, upper tract urothelial cancer; BEN, Balkan endemic nephropathy; HCC, hepatocellular carcinoma.

            2.2.1 AA bioactivation exacerbates toxicity

            The main reason AAs are toxic is bioactivation. This process can be summarized in the following key steps:

            ① Nitroreduction of AAI or AAII (compound a) to the corresponding N-hydroxyaristolactam (compound b), which is the main bioactivation pathway responsible for AA genotoxicity.

            ② AL-NOHs form an electrophilic cyclic aristolactam-nitrenium ion with a delocalized positive charge (compound c).

            ③ Compound c is the ultimate reactive species that covalently bind to the exocyclic amino groups of purine nucleotides in DNA to generate adducts [21, 4850]. These adducts consist of 7-(deoxyadenosin-N6-yl)-aristolactam I or (dA-AAI or dA-AAII) [compound d] and 7-(deoxyguanosin-N2-yl)-aristolactam I or II (dG-AAI or dG-AAII) [51, 52].

            ④ AA-DNA adducts can become enriched in the TP53 gene, which in turn leads to higher mutagenicity [53].

            ⑤ Notably, apart from inducing the formation of DNA adducts to induce malignant damage, AA metabolism can also affect gene transcription to promote carcinogenesis. Data analysis predicts the function of Akt3, FGFR3, PSEN1, and VEGFA genes in the process of tumor progression on AA exposure [54, 55].

            ⑥ Ultimately, various pathologic factors can act in synergy with AAs to cause tumor occurrence [56].

            Several cytosolic and microsomal enzymes capable of catalyzing the nitroreduction of AAI or AAII have been identified in Step ① ( Figure 2 ). Among the enzymes, NQO1 is the main cytosolic reductase that catalyzes the formation of AA-DNA adducts in vivo and in vitro [46, 5760]. Cytosolic reductases have a vital catalytic role in the reductive activation of AAs in the liver and kidneys. Additionally, microsomal enzymes, including cytochrome P450 (CYP) 1A1 and 1A2, P450 oxidoreductase (POR), and cyclooxygenase (COX), have the potential to activate metabolic AAs via nitroreduction in hepatic or renal microsomes [7, 47, 59, 61, 62]. CYP1A2 contributes to the activation of AAs, while CYP1A1 has a minor role in this process [51, 63, 64]. CYP1A1 is highly expressed in renal microsomes and can be induced by AAs. CYP1A1 has an important role in AA activation within the kidney [65]. POR only has a minor role in the activation of AA through simple nitroreduction and COX is another enzyme capable of activating AAI [51, 61]. Overall, although many enzymes catalyzing the metabolic activation of AAs in vivo or in vitro have been identified, whether these enzymes are involved in AA-related mutagenesis in humans remains to be determined.

            AA-DNA adducts have exceptionally long-term persistence in renal tissues of AAN patients, as shown in Step ③ [66]. The mutagenicity of dA-AA adducts is more significant than dG-AA adducts [67]. Moreover, levels of AAI-derived DNA adducts are significantly higher than AAII-derived DNA adducts in vivo and in vitro [41, 68].

            2.2.2 AA detoxification reduces toxicity

            In contrast to the reductive activation of AAI, oxidative reaction of AAs is an important detoxification process by which AA-induced mutagenic and carcinogenic effects are reduced ( Figure 2 ). This process can also be summarized in the following key steps.

            ① AAI is detoxified to the less toxic aristolochic acid Ia (AAIa [compound e]) through O-demethylation, resulting in a decrease of the AAI concentration to attenuate the nephrotoxicity and genotoxicity of AAs [63, 69]. AAIa or AAIa conjugated metabolites, including O-sulfate, O-acetate, and O-glucuronide esters, are considered to be the main detoxification metabolites [1] because these metabolites are excreted.

            ② AAIa (compound e) can also be reduced to N-hydroxyaristolactam Ia.

            ③ N-hydroxyaristolactam Ia further forms aristolactam Ia (AlacIa [compound f]) followed by formation of conjugated metabolites by UDP glucuronosyltransferase (UGT) and sulfotransferases (SULTs), such as N-glucuronides and O-glucuronides [6, 70]. AlacIa might be generated from demethylation of aristolactam I [71].

            ④ N-hydroxyaristolactam I can also be rearranged to 7-hydroxyaristolactam I (compound g) or further generate AL-I, which is also considered to be a detoxification pathway [6, 46, 51].

            CYP1A1/2 has a predominant role in suppressing the nephrotoxic, mutagenic, and carcinogenic effects of AAI [49, 62, 72]. In addition to CYP1A1/2, the tissue oxygen level is also a crucial factor that affects the balance between metabolic activation and detoxification of AAI. Under anaerobic conditions CYP1A1/2 directly binds to AAI and promotes AAI oxidation. Indeed, AAI may be equivalent to a ligand under anerobic conditions [51, 59, 65]. In contrast, under aerobic conditions CYP enzymes are capable of catalyzing O-demethylate AAI to non-toxic AAIa. Furthermore, AAI may be acting as a classical substrate of human CYP1A1/2 under aerobic conditions [7, 46, 51, 7375]. Moreover, UGT and SULTs effectively perform functions in the process of AA detoxification. Therefore, differences in AA metabolism (nitroreductive activation versus oxidative detoxification) may not only lead to an individual’s susceptibility to cancer, liver, and kidney toxicity, but also act as a key determinant of cancer risk.

            2.3 AA excretion and transportation

            AAs transformed to free and conjugated metabolites (O-glucuronide, O-acetate, and O-sulfate) can be excreted via the urine and feces in rodents and humans [70, 71, 76]. In addition to the effect of metabolic enzymes, reabsorption in the proximal tubules through several organic anion transporters (OATs) is known to affect the transport and secretion of AAs [3, 46, 77, 78]. OAT1 and OAT3 are mainly expressed in the kidney, while OAT2 is predominantly expressed in the liver [79]. OAT1, OAT2, and OAT3 are mainly located at the basolateral membrane of proximal tubular epithelial cells (PTECs) and regulate the transport of basic substances from the blood to the cytoplasm [8082]. OAT4, an asymmetric organic anion exchanger, is located at the apical membrane of PTECs and expressed in human kidneys. OAT4 has a key role in regulating anion reabsorption and excretion [78]. Because AAs have anionic properties and can bind to albumin, the OAT family is considered to be a crucial factor in AA-mediated toxicity by regulating AA transport and secretion [3, 77, 78, 81]. Several studies have reported that OATs in the basolateral membrane of PTECs (especially OAT1 and OAT3) contribute to the uptake of AAI through kidney cells, thus partially contributing to AAI-induced nephrotoxicity [78, 80, 83, 84]. Recently, Chang and colleagues [85] demonstrated that the toxic metabolite of AAI (sulfate-conjugated ALI [AL-I-NOSO3H]) can be transported out of the liver through multidrug resistance-associated protein (MRP) transporters, including MRP3 and MRP4, then reabsorbed into the kidney via OATs, which enhances the nephrotoxicity of AAs using the organs-on-chips technique. Although the transport and excretion of AAs has been studied to some extent, the underlying mechanism is unclear and warrants further exploration.

            3. AA-INDUCED HEPATOTOXICITY AND LIVER CANCER

            3.1 AA-induced gene mutation is a major cause of liver cancer

            Notably, AAs can cause nephrotoxicity and UTUC. The AA-induced UTUC mutational signature is characterized by A:T to T:A transversion, which is located primarily on non-transcribed strands as determined by whole-genome and exome analysis and epidemiologic statistics [1, 24, 8688]. However, it is controversial whether the mutational fingerprint of AAs is a causative factor for the pathogenesis of liver cancer induced by AAs.

            In 2013 researchers identified 11 AA-like mutational signatures from 93 HCC patients through whole-genome and exome analysis, suggesting that AAs may be partially responsible for the occurrence of HCC [86]. Ng et al. [33] recently reported that AAs are closely associated with liver cancer in Taiwan and throughout Asia. Specifically, the A:T to T:A mutational signature (Catalogue Of Somatic Mutations In Cancer [COSMIC] signature 22) detected rapidly by exon sequencing was used as a unique genetic fingerprint for AA exposure. The results showed that 78% (76/98) of patients had the specific mutational signature of AAs. To systematically assess the potential effects of AAs on HCC, Ng et al. [33] then searched for the AAs mutational fingerprint in 1400 HCCs from diverse geographic regions, finding that 47% (42/89) of HCCs from mainland China had the signature. Based on these results, Ng et al. [33] concluded that AAs were associated with the occurrence of liver cancer in Taiwan and throughout Asia, where residents have used more AA-containing herbal drugs than in other geographic regions [89]. At the same time, Ng et al. [33] showed that AAs were responsible for the A:T>T:A mutational signature in the genome and were a causal factor for the occurrence of liver cancer. This type of mutational signature can also be induced by other environmental xenobiotics and carcinogens that have potential correlations with liver tumor occurrence [90, 91] ( Table 1 ). The mutation pattern in HCC induced by DEN and AAI in mice is also known as a T>A mutation [99] ( Figure 3 ). Therefore, an AA-induced T>A transversion is a causative factor for the pathogenesis of liver cancer and this mutation can directly induce liver cancer.

            Table 1 |

            Carcinogens causing the AA-mutational signatures.

            CarcinogensModelType of diseaseMutational geneMutational signatureReference
            AAsHuman, rat, mouseLiver, bladder, UTUC, stomachTP53
            Hras
            T>A, C>T[34, 9294]
            VCHumanLiverTP53, Ki-rasG>A, T>A[95, 96]
            4-ABPHuman, mouseLiver, urinary, bladderHrasG>T, G>A,
            A>C, T>A
            [97, 98]
            DENMouseLiverHras, Braf, Apc, EgfrT>C, C>T, T>A, T>G[99]
            DMBAHuman, mouseSkinHras, Kras, Rras2, Trp53T>A, G>T[100]
            1,3-butadieneHuman, rat, mouseBone marrowHPRTT>A[95]
            Chlorambucil and melphalanHumanBreast, ovarian, marrowTP53, Nras, HprtT>A, A>C, T>G[101, 102]
            Chloroethylene oxideHumanStomach, marrowHrasG>A[103, 104]
            Next follows the figure caption
            Figure 3 |

            Mutation pattern in HCC induced by DEN and AAI in mice.

            The A:T > T:A transversion signature is also known as a T>A mutation. (A) Mutation patterns in DEN-induced HCC. (B) Mutation patterns in AAI-induced HCC. (C) Mutation patterns in HCC induced by a combination of AAI with carbon tetrachloride (CCl4).

            AAs not only directly induce liver cancer but also indirectly form liver cancer by inducing other liver diseases ( Figure 4 ). First, AAI has been reported to induce hepatic premalignant alterations (HPAs) with elevated expression of c-Myc and Lin28B in canine livers [105]. Li et al. [106] demonstrated the characteristics of HPAs from AAI exposure in mouse embryonic stem (ES) cell-derived liver-like tissues. Second, the T>A transversion was first demonstrated in HBV-infected patients with HCC from China by whole-exome sequencing analysis [106]. Research showed that among a total of 802,642 HBV-infected patients that reported consuming herbal medicines up to 1 y before being diagnosed with liver cancer, 59.4% had recorded exposure to AA-containing herbal medicines. Li et al. [106] also found a significant linear dose-response relationship between estimated AA consumption and HCC pathogenesis in HBV-infected patients, suggesting that increased exposure to AAs among HBV-positive individuals carries an elevated risk of HCC. Moreover, Chen et al. [35] reported a higher incidence of HCC in HBV-infected patients who used AA-containing herbal remedies.

            Next follows the figure caption
            Figure 4 |

            Toxic mechanism and detoxification strategies of AA-induced hepatotoxicity.

            Third, chronic infection with hepatitis C virus (HCV) is a public health problem and has been a major risk factor for HCC affecting millions of people worldwide [107, 108]. Chen and colleagues [36] recently investigated the association between AA-containing herbal remedies and primary liver cancer (PLC) among patients with HCV infection in Taiwan. Drawing from the National Health Insurance Research Database in Taiwan, 59.5% of 223,467 patients with HCV infections who had visited herbal medicine clinics up to 1 y before being diagnosed with liver cancer were shown to have using AA-containing herbal medicines. This study [36] suggested that there was a significant relationship between AA-containing herbal medicine consumption and PLC in HCV-infected patients in Taiwan. Therefore, Chinese herbal medicines containing AAs should be taken cautiously and are contraindicated in patients with HBV or HCV infections, and patients with current or past exposure to AA-containing herbal medicines should be followed closely.

            Moreover, AAs alone directly induce liver cancer in young mice and in mice with CCl4-induced liver injury. AAI alone can induce liver cancers, including HCC and combined hepatocellular and intrahepatic cholangiocarcinoma (cHCC-ICC), in a time-dose dependent manner in 2-week-old mice [34]. This finding means that AA carries a risk of liver cancer at a younger age. AAs can indirectly form liver cancer by inducing other liver diseases. Further details, including the underlying mechanisms and the situation in other parts of the world, remain to be clarified.

            3.2 Molecular mechanisms underlying AAs associated with liver cancer

            Exposure to AAs is known to be related to AAN. The molecular mechanism underlying AA-induced nephrotoxicity has been widely studied and is known to mainly involve oxidative stress, apoptosis, inflammation, and fibrosis [19, 109]. However, the specific molecular mechanism underlying AA-induced hepatotoxicity or liver cancer is largely unknown, although several specific mechanisms have been proposed and will be described below. IL6R/NF-κB signaling promotes Lin28B/let-7 changes in the liver caused by short-term AAI exposure, suggesting that the IL6R/NF-κB and c-Myc/Lin28B/let-7 signaling pathway may regulate AAI-induced acute HPAs in the canine liver [105, 110, 111]. Furthermore, the Ras/Raf signaling pathway has been reported to be significantly activated in AAI-induced liver cancer in mice according to whole-genome sequencing (WGS) and gene ontology (GO) analyses [34], suggesting that Ras/Raf signaling pathway may also be involved in AAI-induced liver cancer. Furthermore, Han et al. [34] reported that target protein genes of some important pro-carcinogenesis pathways are up-regulated, including the Ras, Notch, Wnt, Hippo, and PI3K-AKT signaling pathways, through transcriptome data analyses and western blotting assays. In contrast, AAI-induces apoptosis via inhibition of the PI3K/AKT signaling pathway [112, 113]. However, the specific role of these signaling pathways in the pathogenesis of AA-induced liver cancer remains to be determined. Overall, current evidence suggests that AAs may induce liver injury/cancer via the Ras/Raf and PI3K-AKT signaling pathways but the mechanism needs to be further clarified.

            4. AA-induced Hepatotoxicity Detoxification Strategies

            4.1 Processing of crude medicines that contain AAs

            The processing of TCM is a unique and traditional pharmaceutical technology in China, which has a direct impact on the medicinal properties and clinical efficacy. The fundamental purpose of TCM is to “take advantage and eliminate harm,” that is to ensure the safety and effectiveness of medication [114]. A variety of processing methods, such as frying and broiling, have been applied to toxic medicinal materials (aristolochia and asarum) since ancient times and have been continuously improved and are still the most commonly used methods for reducing toxicity and increasing efficiency in TCMs that contain AAs. The key process involves high temperature treatment degrading AAs and reducing the content of AAs. Because the structure of AAs contains carboxyl groups, AAs can be dissolved in water by reacting with alkali or strong alkali salts and the residual lye and aristolochlate can be removed by washing with water [115]. Vinegar burning reduces the residual and frying removes trace amounts of AA in the processed product, which are difficult to decoct; therefore, AAs are removed or difficult to fry. Li et al. [116] found that the content of five AAs in aristolochic bells, including AAI and AAII, are decreased by 16.9%–50.6% after honey-frying. Yang et al. [117] compared different processing methods of Aristolochia. The percentage of AAs in each processed product (alkali vinegar, alkali, honey burning, salt burning, ginger, fried coke, and vinegar) and the preparation method of alkali vinegar reduced the content of six AAs in Aristolocha by 50.54%. Yuan et al. [118] evaluated the toxic effects of Aristolochia and honey-fried products. The LD50 of raw and honey-fried products was 34.1 ± 7.2 and 62.6 ± 8.0 g·kg-1·d-1, respectively, and the LD50 of honey-fried products was approximately 1.84 times that of raw products.

            4.2 Compatibility with other drugs

            In view of the mechanism underlying AA toxicity, AAs can be combined with TCM to reduce toxicity. Studies have confirmed that licorice, Hops astragalus, Angelica, Salvia, Ophiopogon vulgaris, peony bark, bamboo leaves, Coptis chinensis, rhubarb, ginger, Cordyceps, aconite, Rehmannia, and Scrophularia radix are compatible with TCMs that contain AAs for two reasons: ① Chemical reactions, such as oxidation, reduction, and decomposition, occur during the decoction process, which reduces the content of AA. ② The combination of alkaloids and metal ions in TCM can inhibit the dissolution of toxic components, alleviate adverse reactions, such as nephrotoxicity and hepatotoxicity, and play a role in enhancing drug efficacy [119, 120]. Ruan et al. [121] used RP-HPLC to compare the AAI content in the Angelica sinensis group and guanmutong, and found that the AAI content in the entire formula of A. sinensis decoction was the lowest (approximately 31.97% of the guanmutong unilateral group). Wang et al. [122] showed that AA can self-assemble with berberine to form a stable supramolecular structure during water decoction and toxicologic experiments in zebrafish and mice showed that the self-assembled supramolecule formed by berberine and AA significantly reduced the toxicity of AA and alleviated the acute kidney injury caused by AA. These findings all indicate that AA toxicity can be reduced by the use of compatibility.

            4.3 Molecular breeding

            Understanding the synthesis pathways and key enzymes of AA in plants, and molecular breeding for silencing and knocking out key enzyme genes is a worthwhile and feasible method. Yang et al. [123] amplified the full-length cDNA sequence of the Aristolochia tyrosine decarboxylase (TyrDC) gene. Tyrosine decarboxylase in different plants has common sequence similarity and TyrDC is the first enzyme-encoded gene in the AA biosynthesis pathway. Nevertheless, there is a lack of in-depth exploration in this type of research.

            4.4 Detoxification from the current molecular mechanism

            In addition, various techniques, such as structural modification [105], microbial transformation [106], molecular imprinting (MIT) [107], and the use of antioxidants [108], have been used for attenuation of AAs and have achieved important results and progress, but these methods have not been widely used due to the lack of in-depth research. Antioxidant therapy is considered to be a major detoxification approach, focusing on neutralizing free radicals and reducing oxidative stress to protect the liver from AA-induced damage. Vitamin E, being lipid-soluble, protects cell membranes from oxidative damage, while vitamin C neutralizes free radicals in aqueous environments, thus lowering cellular oxidative stress associated with AAs [124, 125]. Moreover, N-acetylcysteine (NAC), a critical detoxifying agent, has shown promise in alleviating AA-induced oxidative stress and liver damage [126]. Novel detoxification agents include innovations, such as nanomaterials and targeted drug delivery systems. Nanoparticles can be designed to deliver drugs directly to the liver, thereby improving detoxification efficiency and minimizing systemic side effects [127].

            4.5 Awareness and precautions for AA toxicity

            AAs can induce renal toxicity, carcinogenicity, and mutagenicity but the impact on human health has not been fully recognized. The clinical impact of exposure to AAs has been a global public health issue with consequences ranging from AAN to liver injury in many regions and countries around the world ( Figure 5 ). Recent studies regarding the association between AAs and liver cancer have allowed us to further understand the clinical impact of AAs and provided us with some new insight for preventing AA-induced diseases, especially HCC. First, mice with CCl4-induced liver injury are more susceptible to developing liver cancer after AA exposure. AA exposure among HBV- or HCV-infected patients likewise increases the risk of acquiring HCC [3436]. This finding indicates that patients with some basic liver diseases may increase the risk of developing liver cancer by taking herbal remedies that contain AAs and these patients should be cautious about using Chinese herbal medicines that contain AAs. Second, AAI alone is known to induce the occurrence of liver cancer in 2-week old mice [34]. It has been postulated that herbal remedies that contain AAs may pose a greater risk for the occurrence of HCC in infants or young patients [128], though such effects need to be further evaluated. Third, the majority of patients with AA-related mutation characteristics are similar to patients with chronic viral hepatitis [33]. Thus, it would be interesting to clarify whether AAs are an independent liver carcinogen risk factor when conducting epidemiologic studies in the future. Finally, Chinese herbal medicines that contain AAs are still widely used in some countries and regions, where there may be a lack of comprehensive epidemiologic studies on the toxicity and carcinogenic effects of herbal medicines that contain AAs with respect to HCC and urinary system tumors. Furthermore, a large-scale epidemiologic study needs to be conducted for Chinese patients with HBV infections who are using or have used herbal drugs that contain AAs to clarify the correlation between AAs and the occurrence of HCC.

            Next follows the figure caption
            Figure 5 |

            The clinical impact of exposure to AAs from AAN to HCC is a global public health issue.

            (1) AAs are associated with important events of kidney damage (Left). (2) AAs cause liver injury. AAs, aristolochic acids; CHN, Chinese herb nephropathy; AAN, aristolochic acid nephropathy; UTUC, upper tract urothelial cancer; HCC, hepatocellular carcinoma; HBV, hepatitis B virus; HCV, hepatitis C virus.

            Moreover, several corresponding measures and approaches should be taken to prevent AA-induced toxicity from the perspective of AA exposure and ADME process. There are several possible precautionary steps with respect to AA exposure and intake, as follows: (1) enhance education and public awareness for primary prevention of AA exposure to reduce the intake of AAs; (2) strengthen the supervision of herbal materials that contain AAs to strictly control the use of AA-containing herbal drugs; (3) reduce the synthesis of AAs in Aristolochia and Asarum plants by gene editing technology to knock out or silence the key enzymes of AA biosynthesis; and (4) remove AAs from the agricultural soil and groundwater to prevent exposure to AAs via dietary intake [31, 32]. In addition, there are also various possible methods from the perspective of the ADME process of AAs to prevent AA-induced toxicity, as follows: (I) inhibit organic anion transporters to reduce reabsorption of AAs in renal cells [84]; (2) suppress the activity and expression of enzymes related to AA metabolic activation to decrease the level of AA-DNA adducts [62, 129, 130]; and (3) increase the activity and expression of enzymes associated with AA metabolic detoxification to accelerate AA transformation into free and conjugated metabolites that can then be excreted [7].

            5. PERSPECTIVE AND CONCLUSION

            AAs are a group of compounds with potent nephrotoxicity and carcinogenicity. Human exposure to AAs via herbal or dietary intake is recognized as a crucial causative factor of AAN and UTUC. At present, the avenues of AA intake into the human body are mainly through the consumption of herbs and food that contain AAs, as well as groundwater contaminated by AAs. AA-DNA adducts have been used as specific biomarkers to assess AA exposure. In addition to exposure to AAs and the ingested dosage, AA metabolism is an important factor in determining the bioavailability and effective concentration of AAs in individuals. There are two major pathways of AA metabolism (bioactivation and detoxification). This review summarized the metabolic process of AAs in the body and the cytosolic and microsomal enzymes that affect the biological metabolism of AAs. Although several enzymes that catalyze the biotransformation of AAs have been identified, whether there are other metabolic enzymes that also affect the metabolism of AAs remains to be answered. More importantly, this review objectively summarizes and analyzes the literature published in the last several years regarding the association of AAs with liver cancer, as outlined earlier: (1) The specific mutation signature of AAs is not only induced by AAs, but also by other environmental carcinogens. (2) Exposure to AAs alone or a combination of AAs with CCl4 causes liver cancer in young mice, but there is no evidence of AAs-induced HCC in adult animals. (3) AA-containing herbal medicines may be an important risk factor for HBV- or HCV-infected patients developing HCC. Therefore, AAs may increase the risk of developing liver cancer in patients with basic liver diseases. Moreover, we put forward new insights and precautions for preventing AA-induced diseases, especially liver cancer. In summary, although several existing research results indicate that AAs may be one of the causative factors of liver cancer, it is still urgent to further confirm whether there is actually a causative relationship between AA and HCC. Importantly, the relevant molecular mechanisms underlying AA-induced liver injury need to be further explored [131133]. We also provide insight and strategies for the prevention and treatment of AA-induced liver and kidney injury, thereby reducing the burden of AA-induced liver and kidney injury. At the same time, the feasibility and applicability of the platform for analyzing the toxicity evaluation and toxicologic mechanism research of TCM components were also demonstrated.

            CONFLICTS OF INTEREST

            All authors declare no conflicts of interest.

            REFERENCES

            1. Stiborová M, Arlt VM, Schmeiser HH. Balkan Endemic Nephropathy: An Update on Its Aetiology. Archives of Toxicology. 2016. Vol. 90:2595–2615

            2. Yang HY, Chen PC, Wang JD. Chinese Herbs Containing Aristolochic Acid Associated with Renal Failure and Urothelial Carcinoma: A Review from Epidemiologic Observations to Causal Inference. BioMed Research International. 2014. Vol. 2014:569325

            3. Dickman KG, Sweet DH, Bonala R, Ray T, Wu A. Physiological and Molecular Characterization of Aristolochic Acid Transport by the Kidney. The Journal of Pharmacology and Experimental Therapeutics. 2011. Vol. 338:588–597

            4. Kumar V, Poonam, Prasad AK, Parmar VS. Naturally Occurring Aristolactams, Aristolochic Acids and Dioxoaporphines and their Biological Activities. Natural Product Reports. 2003. Vol. 20:565–583

            5. Michl J, Ingrouille MJ, Simmonds MS, Heinrich M. Naturally Occurring Aristolochic Acid Analogues and their Toxicities. Natural Product Reports. 2014. Vol. 31:676–693

            6. Stiborová M, Frei E, Arlt VM, Schmeiser HH. Metabolic Activation of Carcinogenic Aristolochic Acid, A Risk Factor for Balkan Endemic Nephropathy. Mutation Research. 2008. Vol. 658:55–67

            7. Stiborová M, Frei E, Schmeiser HH, Arlt VM, Martínek V. Mechanisms of Enzyme-Catalyzed Reduction of Two Carcinogenic Nitro-Aromatics, 3-Nitrobenzanthrone and Aristolochic Acid I: Experimental and Theoretical Approaches. International Journal of Molecular Sciences. 2014. Vol. 15:10271–10295

            8. Anandagoda N, Lord GM. Preventing Aristolochic Acid Nephropathy. Clinical Journal of the American Society of Nephrology. 2015. Vol. 10:167–168

            9. Abdullah R, Diaz LN, Wesseling S, Rietjens IM. Risk Assessment of Plant Food Supplements and Other Herbal Products Containing Aristolochic Acids Using the Margin of Exposure (MOE) Approach. Food Additives & Contaminants Part A, Chemistry, Analysis, Control, Exposure & Risk Assessment. 2017. Vol. 34:135–144

            10. Ioset JR, Raoelison GE, Hostettmann K. Detection of Aristolochic Acid in Chinese Phytomedicines and Dietary Supplements Used as Slimming Regimens. Food and Chemical Toxicology: An International Journal Published for the British Industrial Biological Research Association. 2003. Vol. 41:29–36

            11. Arlt VM, Stiborova M, Schmeiser HH. Aristolochic Acid as a Probable Human Cancer Hazard in Herbal Remedies: A Review. Mutagenesis. 2002. Vol. 17:265–77

            12. Vanherweghem JL, Depierreux M, Tielemans C, Abramowicz D, Dratwa M, Jadoul M, et al.. Rapidly Progressive Interstitial Renal Fibrosis in Young Women: Association with Slimming Regimen Including Chinese Herbs. Lancet (London, England). 1993. Vol. 341:387–391

            13. Nortier JL, Vanherweghem JL. For Patients Taking Herbal Therapy--Lessons from Aristolochic Acid Nephropathy. Nephrology Dialysis Transplantation. 2007. Vol. 22:1512–1517

            14. Jelaković B, Dika Ž, Arlt VM, Stiborova M, Pavlović NM, Nikolić J, et al.. Balkan Endemic Nephropathy and the Causative Role of Aristolochic Acid. Seminars in Nephrology. 2019. Vol. 39:284–296

            15. Grollman AP, Shibutani S, Moriya M, Miller F, Wu L, Moll U, et al.. Aristolochic Acid and the Etiology of Endemic (Balkan) Nephropathy. Proceedings of the National Academy of Sciences of the United States of America. 2007. Vol. 104:12129–12134

            16. Vanhaelen M, Vanhaelen-Fastre R, But P, Vanherweghem JL. Identification of Aristolochic Acid in Chinese Herbs. Lancet (London, England). 1994. Vol. 343:174

            17. Premužić V, Ivković V, Leko N, Stipančić Ž, Karanović S, Jelaković A, et al.. Arterial Stiffness in Balkan Endemic Nephropathy, an Environmental Form of Aristolochic Acid Nephropathy. Frontiers in Cardiovascular Medicine. 2018. Vol. 5:166

            18. But PP, Ma SC. Chinese-Herb Nephropathy. Lancet (London, England). 1999. Vol. 354:1731–1732

            19. Jadot I, Declèves AE, Nortier J, Caron N. An Integrated View of Aristolochic Acid Nephropathy: Update of the Literature. International Journal of Molecular Sciences. 2017. Vol. 18:297

            20. Zhang HM, Zhao XH, Sun ZH, Li GC, Liu GC, Sun LR, et al.. Recognition of the Toxicity of Aristolochic Acid. Journal of Clinical Pharmacy and Therapeutics. 2019. Vol. 44:157–162

            21. Stiborová M, Arlt VM, Schmeiser HH. DNA Adducts Formed by Aristolochic Acid Are Unique Biomarkers of Exposure and Explain the Initiation Phase of Upper Urothelial Cancer. International Journal of Molecular Sciences. 2017. Vol. 18:2144

            22. Nortier JL, Martinez MC, Schmeiser HH, Arlt VM, Bieler CA, Petein M, et al.. Urothelial Carcinoma Associated with the Use of a Chinese Herb (Aristolochia Fangchi). The New England Journal of Medicine. 2000. Vol. 342:1686–1692

            23. De Broe ME. Chinese Herbs Nephropathy and Balkan Endemic Nephropathy: Toward A Single Entity, Aristolochic Acid Nephropathy. Kidney International. 2012. Vol. 81:513–515

            24. Aydin S, Ambroise J, Cosyns JP, Gala JL. TP53 Mutations in p53-Negative Dysplastic Urothelial Cells from Belgian AAN Patients: New Evidence for Aristolochic Acid-Induced Molecular Pathogenesis and Carcinogenesis. Mutation Research - Genetic Toxicology and Environmental Mutagenesis. 2017. Vol. 818:17–26

            25. Imai H, Matsuoka M, Kumagai T, Sakamoto T, Koumura T. Lipid Peroxidation-Dependent Cell Death Regulated by GPx4 and Ferroptosis. Current Topics in Microbiology and Immunology. 2017. Vol. 403:143–170

            26. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Some Traditional Herbal Medicines, Some Mycotoxins, Naphthalene and Styrene. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. 2002. Vol. 82. p. 1–556

            27. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: Pharmaceuticals. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. 2012. Vol. 100(Pt A):1–401

            28. Abdullah R, Alhusainy W, Woutersen J, Rietjens IM, Punt A. Predicting Points of Departure for Risk Assessment Based on in Vitro Cytotoxicity Data and Physiologically Based Kinetic (PBK) Modeling: The Case of Kidney Toxicity Induced by Aristolochic Acid I. Food and Chemical Toxicology: An International Journal Published for the British Industrial Biological Research Association. 2016. Vol. 92:104–116

            29. Debelle FD, Vanherweghem JL, Nortier JL. Aristolochic acid Nephropathy: A Worldwide Problem. Kidney International. 2008. Vol. 74:158–169

            30. Stoyanovsky DA, Tyurina YY, Shrivastava I, Bahar I, Tyurin VA, Protchenko O, et al.. Iron Catalysis of Lipid Peroxidation in Ferroptosis: Regulated Enzymatic or Random Free Radical Reaction? Free Radical Biology & Medicine. 2019. Vol. 133:153–161

            31. Chan CK, Tung KK, Pavlović NM, Chan W. Remediation of Aristolochic Acid-Contaminated Soil by An Effective Advanced Oxidation Process. The Science of the Total Environment. 2020. Vol. 720:137528

            32. Tung KK, Chan CK, Zhao Y, Chan KJ, Liu G, Pavlović NM, et al.. Occurrence and Environmental Stability of Aristolochic Acids in Groundwater Collected from Serbia: Links to Human Exposure and Balkan Endemic Nephropathy. Environmental Science & Technology. 2020. Vol. 54:1554–1561

            33. Ng AWT, Poon SL, Huang MN, Lim JQ, Boot A, Yu W, et al.. Aristolochic Acids and their Derivatives are Widely Implicated in Liver Cancers in Taiwan and Throughout Asia. Science Translational Medicine. 2017. Vol. 9:eaan6446

            34. Lu ZN, Luo Q, Zhao LN, Shi Y, Wang N, Wang L, et al.. The Mutational Features of Aristolochic Acid-Induced Mouse and Human Liver Cancers. Hepatology (Baltimore, Md). 2020. Vol. 71:929–942

            35. Chen CJ, Yang YH, Lin MH, Lee CP, Tsan YT, Lai MN, et al.. Herbal Medicine Containing Aristolochic Acid and the Risk of Hepatocellular Carcinoma in Patients with Hepatitis B Virus Infection. International Journal of Cancer. 2018. Vol. 143:1578–1587

            36. Chen CJ, Yang YH, Lin MH, Lee CP, Tsan YT, Lai MN, et al.. Herbal Medicine Containing Aristolochic Acid and the Risk of Primary Liver Cancer in Patients with Hepatitis C Virus Infection. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, Cosponsored by the American Society of Preventive Oncology. 2019. Vol. 28:1876–1883

            37. Nitzsche D, Melzig MF, Arlt VM. Evaluation of the Cytotoxicity and Genotoxicity of Aristolochic Acid I - A Component of Aristolochiaceae Plant Extracts Used in Homeopathy. Environmental Toxicology and Pharmacology. 2013. Vol. 35:325–334

            38. Michl J, Jennings HM, Kite GC, Ingrouille MJ, Simmonds MS, Heinrich M. Is Aristolochic Acid Nephropathy A Widespread Problem in Developing Countries? A Case Study of Aristolochia Indica L. in Bangladesh Using An Ethnobotanical-Phytochemical Approach. Journal of Ethnopharmacology. 2013. Vol. 149:235–244

            39. Schmeiser HH, Kucab JE, Arlt VM, Phillips DH, Hollstein M, Gluhovschi G, et al.. Evidence of Exposure to Aristolochic Acid in Patients with Urothelial Cancer from A Balkan Endemic Nephropathy Region of Romania. Environmental and Molecular Mutagenesis. 2012. Vol. 53:636–641

            40. Pavlović NM, Maksimović V, Dragišić Maksimović J, Orem WH, Tatu CA, Lerch HE, et al.. Possible Health Impacts of Naturally Occurring Uptake of Aristolochic Acids by Maize and Cucumber Roots: Links to the Etiology of Endemic (Balkan) Nephropathy. Environmental Geochemistry and Health. 2013. Vol. 35:215–226

            41. Shibutani S, Dong H, Suzuki N, Ueda S, Miller F, Grollman AP. Selective Toxicity of Aristolochic Acids I and II. Drug Metabolism and Disposition: The Biological Fate of Chemicals. 2007. Vol. 35:1217–1222

            42. Wang L, Ding X, Li C, Zhao Y, Yu C, Yi Y, et al.. Oral Administration of Aristolochia Manshuriensis Kom in Rats Induces Tumors in Multiple Organs. Journal of Ethnopharmacology. 2018. Vol. 225:81–89

            43. Liu X, Wu J, Wang J, Feng X, Wu H, Huang R, et al.. Mitochondrial Dysfunction is Involved in Aristolochic Acid I-Induced Apoptosis in Renal Proximal Tubular Epithelial Cells. Human & Experimental Toxicology. 2020. Vol. 39:673–682

            44. National Toxicology Program. Final report on carcinogens background document for aristolochic acids. Report on Carcinogens Background Document for [Substance Name]. 2008. Vol. i-xxv:1–246

            45. Healy ME, Chow JD, Byrne FL, Breen DS, Leitinger N, Li C, et al.. Dietary Effects on Liver Tumor Burden in Mice Treated with the Hepatocellular Carcinogen Diethylnitrosamine. Journal of Hepatology. 2015. Vol. 62:599–606

            46. Stiborová M, Martínek V, Frei E, Arlt VM, Schmeiser HH. Enzymes Metabolizing Aristolochic Acid and their Contribution to the Development of Aristolochic Acid Nephropathy and Urothelial Cancer. Current Drug Metabolism. 2013. Vol. 14:695–705

            47. Milichovský J, Bárta F, Schmeiser HH, Arlt VM, Frei E, Stiborová M, et al.. Active Site Mutations as A Suitable Tool Contributing to Explain A Mechanism of Aristolochic Acid I Nitroreduction by Cytochromes P450 1A1, 1A2 and 1B1. International Journal of Molecular Sciences. 2016. Vol. 17:213

            48. Pfau W, Schmeiser HH, Wiessler M. Aristolochic Acid Binds Covalently to the Exocyclic Amino Group of Purine Nucleotides in DNA. Carcinogenesis. 1990. Vol. 11:313–319

            49. Stiborová M, Bárta F, Levová K, Hodek P, Schmeiser HH, Arlt VM, et al.. A Mechanism of O-Demethylation of Aristolochic Acid I by Cytochromes P450 and Their Contributions to This Reaction in Human and Rat Livers: Experimental and Theoretical Approaches. International Journal of Molecular Sciences. 2015. Vol. 16:27561–27575

            50. Asher G, Dym O, Tsvetkov P, Adler J, Shaul Y. The Crystal Structure of NAD(P)H Quinone Oxidoreductase 1 in Complex with Its Potent Inhibitor Dicoumarol. Biochemistry. 2006. Vol. 45:6372–6378

            51. Stiborová M, Levová K, Bárta F, Shi Z, Frei E, Schmeiser HH, et al.. Bioactivation Versus Detoxication of the Urothelial Carcinogen Aristolochic Acid I by Human Cytochrome P450 1A1 and 1A2. Toxicological Sciences: An Official Journal of the Society of Toxicology. 2012. Vol. 125:345–358

            52. Bieler CA, Stiborova M, Wiessler M, Cosyns JP, van Ypersele de Strihou C, Schmeiser HH. 32P-Post-Labelling Analysis of DNA Adducts Formed by Aristolochic Acid in Tissues from Patients with Chinese Herbs Nephropathy. Carcinogenesis. 1997. Vol. 18:1063–1067

            53. Sidorenko VS, Yeo JE, Bonala RR, Johnson F, Schärer OD, Grollman AP. Lack of Recognition by Global-Genome Nucleotide Excision Repair Accounts for the High Mutagenicity and Persistence of Aristolactam-DNA Adducts. Nucleic Acids Research. 2012. Vol. 40:2494–2505

            54. Tao L, Zeng Y, Wang J, Liu Z, Shen B, Ge J, et al.. Differential MicroRNA Expression in Aristolochic Acid-Induced Upper Urothelial Tract Cancers Ex Vivo. Molecular Medicine Reports. 2015. Vol. 12:6533–6546

            55. Chen CH, Dickman KG, Huang CY, Shun CT, Tai HC, Huang KH, et al.. Recurrence Pattern and TP53 Mutation in Upper Urinary Tract Urothelial Carcinoma. Oncotarget. 2016. Vol. 7:45225–45236

            56. Chen CH, Dickman KG, Huang CY, Moriya M, Shun CT, Tai HC, et al.. Aristolochic Acid-Induced Upper Tract Urothelial Carcinoma in Taiwan: Clinical Characteristics and Outcomes. International Journal of Cancer. 2013. Vol. 133:14–20

            57. Stiborová M, Frei E, Sopko B, Sopková K, Marková V, Lanková M, et al.. Human Cytosolic Enzymes Involved in the Metabolic Activation of Carcinogenic Aristolochic Acid: Evidence for Reductive Activation by Human NAD(P)H:Quinone Oxidoreductase. Carcinogenesis. 2003. Vol. 24:1695–1703

            58. Bárta F, Levová K, Frei E, Schmeiser HH, Arlt VM, Stiborová M. The Effect of Aristolochic Acid I on Expression of NAD(P)H:Quinone Oxidoreductase in Mice and Rats--A Comparative Study. Mutation Research Genetic Toxicology and Environmental Mutagenesis. 2014. Vol. 768:1–7

            59. Stiborová M, Sopko B, Hodek P, Frei E, Schmeiser HH, Hudecek J. The Binding of Aristolochic Acid I to the Active Site of Human Cytochromes P450 1A1 and 1A2 Explains their Potential to Reductively Activate this Human Carcinogen. Cancer Letters. 2005. Vol. 229:193–204

            60. Levova K, Moserova M, Nebert DW, Phillips DH, Frei E, Schmeiser HH, et al.. NAD(P)H:Quinone Oxidoreductase Expression in Cyp1a-Knockout and CYP1A-Humanized Mouse Lines and Its Effect on Bioactivation of the Carcinogen Aristolochic Acid I. Toxicology and Applied Pharmacology. 2012. Vol. 265:360–367

            61. Stiborová M, Frei E, Hodek P, Wiessler M, Schmeiser HH. Human Hepatic and Renal Microsomes, Cytochromes P450 1A1/2, NADPH: Cytochrome P450 Reductase and Prostaglandin H Synthase Mediate the Formation of Aristolochic Acid-DNA Adducts Found in Patients with Urothelial Cancer. International Journal of Cancer. 2005. Vol. 113:189–197

            62. Dračínská H, Bárta F, Levová K, Hudecová A, Moserová M, Schmeiser HH, et al.. Induction of Cytochromes P450 1A1 and 1A2 Suppresses Formation of DNA Adducts by Carcinogenic Aristolochic Acid I in Rats in Vivo. Toxicology. 2016. Vol. 344–346:7–18

            63. Stiborová M, Frei E, Schmeiser HH. Biotransformation Enzymes in Development of Renal Injury and Urothelial Cancer Caused by Aristolochic Acid. Kidney International. 2008. Vol. 73:1209–1211

            64. Stiborová M, Martínek V, Rýdlová H, Hodek P, Frei E. Sudan I is A Potential Carcinogen for Humans: Evidence for its Metabolic Activation and Detoxication by Human Recombinant Cytochrome P450 1A1 and Liver Microsomes. Cancer Research. 2002. Vol. 62:5678–5684

            65. Stiborová M, Frei E, Wiessler M, Schmeiser HH. Human Enzymes Involved in the Metabolic Activation of Carcinogenic Aristolochic Acids: Evidence for Reductive Activation by Cytochromes P450 1A1 and 1A2. Chemical Research in Toxicology. 2001. Vol. 14:1128–1137

            66. Schmeiser HH, Nortier JL, Singh R, Da Costa GG, Sennesael J, Cassuto-Viguier E, et al.. Exceptionally Long-Term Persistence of DNA Adducts Formed by Carcinogenic Aristolochic Acid I in Renal Tissue from Patients with Aristolochic Acid Nephropathy. International Journal of Cancer. 2014. Vol. 135:502–507

            67. Attaluri S, Bonala RR, Yang IY, Lukin MA, Wen Y, Grollman AP, et al.. DNA Adducts of Aristolochic Acid II: Total Synthesis and Site-Specific Mutagenesis Studies in Mammalian Cells. Nucleic Acids Research. 2010. Vol. 38:339–352

            68. Stiborová M, Frei E, Breuer A, Wiessler M, Schmeiser HH. Evidence for Reductive Activation of Carcinogenic Aristolochic Acids by Prostaglandin H Synthase -- (32)P-Postlabeling Analysis of DNA Adduct Formation. Mutation Research. 2001. Vol. 493:149–160

            69. Xiao Y, Ge M, Xue X, Wang C, Wang H, Wu X, et al.. Hepatic Cytochrome P450s Metabolize Aristolochic Acid and Reduce Its Kidney Toxicity. Kidney International. 2008. Vol. 73:1231–1239

            70. Chan W, Luo HB, Zheng Y, Cheng YK, Cai Z. Investigation of the Metabolism and Reductive Activation of Carcinogenic Aristolochic Acids in Rats. Drug Metabolism and Disposition. 2007. Vol. 35:866–874

            71. Schmeiser HH, Pool BL, Wiessler M. Identification and Mutagenicity of Metabolites of Aristolochic Acid Formed by Rat Liver. Carcinogenesis. 1986. Vol. 7:59–63

            72. Rosenquist TA, Einolf HJ, Dickman KG, Wang L, Smith A, Grollman AP. Cytochrome P450 1A2 Detoxicates Aristolochic Acid in the Mouse. Drug Metabolism and Disposition. 2010. Vol. 38:761–768

            73. Stiborová M, Levová K, Bárta F, Šulc M, Frei E, Arlt VM, et al.. The Influence of Dicoumarol on the Bioactivation of the Carcinogen Aristolochic Acid I in Rats. Mutagenesis. 2014. Vol. 29:189–200

            74. Stiborová M, Frei E, Arlt VM, Schmeiser HH. Knockout and Humanized Mice as Suitable Tools to Identify Enzymes Metabolizing the Human Carcinogen Aristolochic Acid. Xenobiotica. 2014. Vol. 44:135–145

            75. Arlt VM, Levová K, Bárta F, Shi Z, Evans JD, Frei E, et al.. Role of P450 1A1 and P450 1A2 in Bioactivation Versus Detoxication of the Renal Carcinogen Aristolochic Acid I: Studies in Cyp1a1−/−, Cyp1a2−/−, and Cyp1a1/1a2−/− mice. Chemical Research in Toxicology. 2011. Vol. 24:1710–1719

            76. Chan W, Cui L, Xu G, Cai Z. Study of the Phase I and Phase II Metabolism of Nephrotoxin Aristolochic Acid by Liquid Chromatography/Tandem Mass Spectrometry. Rapid Communications in Mass Spectrometry. 2006. Vol. 20:1755–1760

            77. Babu E, Takeda M, Nishida R, Noshiro-Kofuji R, Yoshida M, Ueda S, et al.. Interactions of Human Organic Anion Transporters with Aristolochic Acids. Journal of Pharmacological Sciences. 2010. Vol. 113:192–196

            78. Hagos Y, Wolff NA. Assessment of the role of Renal Organic Anion Transporters in Drug-Induced Nephrotoxicity. Toxins (Basel). 2010. Vol. 2:2055–2082

            79. Yin J, Wang J. Renal Drug Transporters and Their Significance in Drug-Drug Interactions. Acta Pharmaceutica Sinica B. 2016. Vol. 6:363–373

            80. Bakhiya N, Arlt VM, Bahn A, Burckhardt G, Phillips DH, Glatt H. Molecular Evidence for an Involvement of Organic Anion Transporters (OATs) in Aristolochic Acid Nephropathy. Toxicology. 2009. Vol. 264:74–79

            81. Shen QQ, Wang JJ, Roy D, Sun LX, Jiang ZZ, Zhang LY, et al.. Organic Anion Transporter 1 and 3 Contribute to Traditional Chinese Medicine-Induced Nephrotoxicity. Chinese Journal of Natural Medicines. 2020. Vol. 18:196–205

            82. Ahn SY, Nigam SK. Toward a Systems Level Understanding of Organic Anion and Other Multispecific Drug Transporters: A Remote Sensing and Signaling Hypothesis. Molecular Pharmaceutics. 2009. Vol. 76:481–490

            83. Xue X, Gong LK, Maeda K, Luan Y, Qi XM, Sugiyama Y, et al.. Critical Role of Organic Anion Transporters 1 and 3 in Kidney Accumulation and Toxicity of Aristolochic Acid I. Molecular Pharmaceutics. 2011. Vol. 8:2183–2192

            84. Baudoux TE, Pozdzik AA, Arlt VM, De Prez EG, Antoine MH, Quellard N, et al.. Probenecid Prevents Acute Tubular Necrosis in a Mouse Model of Aristolochic Acid Nephropathy. Kidney International. 2012. Vol. 82:1105–1113

            85. Chang SY, Weber EJ, Sidorenko VS, Chapron A, Yeung CK, Gao C, et al.. Human Liver-Kidney Model Elucidates the Mechanisms of Aristolochic Acid Nephrotoxicity. JCI Insight. 2017. Vol. 2:e95978

            86. Poon SL, Pang ST, McPherson JR, Yu W, Huang KK, Guan P, et al.. Genome-Wide Mutational Signatures of Aristolochic Acid and Its Application as a Screening Tool. Science Translational Medicine. 2013. Vol. 5:197ra01

            87. Hoang ML, Chen CH, Sidorenko VS, He J, Dickman KG, Yun BH, et al.. Mutational Signature of Aristolochic Acid Exposure as Revealed by Whole-Exome Sequencing. Science Translational Medicine. 2013. Vol. 5:197ra02

            88. Kucab JE, Phillips DH, Arlt VM. Linking Environmental Carcinogen Exposure to TP53 Mutations in Human Tumours Using the Human TP53 Knock-in (Hupki) Mouse Model. The FEBS Journal. 2010. Vol. 277:2567–2583

            89. Hsieh SC, Lin IH, Tseng WL, Lee CH, Wang JD. Prescription Profile of Potentially Aristolochic Acid Containing Chinese Herbal Products: An Analysis of National Health Insurance Data in Taiwan between 1997 and 2003. Chinese Medicine. 2008. Vol. 3:13

            90. Yu XJ, Yang MJ, Zhou B, Wang GZ, Huang YC, Wu LC, et al.. Characterization of Somatic Mutations in Air Pollution-Related Lung Cancer. EBioMedicine. 2015. Vol. 2:583–590

            91. Zhou G, Zhao X. Carcinogens that Induce the A:T > T:A Nucleotide Substitutions in the Genome. Frontiers of Medicine. 2018. Vol. 12:236–238

            92. Chen CH, Dickman KG, Moriya M, Zavadil J, Sidorenko VS, Edwards KL, et al.. Aristolochic Acid-Associated Urothelial Cancer in Taiwan. Proceedings of the National Academy of Sciences of the United States of America. 2012. Vol. 109:8241–8246

            93. Sborchia M, De Prez EG, Antoine MH, Bienfait L, Indra R, Valbuena G, et al.. The Impact of p53 on Aristolochic Acid I-Induced Nephrotoxicity and DNA Damage In Vivo and In Vitro. Archives of Toxicology. 2019. Vol. 93:3345–3366

            94. Wang L, Li C, Tian J, Liu J, Zhao Y, Yi Y, et al.. Genome-Wide Transcriptional Analysis of Aristolochia Manshuriensis Induced Gastric Carcinoma. Pharmaceutical Biology. 2020. Vol. 58:98–106

            95. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans: IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Volume 97. 1,3-Butadiene, Ethylene Oxide and Vinyl Halides (Vinyl Fluoride, Vinyl Chloride and Vinyl Bromide. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. 2008. Vol. 97:3–471

            96. Sherman M. Vinyl chloride and the liver. Journal of Hepatology. 2009. Vol. 51:1074–1081

            97. Wang S, Sugamori KS, Brenneman D, Hsu I, Calce A, Grant DM. Influence of Arylamine N-Acetyltransferase, Sex, And Age On 4-Aminobiphenyl-Induced In Vivo Mutant Frequencies And Spectra In Mouse Liver. Environmental and Molecular Mutagenesis. 2012. Vol. 53:350–357

            98. Fletcher K, Tinwell H, Ashby J. Mutagenicity of the Human Bladder Carcinogen 4-Aminobiphenyl to the Bladder of MutaMouse Transgenic Mice. Mutation Research. 1998. Vol. 400:245–250

            99. Connor F, Rayner TF, Aitken SJ, Feig C, Lukk M, Santoyo-Lopez J, et al.. Mutational Landscape of a Chemically-Induced Mouse Model of Liver Cancer. Journal of Hepatology. 2018. Vol. 69:840–850

            100. Nassar D, Latil M, Boeckx B, Lambrechts D, Blanpain C. Genomic Landscape of Carcinogen-Induced and Genetically Induced Mouse Skin Squamous Cell Carcinoma. Nature Medicine. 2015. Vol. 21:946–954

            101. Wang P, Bauer GB, Kellogg GE, Abraham DJ, Povirk LF. Effect of Distamycin On Chlorambucil-Induced Mutagenesis in pZ189: Evidence of a Role for Minor Groove Alkylation at Adenine N-3. Mutagenesis. 1994. Vol. 9:133–139

            102. Wang P, Bennett RA, Povirk LF. Melphalan-Induced Mutagenesis in an SV40-Based Shuttle Vector: Predominance of A.T–T.A Transversions. Cancer Research. 1990. Vol. 50:7527–7531

            103. Guengerich FP. Roles of the Vinyl Chloride Oxidation Products 1-Chlorooxirane and 2-Chloroacetaldehyde in the In Vitro Formation of Etheno Adducts of Nucleic Acid Bases [Corrected]. Chemical Research in Toxicology. 1992. Vol. 5:2–5

            104. Bren U, Zupan M, Guengerich FP, Mavri J. Chemical Reactivity as a Tool to Study Carcinogenicity: Reaction Between Chloroethylene Oxide and Guanine. The Journal of Organic Chemistry. 2006. Vol. 71:4078–4084

            105. Jin K, Su KK, Li T, Zhu XQ, Wang Q, Ge RS, et al.. Hepatic Premalignant Alterations Triggered by Human Nephrotoxin Aristolochic Acid I in Canines. Cancer Prevention Research (Philadelphia). 2016. Vol. 9:324–334

            106. Huang J, Deng Q, Wang Q, Li KY, Dai JH, Li N, et al.. Exome sequencing of Hepatitis B Virus-Associated Hepatocellular Carcinoma. Nature Genetics. 2012. Vol. 44:1117–1721

            107. Torres HA, Shigle TL, Hammoudi N, Link JT, Samaniego F, Kaseb A, et al.. The Oncologic Burden of Hepatitis C Virus Infection: A Clinical Perspective. CA: Cancer Journal for Clinicians. 2017. Vol. 67:411–431

            108. Spearman CW, Dusheiko GM, Hellard M, Sonderup M. Hepatitis C. Lancet. 2019. Vol. 394:1451–1466

            109. Anger EE, Yu F, Li J. Aristolochic Acid-Induced Nephrotoxicity: Molecular Mechanisms and Potential Protective Approaches. International Journal of Molecular Sciences. 2020. Vol. 21:1157

            110. Li T, Jin K, Zhu DY, Li L, Mao ZR, Wu BW, et al.. Premalignant Alteration Assessment in Liver-Like Tissue Derived from Embryonic Stem Cells by Aristolochic Acid I Exposure. Oncotarget. 2016. Vol. 7:78872–7882

            111. Iliopoulos D, Hirsch HA, Struhl K. An Epigenetic Switch Involving NF-KappaB, Lin28, Let-7 MicroRNA, and IL6 Links Inflammation to Cell Transformation. Cell. 2009. Vol. 139:693–706

            112. Xie XC, Zhao N, Xu QH, Yang X, Xia WK, Chen Q, et al.. Relaxin Attenuates Aristolochic Acid Induced Human Tubular Epithelial Cell Apoptosis In Vitro by Activation of the PI3K/Akt Signaling Pathway. Apoptosis. 2017. Vol. 22:769–776

            113. Yang X, Thorngren D, Chen Q, Wang M, Xie X. Protective Role of Relaxin in a Mouse Model of Aristolochic Acid Nephropathy. Biomedicine Pharmacotherapy. 2019. Vol. 115:108917

            114. Zhao Z, Liang Z, Chan K, Lu G, Lee ELM, Chen H, et al.. A unique Issue in the Standardization of Chinese Materia Medica: Processing. Planta Medica. 2010. Vol. 76:1975–1986

            115. Ang LP, Ng PW, Lean YL, Kotra V, Kifli N, Goh HP, et al.. Herbal Products Containing Aristolochic Acids: A Call to Revisit the Context of Safety. Journal of Herbal Medicine. 2021. Vol. 28:100447

            116. Li Z-H, Yang B, Yang W-L, Chen H-F, Yang M, Yuan J-B, et al.. Effect of Honey-Toasting on the Constituents and Contents of Aristolochic Acid Analogues in Aristolochiae Fructus. Zhong Yao Cai. 2013. Vol. 36:538–541

            117. Yang B, Li Z, Yang W, Chen H, Yuan J. The Effect of Various Drug Processing Technologies on the Contents of Aristolochic Acid Analogues in Axistolchiae Fructus. Lishizhen Medicine and Materia Medica Research. 2012. Vol. 23:2553

            118. Yuan J-B, Huang Q, Ren G, Shi M, Chen L, Yang W-L, et al.. Acute and Subacute Toxicity of the Extract of Aristolochiae Fructus and Honey-Fried Aristolochiae Fructus in Rodents. Biological and Pharmaceutical Bulletin. 2014. Vol. 37:387–393

            119. Fan Y, Li Z, Xi J. Recent Developments in Detoxication Techniques for Aristolochic Acid-Containing Traditional Chinese Medicines. RSC Advances. 2020. Vol. 10:1410–1425

            120. Wang W, Zhang J, Cheng J. Effect of Salvia Miltiorrhiza on Renal Pathological Change and Expression of ACE and ACE2 in Rats with Aristolochic Acid Induced Nephropathy. Chinese Journal of Integrated Traditional and Western Nephrology. 2009. Vol. 10:109–112

            121. Ruan Y, Hu X, Zhao Y. Effect of Chinese Herbal Medicine Compatibility on Aristolochic Acid Content in Dangguisini Decoction. Chinese Archives of Traditional Chinese Medicine. 2012. Vol. 30:549–551

            122. Wang P, Guo W, Huang G, Zhen J, Li Y, Li T, et al.. Berberine-Based Heterogeneous Linear Supramolecules Neutralized THE Acute Nephrotoxicity OF Aristolochic Acid by the Self-Assembly Strategy. ACS Applied Materials & Interfaces. 2021. Vol. 13:32729–32742

            123. Wang X, Hui F, Yang Y, Yang S. Deep Sequencing and Transcriptome Analysis to Identify Genes Related to Biosynthesis of Aristolochic Acid in Asarum Heterotropoides. Scientific Reports. 2018. Vol. 8:17850

            124. Wu TK, Pan YR, Wang HF, Wei CW, Yu YL. Vitamin E (α-tocopherol) ameliorates aristolochic acid-induced renal tubular epithelial cell death by attenuating oxidative stress and caspase-3 activation. Molecular Medicine Reports. 2018. Vol. 17:31–36

            125. Wu TK, Wei CW, Pan YR, Cherng SH, Chang WJ, Wang HF, et al.. Vitamin C Attenuates the Toxic Effect of Aristolochic Acid on Renal Tubular Cells Via Decreasing Oxidative Stress-Mediated Cell Death Pathways. Molecular Medicine Reports. 2015. Vol. 12:6086–6092

            126. Zhang J, Chan CK. Identifying Cysteine, N-Acetylcysteine, and Glutathione Conjugates as Novel Metabolites of Aristolochic Acid I: Emergence of a New Detoxification Pathway. Chemical Research in Toxicology. 2020. Vol. 33:1374–1381

            127. Zhang YL, Wang YL, Yan K, Deng QQ, Li FZ, Liang XJ. Nanostructures in Chinese Herbal Medicines (CHMs) for Potential Therapy. Nanoscale Horizons. 2023. Vol. 8:976–990

            128. Wang JB, Bai ZF, Xiao XH. Letter to the Editor: Is Aristolochic Acid the Major Cause of Liver Cancer in China and Asia? Hepatology. 2020. Vol. 71:1130

            129. Feng C, Xie X, Wu M, Li C, Gao M, Liu M, et al.. Tanshinone I Protects Mice from Aristolochic Acid I-Induced Kidney Injury by Induction of CYP1A. Environmental Toxicology and Pharmacology. 2013. Vol. 36:850–857

            130. Chang MM, Lin CN, Fang CC, Chen M, Liang PI, Li WM, et al.. Glycine N-methyltransferase Inhibits Aristolochic Acid Nephropathy by Increasing CYP3A44 and Decreasing NQO1 Expression in Female Mouse Hepatocytes. Scientific Reports. 2018. Vol. 8:6960

            131. Zhang Q, Luo P, Chen J, Yang C, Xia F, Zhang J, et al.. Dissection of Targeting Molecular Mechanisms of Aristolochic Acid-Induced Nephrotoxicity via a Combined Deconvolution Strategy of Chemoproteomics and Metabolomics. International Journal of Biological Sciences. 2022. Vol. 18:2003–2017

            132. Luo P, Chen J, Zhang Q, Xia F, Wang C, Bai Y, et al.. Dissection of Cellular and Molecular Mechanisms of Aristolochic Acid-Induced Hepatotoxicity via Single-Cell Transcriptomics. Precision Clinical Medicine. 2022. Vol. 5:pbac023

            133. Chen J, Luo P, Wang C, Yang C, Bai Y, He X, et al.. Integrated Single-Cell Transcriptomics and Proteomics Reveal Cellular-Specific Responses and Microenvironment Remodeling in Aristolochic Acid Nephropathy. JCI Insight. 2022. Vol. 7:e157360

            Author and article information

            Journal
            amm
            Acta Materia Medica
            Compuscript (Ireland )
            2737-7946
            30 September 2024
            : 3
            : 3
            : 349-362
            Affiliations
            [a ]Department of Nephrology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China
            [b ]School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong, China
            [c ]State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
            [d ]State Key Laboratory of Southwestern Chinese Medicine Resource, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
            [e ]National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
            [f ]Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
            [g ]Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou, China
            Author notes

            1These authors contributed equally.

            Article
            10.15212/AMM-2024-0023
            91a7f68e-32fd-4b8c-8e5e-8b648be9b4c8
            2024 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 04 May 2024
            : 05 August 2024
            : 22 August 2024
            Page count
            Figures: 5, Tables: 1, References: 133, Pages: 14
            Funding
            Funded by: National Key Research and Development Program of China
            Award ID: 2022YFC2303600
            Funded by: Science and Technology Foundation of Shenzhen (Shenzhen Clinical Medical Research Center for Geriatric Diseases)
            Award ID: 2017ZX09101002-001-001-05
            Funded by: Major National Science and Technology Program of China for Innovative Drug
            Award ID: 2017ZX09101002-001-001-05
            Funded by: National Natural Science Foundation of China
            Award ID: 82074098
            Funded by: National Natural Science Foundation of China
            Award ID: 82304827
            Funded by: National Natural Science Foundation of China
            Award ID: 82305014
            Funded by: Science and Technology Project of Guangzhou City
            Award ID: 2024A04J4155
            Funded by: Science and Technology Project of Guangzhou City
            Award ID: 2024A04J4573
            Funded by: Guangdong Provincial Bureau of Traditional Chinese Medicine Research Project
            Award ID: 20241202
            Funded by: Guangdong Provincial Bureau of Traditional Chinese Medicine Research Project
            Award ID: 20241205
            The authors gratefully acknowledge the National Key Research and Development Program of China (2022YFC2303600); the Science and Technology Foundation of Shenzhen (Shenzhen Clinical Medical Research Center for Geriatric Diseases), the Major National Science and Technology Program of China for Innovative Drug (2017ZX09101002-001-001-05), the National Natural Science Foundation of China (82074098, 82304827, and 82305014), the Science and Technology Project of Guangzhou City (2024A04J4155 and 2024A04J4573), and the Guangdong Provincial Bureau of Traditional Chinese Medicine Research Project (20241202 and 20241205).
            Categories
            Review Article

            Toxicology,Pathology,Biochemistry,Clinical chemistry,Pharmaceutical chemistry,Pharmacology & Pharmaceutical medicine
            Genotoxicity,Hepatotoxicity,Aristolochic acid,Carcinogenicity,Herbal medicine

            Comments

            Comment on this article