Lupine Publishers Group

Lupine Publishers

  Submit Manuscript

ISSN: 2641-1652

Current Trends in Gastroenterology and Hepatology

Mini Review(ISSN: 2641-1652)

Different Toxicity of Aristolochic Acids in Kidney and Liver Volume 3 - Issue 2

Shuzhen Chen1,2 and Hongyang Wang1,2*

  • 1National Center for Liver Cancer, Second Military Medical University, China
  • 2International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, China

Received:November 17, 2021   Published: November 24, 2021

*Corresponding author: Hongyang Wang, International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, China

DOI: 10.32474/CTGH.2021.03.000161

 

Abstract PDF

Introduction

Aristolochic acid (AAs) is a group of nitrophenanthrene compounds comprised of AAI, AAII, AAIII and AAIV, which are widely found in Aristolochia plants and used in herbal therapy and traditional Chinese medicine [1]. Consistent use of aristolochic acids- containing drugs could lead to aristolochic acid nephropathy and subsequent urinary tract tumors [2-4]. Active metabolites of AAs form adducts with DNA, inducing characteristic A-T transversion (A:T to T:A mutation) known as AA mutational signature [5]. In 2017, a study has analyzed AA mutational signature of several datasets and concluded that AAs and their derivatives were widely implicated in liver cancers in Taiwan and throughout Asia [6]. Ever since the paper published, there has been an intensive debate on whether the prevalence of AA signature mutation is high in HCC patients and if this mutation spectra is really correlate with traditional Chinese medicine consumption in Asia. Since no case report has linked AAI to liver cancer by far, many researchers held doubts regarding AA-induced liver cancer. Herein, we summarized previous reports of animal experiments indicating the organ specified toxicity in kidney other than liver and shared our opinion about the possible reasons.

For long, several reports have linked AAs to the development of urothelial cancer, kidney and forestomach tumors in rodents [7-11]. Although AA could be bioactivated in both kidney and liver, in most studies, it only induces tumors in kidney [12]. Therefore, kidney was usually considered as the prior target organ of AAs. AA-DNA adduct is a well-known biomarker for AA exposure. Studies conducted on rat kidney and liver found that kidney had at least two-fold higher levels of DNA adducts and mutant frequency than livers inducted by AAI [13, 14]. The same dose didn’t cause liver tumor in rat, but DNA adducts were detectable at lower levels than kidney [13]. The experiment on Muta mice showed the same tendency [15]. A most recent study also indicated that although forestomach carcinoma was the main cause of death in long-term small dose (0.3-3.0 mg/ kg) AAI-treated mice, kidney was still the organ with most AA-DNA adducts accumulation compared with forestomach and liver [16].

There are several possible reasons for the tissue specificity of AA, one of which could be the ability of proximal tubules to transport and concentrate AA and their metabolites, resulting in renal toxicity. OAT family, mainly expressed on renal proximal tubules, is considered to be one of the pivotal determinants mediating the accumulation of AAI into the proximal tubules [17]. In addition, the level of enzymes catalyzing the reductive activation of AAI are varied in different cells. The activation pathway for AAI is nitroreduction catalyzed by both cytosolic and microsomal enzymes. One of the main human and rat enzymes activating AA-I toxicity was NAD(P) H:quinone oxidoreductase (NQO1), present in hepatic and renal cytosolic subcellular fractions. Other involving enzymes include NADPH: CYP reductase (POR) in kidney microsomes and protaglandin H synthase (cyclooxygenase, COX) in urothelial tissues [18]. In addition to gene expression level of the AAI activation related enzymes in liver and kidney, in vivo oxygen concentration in specific tissues might also affect the balance between AAI nitroreduction and demethylation, which in turn would influence tissue-specific toxicity or carcinogenicity [19]. A recent study also indicated that hepatocyte-specific metabolism of AA-I substantially increases its cytotoxicity toward kidney proximal tubular epithelial cells, including formation of aristolactam adducts and release of kidney injury biomarkers [20].Moreover, AA exposure could cause significantly altered gene expression profiles between kidney and liver, involving defense response, apoptosis and immune response, cell cycle etc, which might also be possible reasons for the tissue-specific toxicity and carcinogenicity of AA [12, 21].

Although the toxicity and carcinogenesis of AAs in kidney is well-defined, their role in liver damage and tumor development may be different. Besides, AA exposure as the main cause of liver cancer was not consistent with the actual scenario in Asia since hepatitis B virus infection remains as the highest risk. Therefore, we believe the toxicity of AAs in liver and kidney should be considered separately.

References

  1. Yang HY, Chen PC, Wang JD (2014) Chinese herbs containing aristolochic acid associated with renal failure and urothelial carcinoma: A review from epidemiologic observations to causal inference. Biomed Res Int: 569325.
  2. Vanherweghem JL, Depierreux M, Tielemans C, Abramowicz D, Dratwa M, et al. (1993) Rapidly progressive interstitial renal fibrosis in young women: Association with slimming regimen including Chinese herbs. Lancet 341(8842): 387-391.
  3. Nortier JL, Martinez MC, Schmeiser HH, Arlt VM, Bieler CA, et al. (2000) Urothelial carcinoma associated with the use of a Chinese herb (Aristolochia fangchi). N Engl J Med 342(23): 1686-1692.
  4. Nortier JL, Vanherweghem JL (2002) Renal interstitial fibrosis and urothelial carcinoma associated with the use of a Chinese herb (Aristolochia fangchi). Toxicology 181-182.
  5. Hoang ML, Chen CH, Sidorenko VS, He J, Dickman KG, et al. (2013) Mutational signature of aristolochic acid exposure as revealed by whole-exome sequencing. Sci Transl Med 5(197): 197ra02.
  6. Ng AWT, Poon SL, Huang MN, Lim JQ, Boot A, et al. (2017) Aristolochic acids and their derivatives are widely implicated in liver cancers in Taiwan and throughout Asia. Sci Transl Med 9(412): 6446.
  7. Yi JH, Han SW, Kim WY, Kim J, Park MH (2018) Effects of aristolochic acid I and/or hypokalemia on tubular damage in C57BL/6 rat with aristolochic acid nephropathy. Korean J Intern Med 33(4): 763-773.
  8. Wang YY, Li Z, Chen T, Zhao XM (2015) Understanding the aristolochic acid toxicities in rat kidneys with regulatory networks. IET Syst Biol 9(4): 141-146.
  9. Yeh YH, Lee YT, Hsieh HS, Hwang DF (2008) Short-term toxicity of aristolochic acid, aristolochic acid-I and aristolochic acid-II in rats. Food Chem Toxicol 46(3): 1157-1163.
  10. Mengs U, Stotzem CD (1993) Renal toxicity of aristolochic acid in rats as an example of nephrotoxicity testing in routine toxicology. Arch Toxicol 67(5): 307-311.
  11. Mengs U (1988) Tumour induction in mice following exposure to aristolochic acid. Arch Toxicol 61(6): 504-505.
  12. Chen T, Guo L, Zhang L, Shi L, Fang H, et al. (2006) Gene expression profiles distinguish the carcinogenic effects of aristolochic acid in target (kidney) and non-target (liver) tissues in rats. BMC Bioinformatics 7(Suppl 2): 20.
  13. Mei N, Arlt VM, Phillips DH, Heflich RH, Chen T (2006) DNA adduct formation and mutation induction by aristolochic acid in rat kidney and liver. Mutat Res 602(1-2): 83-91.
  14. Li XL, Guo XQ, Wang HR, Chen T, Mei N (2020) Aristolochic Acid-Induced Genotoxicity and Toxicogenomic Changes in Rodents. World J Tradit Chin Med 6(1): 12-25.
  15. Kohara A, Suzuki T, Honma M, Ohwada T, Hayashi M (2002) Mutagenicity of aristolochic acid in the lambda/lacZ transgenic mouse (MutaMouse). Mutat Res 515(1-2): 63-72.
  16. Chen SZ, Dong YP, Qi XM, Cao QQ, Luo T, et al. (2021) Aristolochic acids exposure was not the main cause of liver tumorigenesis in adulthood. Acta Pharmaceutica Sinica B.
  17. Dickman KG, Sweet DH, Bonala R, Ray T, Wu A (2011) Physiological and molecular characterization of aristolochic acid transport by the kidney. J Pharmacol Exp Ther 338(2): 588-597.
  18. Stiborova M, Hudecek J, Frei E, Schmeiser HH (2008) Contribution of biotransformation enzymes to the development of renal injury and urothelial cancer caused by aristolochic acid: Urgent questions, difficult answer. Interdiscip Toxicol 1(1): 8-12.
  19. Stiborova M, Levova K, Barta F, Shi Z, Frei E, Schmeiser HH, et al. (2012) Bioactivation versus detoxication of the urothelial carcinogen aristolochic acid I by human cytochrome P450 1A1 and 1A2. Toxicol Sci 125(2): 345-358.
  20. Chang SY, Weber EJ, Sidorenko VS, Chapron A, Yeung CK, et al. (2017) Human liver-kidney model elucidates the mechanisms of aristolochic acid nephrotoxicity. JCI Insight 2(22): 95978.
  21. Arlt VM, Zuo J, Trenz K, Roufosse CA, Lord GM, Nortier JL, et al. (2011) Gene expression changes induced by the human carcinogen aristolochic acid I in renal and hepatic tissue of mice. Int J Cancer 128(1): 21-32.

https://www.high-endrolex.com/21