Academia.eduAcademia.edu
review http://www.kidney-international.org & 2008 International Society of Nephrology Aristolochic acid nephropathy: A worldwide problem Frédéric D. Debelle1,2, Jean-Louis Vanherweghem1 and Joëlle L. Nortier1,2 1 Department of Nephrology, Dialysis and Renal Transplantation, Erasme Hospital, Brussels, Belgium and 2Experimental Nephrology Unit, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium Aristolochic acid nephropathy (AAN), a progressive renal interstitial fibrosis frequently associated with urothelial malignancies, was initially reported in a Belgian cohort of more than 100 patients after the intake of slimming pills containing a Chinese herb, Aristolochia fangchi. Although botanicals known or suspected to contain aristolochic acid (AA) were no longer permitted in many countries, several AAN cases were regularly observed all around the world. The incidence of AAN is probably much higher than initially thought, especially in Asia and the Balkans. In Asian countries, where traditional medicines are very popular, the complexity of the pharmacopoeia represents a high risk for AAN because of the frequent substitution of the botanical products by AA-containing herbs. In the Balkan regions, the exposure to AA found in flour obtained from wheat contaminated with seeds of Aristolochia clematitis could be responsible for the so-called Balkan-endemic nephropathy. Finally, despite the Food and Drug Administration’s warnings concerning the safety of botanical remedies containing AA, these herbs are still sold via the Internet. Kidney International (2008) 74, 158–169; doi:10.1038/ki.2008.129; published online 16 April 2008 KEYWORDS: aristolochic acid; Aristolochia; renal interstitial fibrosis; urothelial carcinoma; herbal remedies; Balkan-endemic nephropathy Aristolochic acid nephropathy (AAN), a rapidly progressive interstitial nephritis leading to end-stage renal disease and urothelial malignancy, was originally reported in Belgium in a group of patients who had ingested slimming pills containing powdered root extracts of Chinese herbs (Figure 1a and b).1–4 This nephropathy, initially called Chinese-herb nephropathy (CHN), appeared to be the dramatic consequence of a substitution of Stephania tetrandra by Aristolochia fangchi rich in aristolochic acid (AA), because both herbs share the same common name in Pin Yin (Han Fang Ji and Guang Fang Ji), and one can be used instead of the other in traditional Chinese medicine irrespective of their botanical classification.1,3,5 After the publication of the index cases, new cases of AAN were regularly reported, not only in Belgium but also worldwide (Figure 1a).1–4,6–23 Actually, the true incidence of AAN is largely unknown and probably underestimated, as numerous ingredients known or suspected to contain AA are used in traditional medicine in China, Japan, and India (Figure 1c).24–26 Another reason to suspect a higher number of AAN cases is based on the hypothesis that AA could be an environmental cause of Balkan-endemic nephropathy (BEN), which is a familial chronic tubulointerstitial disease characterized by an insidious onset, a slow progression to end-stage renal disease, and an increased frequency of urothelial cancer.23 This nephropathy is endemic in Serbia, Bosnia, Croatia, Bulgaria, and Romania (Figure 1d).27 Finally, despite the Food and Drug Administration’s warnings regarding the safety of botanical remedies containing AA (known or suspected to contain AA), plants containing AA are still available via the Internet.28 AAN: THE BELGIAN OUTBREAK OF CHN Correspondence: Joëlle L. Nortier, Nephrology Department, Erasme Hospital, Route de Lennik, 808, Brussels B-1070, Belgium. E-mails: jnortier@ulb.ac.be or Joelle.Nortier@erasme.ulb.ac.be Received 3 December 2007; revised 24 January 2008; accepted 6 February 2008; published online 16 April 2008 158 In 1992, two young women with no previous history of renal disease were admitted in our Nephrology department in Brussels (Belgium) with severe interstitial nephritis that progressed over a couple of months to end-stage renal disease.1 One year before, these two patients had followed the same weight-loss program in the same medical clinic in Brussels. Interestingly, the composition of weight-reducing pills was modified in June 1990 by introducing root extracts from two Chinese herbs, labeled as S. tetrandra and Magnolia officinalis.1 An epidemiological survey of the Nephrology centers of Brussels revealed an unusual, increased incidence of patients with ‘interstitial nephritis of unknown origin’ Kidney International (2008) 74, 158–169 review FD Debelle et al.: Aristolochic acid nephropathy admitted for dialysis in 1991 and 1992.1 This survey identified seven other women who had followed the same slimming regimen containing the Chinese herbs.1 Confronted to this rapidly progressive nephropathy probably related to the ingestion of these two Chinese herbs, the Belgian authorities decided to ban S. tetrandra and M. officinalis from the Belgian market at the end of 1992. Despite this safety measure, more than 100 cases of CHN were reported in Belgium in 1998, 70% of them being in end-stage renal disease.7 The time correlation between the introduction of the Chinese herbs in weight-loss regimens and the appearance of this rapidly progressive interstitial renal fibrosis focused the search of the culprit on the Chinese herbs.1 Very soon, the inadvertent replacement of Stephania by Aristolochia was suspected. First, S. tetrandra (Pin Yin name: Han Fang Ji) and A. fangchi (Pin Yin name: Guang Fang Ji) belong to the same therapeutic ‘Fang Ji’ family in traditional Chinese medicine, and the herbal ingredients are generally traded using their common Pin Yin name. Second, the pathological aspect of CHN is very similar to that of BEN, whose etiology is still controversial, but some suggested that AAs containing A. clematitis are the main culprit (see section below).1,2,29,30 The hypothesis of substitution was strengthened by the phytochemical analyses of the S. tetrandra batches revealing that most of them did not contain tetrandrine but AAs (0.65±0.56 mg g1 of powder), the main compounds of the Aristolochia sp.3 A survey including 71 CHN/AAN patients Belgium: 128 Germany: 1 UK: 4 Korea: 1 France: 4 Japan: 6 USA: 2 Spain: 1 Taiwan: 33 China: 116 3 2 1 Slimming pill and crosssection of a root of aristolochia Extensive paucicellular interstitial fibrosis and tubular atrophy typically found in end-stage CHN Autoradiographic pattern of specific aristolochic acid-related DNA adducts detected in human renal tissue Figure 1 | Aristolochic acid nephropathy: a worldwide problem. (a) Counting cases of CHN/AAN around the world reported in the literature.1–4,6–23 (b) CHNA/AAN is a rapidly progressive interstitial nephritis leading to end-stage renal disease and urothelial malignancy, which was originally reported in Belgium in the context of the intake of slimming pills containing powdered Chinese herbs (A. fangchi). (c) The true incidence of AAN is largely unknown and probably underestimated, as numerous ingredients known or suspected to contain AA are used in traditional medicines in India and Eastern Asia (see Tables 1–3 for more details). (d) Finally, another reason to suspect a higher number of AAN cases is based on the hypothesis that the exposure to seeds of Aristolochia clematitis comingled with wheat grain during the annual harvest could be responsible for BEN. Kidney International (2008) 74, 158–169 159 review FD Debelle et al.: Aristolochic acid nephropathy A pharmacy selling traditional herbal remedies including Fang Chi and Mu Tong Aristolochic acid containing Aristolochia clematitis growing in a field of wheat (Croatia) By courtesy of Dr Bojan Jelaković Figure 1 | Continued. followed in our department demonstrated in a multiple regression analysis that the cumulative dose of the so-called Stephania (in fact, Aristolochia) was the only significant factor predicting the slope of the inverse of plasma creatinine levels.31 The causal role of AA was definitively confirmed by the detection of AA DNA adducts in kidney removed from CHN/AAN patients showing evidence of a previous exposure to AA.32,33 In addition, the main histological and functional features of CHN/AAN were successfully reproduced by administrating AA to New Zealand white rabbits or male Wistar rats.34,35 Consequently, the term Chinese-herb nephropathy has been progressively abandoned and replaced by aristolochic acid nephropathy. After the first report of cases in Belgium, other similar cases were sporadically observed in France, Spain, Germany, United Kingdom, and the United States, in the context of herbal remedies for slimming purposes but also for all kinds of indications such as eczema, hepatitis B, ‘liver enhancement’, arthritis, rheumatism, and pain relief.6,8–10,13,15,17,20 As expected, numerous cases were also reported in Asian countries where the complexities of the traditional pharmacopoeia represent a high risk for AA exposure.12,14,16,19,21,22,36–40 Clinically, the initial presentation of CHN/AAN was usually silent and the renal failure was discovered by routine blood testing.41 However, few cases presenting with a Fanconi syndrome or an acute renal failure due to tubular necrosis were reported in the literature.11,12,15,19,21 Anemia was present and was often more severe than might have been anticipated from the degree of renal failure.42 In most of the cases, urinary sediment was unremarkable and dipstick analysis for albuminuria was negative.43 However, urinary excretion of five low molecular weight proteins (b2-microglobulin, cystatin C, Clara cell protein, retinal160 binding protein, and a1-microglobulin) was markedly increased in five patients with CHN/AAN, and the urinary low molecular weight protein/albumin ratio was higher than in control patients with glomerular diseases.44 Moreover, in 26 patients with CHN/AAN, levels of urinary neutral endopeptidase, a 94 kDa ectoenzyme of the proximal tubule brush border, were significantly decreased in those with moderate renal failure and almost undetectable in those with end-stage renal failure.45 NEP enzymuria positively correlated with individual creatinine clearance values (r ¼ 0.76; P ¼ 0.0001) and negatively correlated with urinary low molecular weight protein levels (r ¼ 0.55; P ¼ 0.00001).45 An in vitro study on the opossum kidney cell line demonstrated that AA intoxication led to a rapid and persistent decrease in megalin expression in parallel with an inhibition of receptor-mediated endocytosis of low molecular weight protein.46 Taking together, these data indicated that proximal tubular cells are the main target of the AA-containing Chinese herb. Macroscopically, the kidneys were shrunk, asymmetric in about half of the cases with irregular outlines in one-third.43 Microscopically, an extensive interstitial fibrosis with atrophy and loss of proximal tubules was the predominant lesion, which was mainly located in the superficial cortex and progressed toward the deep cortex (Figure 1b).2,29 The glomeruli were relatively spared, although, in the later stage of the disease, they displayed a mild collapse of the capillaries and a wrinkling of the basement membrane. Although it was not the general rule, an interstitial inflammatory infiltration was retrieved in several renal biopsy specimens, suggesting an immunological process as a possible pathophysiological mechanism. This observation was the rational basis for a pilot study with corticosteroids performed in 35 CHN/AAN patients with chronic renal failure. Compared to an untreated group, a significant reduction of the number of patients reaching end-stage renal disease was observed after 1 year of steroid therapy.47 Eight years later, in a larger group of CHN patients, the steroid therapy was confirmed to slow down the progression of renal failure.48 Finally, the finding of an endothelial wall thickening in the interlobular and afferent arterioles suggested a possible ischemic process induced by other substances concomitantly administrated with the Chinese herb.49 The appetite suppressant (dex)fenfluramine, a serotonin agonist, could have played a role in the development of CHN because serotonin injection was experimentally shown to induce ischemic renal lesions progressing to renal fibrosis.50 However, this hypothesis can be ruled out on the basis of reports of CHN cases out of the context of a slimming regimen and the demonstration that dexfenfluramine did not enhance the nephrotoxicity of AA in a rat model of CHN/AAN.41,51 In the same line, an extrarenal toxicity of Chinese herbs was also suspected because 30–50% of the CHN patients displayed an aortic insufficiency.43 However, the puzzling association of valvular abnormalities with CHN appeared to be mainly linked to the concomitant presence of anorectic drugs in the Kidney International (2008) 74, 158–169 review FD Debelle et al.: Aristolochic acid nephropathy slimming pills, with the demonstration of a significant dose–response relationship between the cumulative dose of (dex)fenfluramine and the aortic regurgitation.52,53 AA CONTAINING HERBAL REMEDIES IN TRADITIONAL MEDICINES The uncontrolled use and the uncorrected identification of medicinal herbs, which are usually considered by the general population as inherently harmless, were at the heart of all AAN cases encountered in the Western countries.54 On the other hand, in Asia, AAN was the dramatic consequence of the complex pharmacopoeia of herbal remedies used in traditional medicines and the lack of their regulation as applied for conventional drugs.5 For example, two series of 12 and 20 AAN cases, respectively, related to the use of various herbal medications were reported in Taiwan.14,16 The ingestion of AA-containing herbal remedies used in traditional Sino-Japanese Kampo prescriptions (vernacular names: Boui and Mokutsu) resulted in Fanconi syndrome secondary to AAN in four Japanese patients.11,12 In China, acute renal failure secondary to tubular necrosis was observed in eight patients after the intake of Guanmutong (Aristolochia manshuriensis Kom.), an AA-containing Chinese herb widely used for the treatment of urinary and cardiovascular diseases.21 Two series of 58 and 51 AAN cases, respectively, were reported in 2001 in China, most of them after the intake of the liver tonic Longdan Xieganwan that contained Caulis Aristolochia Manshuriensis.22 Five additional AAN cases were also observed in Hong Kong, following the substitution of the nontoxic Herba Solani Lyrati by the AA-containing herb Aristolochia mollissimae.22 The preferential use of vernacular names in traditional medicine terminology and the high risk for substitution of the herbal products might explain the outbreak of AAN in Asia (for more details, see Tables 1–3). However, one can expect that the true incidence of AAN in Asia is largely underestimated. Indeed, Akebia is commonly used in Sino-Japanese prescriptions as well as Fang ji and Mu Tong in traditional Chinese medicine.24,25 It is worthy to stress that traditional medicine is still widely popular in China in 2007: about 3000 hospitals provide traditional Chinese medicine treatments to nearly 234 million patients each year.60 Despite the fact that the AA-containing herbs were theoretically banned in many countries around the world, including Table 1 | Botanicals known or suspected to contain aristolochic acid and their vernacular names55–59 Botanical name Common or other names Aristolochia Aristolochia Aristolochia Aristolochia Aristolochia Aristolochia Aristolochia Aristolochia, Guan Mu tong, Guang Mu tong Oval leaf Dutchman’s pipe Ukulwe Birthwort Ma Dou Ling (fruit), Bei Ma Dou Ling (root), Tian Xian Teng (herb) Mil homens Ma Dou Ling (fruit); Tian Xian Teng (herb), Qing Mu Xiang (root), Sei-Mokkou (Japanese), Birthwort, Long birthwort, Slender Dutchman’s pipe Guang Fang ji (root), Fang ji, Fang chi, Mokuboi (Japanese), Kou-boui (Japanese), Kwangbanggi (Korean) Han Fang Ji Indian birthwort (root), Yin Du Ma Dou Ling Yellowmouth Dutchman’s pipe, Zhu Sha Lian spp. acuminata (Syn. Aristolochia tagala) bracteata clematitis contorta cymbifera debilis (Syn. Aristolochia longa, A. recurvilabra, A. sinarum) Aristolochia fangchi Aristolochia heterophylla Aristolochia indica Aristolochia kaempferi (Syn. Aristolochia chrysops, A. feddei., A. heterophylla, A. mollis, A. setchuenensis, A. shimadai, A. thibetica, Isotrema chrysops, I. heterophylla, I. lasiops) Aristolochia macrophylla (Syn. Aristolochia sipho) Aristolochia manschuriensis (Syn. Hocquartia manshuriensis, Syn. Isotrema manchuriensis) Aristolochia maxima (Syn. Howardia hoffmannii) Aristolochia mollissima Aristolochia moupinensis Aristolochia serpentaria (Syn. Aristolochia serpentaria) Aristolochia triangularis Aristolochia tuberosa Aristolochia tubiflora Aristolochia versicolar Asarum canadense (Syn. Asarum acuminatum, A. ambiguum, A. canadense, A. furcatum, A. medium, A. parvifolium, A. reflexum, A. rubrocinctum) Asarum himalai(y)cum Asarum splendens Dutchman’s-pipe Manchurian birthwort, Manchurian Dutchman’s pipe (stem) Guan Mutong (stem), Kan-Mokutsu (Japanese), Mokuboi (Japanese), Kwangbanggi (Korean) Maxima Dutchman’s pipe, Da Ma Dou Ling Wooly Dutchman’s pipe, Mian Mao Ma Dou Ling Moupin Dutchman’s pipe, Huai Tong Virginia snakeroot, Serpentaria, Virginia serpentary Triangular Dutchman’s pipe, San Jiao Ma Dou Ling Tuberous Dutchman’s pipe, Kuai Jing Ma Dou Ling Tubeflower Dutchman’s pipe, Guan Hua Ma Dou Ling Versicolorous Dutchman’s pipe, Bian Se Ma Dou Ling Wild ginger, Indian ginger, Canada snakeroot, False coltsfoot, Colic root, Heart snakeroot, Vermont snakeroot, Southern snakeroot, Jia Na Da Xi Xin Tanyou-saishin (Japanese) Do-saishin (Japanese) Other botanicals known or suspected to contain aristolochic acid are Aristolochia argentina, A. baetica. (Syn. A. bracteolata), A. chilensis, A. cinnabarina, A. elegans (Syn. A. hassleriana), A. esperanzae, A. fimbriata, A. kwangsiensis (Syn. A. austroszechuanica), A. maurorum, A. rigida. A. rotunda, A. watsoni(i) (Syn. A. porphyrophylla), A. westlandi(i), A. zollingeriana (Syn. A. kankauensis, A. roxburghiana, A. tagala, Hocquartia kankauensis), Bragantia wallichii. Kidney International (2008) 74, 158–169 161 review FD Debelle et al.: Aristolochic acid nephropathy Table 2 | Botanicals that may be adulterated with aristolochic acid and their respective vernacular names55–59 Botanical name Common or other names Akebia spp. Akebia, Mu tong, Ku mu tong, Zi mutong, Bai mu tong, Mokutsu (Japanese), Mokt’ong (Korean) Chocolate vine, Fiveleaf akebia, Bai Mu Tong (stem), Mu Tong Gen (root), Yu zhi zi (seed), Mokutsu (Japanese) Bai Mu Tong (stem), Bai Mu Tong Gen (root), Bai Mu Tong Zi (seed), Three leaf akebia, Yu zhi zi, San Ye Mu Tong Zi (seed), San Ye Mu Tong Gen (root) Wild ginger (root, leaves) Batei-saishin (Japanese) Keirin-saishin (Japanese) Chinese wild ginger, Manchurian wild ginger, Bei Xi Xin, Xin xin Usuba-saishin (Japanese) Chinese wild ginger, Xi Xin, Hua Xi Xin, Manchurian wild ginger, Siebold’s wild ginger Clematis, Mufangji, Clematidis, Ireisen (Japanese), Wojoksum (Korean) Armand’s clematis Chuan Mu tong (stem), Xiao mu tong, Armand’s virgin bower Chinese clematis, Wei Ling Xian Chuan Mu Tong (stem) Cocculus Indian cockle, Yin Du Mu Fang Ji Mu Fang Ji Mu Fang Ji (root) Akebia quinata (Syn. Rajania quinata) Akebia trifoliata Asarum caudatum Asarum forbesii Asarum heterotropoides (Syn. Asarum heterotropoides) Asarum sieboldii (Syn. Asarum sieboldii, A. sieboldii var. seoulensis, A. heterotropoides var. seoulense, A. sieboldii) Clematis spp. Clematis armandii (Syn. Clematis armandii fo. Farquhariana, C. armandii var. biondiana, C. biondiana, C. ornithopus) Clematis chinensis Clematis montana (Syn. Clematis insulari-alpina) Cocculus spp. Cocculus indicus (Syn. Anamirta paniculata) Cocculus orbiculatus (Syn. Cissampelos pareira) Cocculus orbiculatus (Syn. Cocculus cuneatus, C. sarmentosus, C. sarmentosus var. linearis, C. sarmentosus var. pauciflorus, C. sarmentosus var. stenophyllus, C. thunbergii, C. trilobus, Menispermum orbiculatus, M. trilobum, Nephroia sarmentosa) Moku-boui (Japanese) Cocculus palmatus Cocculus palmatus (Syn. Jateorhiza miersii) Cocculus pendulus (Syn. Cebatha pendula, Epibaterium pendulus, Cocculus epibaterium) Cocculus trilobus Diploclisia chinensis Saussurea lappa Sinomenium acutum (Syn. Cocculus diversifolius var. cinereus, C. heterophyllus, Menispermum acutum, Sinomenium acutum var. cinereum, S. diversifolium) Stephania spp. Stephania tetrandra Vladimiria souliei Columba, Columbo Colombo Chui Mu Fang Ji Mu Fang Ji (root) Xiangfangchi Mokkou (Japanese) Orientvine, Xunfengteng, Dafengteng, Daqingmuxinag, Zhuigusan, Da ye qingshener, Mufangji, Hanfangji, Tuteng, Zhuigufeng, Maofangji Stephania Fen fang ji, Fang ji (root), Han fang ji (root), Kanboi (Japanese), Hanbanggi (Korean), Fun-boui (Japanese) Sen-mokkou Other botanicals that may be adulterated with aristolochic acid are: Clematis hexapetala, Clematis uncinata, (Syn. Clematis alsomitrifolia, C. chinensis var. uncinata, C. drakeana, C. floribunda, C. gagnepainiana, C. leiocarpa, C. ovatifolia, C. uncinata var. biternata, C. uncinata var. coriacea, C. uncinata var. floribunda, C. uncinata var. ovatifolia, C. uncinata var. taitongensis), Cocculus carolinus (Syn. Cebatha carolina, Epibaterium carolinum, Menispermum carolinum), Cocculus diversifolius (Syn. Cocculus madagascariensis), Cocculus hirsutus (Syn. Cocculus villosus, Menispermum hirsutum), Cocculus laurifolius (Syn. Cinnamomum esquirolii), Cocculus leaebe, Cocculus madagascariensis (Syn. Cocculus diversifolius), Cocculus thunbergii, Diploclisia affinis (Syn. Diploclisia chinensis, Cocculus affinis), Menispernum dauricum. several in Asia, the risk of their mistaken use in traditional Chinese medicine is still high and could lead to a major concern for public health. Considering this fact, a reasonable way to decrease this risk should be the systematic quality control of herbal preparations by using reproducible and accurate analytical methods, such as high-performance liquid chromatography, liquid chromatography/mass spectrometry, or capillary electrophoresis.61,62 Interestingly, Mani63 reported in a series of 2028 Indian patients with chronic kidney disease that chronic interstitial nephritis was a frequent cause (27.8%). We can speculate that some of them are AAN, as Indian folk medicine used more than 7500 plant species, including Aristolochia bracteata, Aristolochia tagala, and Aristolochia indica.26 The identification of AA-related specific adducts on renal tissue could confirm this hypothesis.32 162 AA-ASSOCIATED UROTHELIAL MALIGNANCIES Clinical findings The striking association between AA exposure and the presence of urothelial abnormalities was described for the first time by Cosyns et al.,29 who observed moderate atypia and atypical hyperplasia of the urothelium in four pieces of nephroureterectomies removed from three CHN/AAN patients before or at the time of transplantation. In the same time, two cases of urothelial carcinoma were reported among the Belgian cohort of CHN/AAN patients.4,64 Considering the high risk for urothelial malignancy related to AA exposure, the prophylactic bilateral removal of the native kidneys and ureters was systematically proposed to CHN/AAN patients treated by dialysis or renal transplantation. It emerged that 40–45% of CHN/AAN patients displayed multifocal high-grade transitional cell carcinomas, mainly in Kidney International (2008) 74, 158–169 FD Debelle et al.: Aristolochic acid nephropathy Table 3 | Products in which Mu Tong and Fang Ji are declared as ingredients55,56 Name Ba Zheng Wan Chun Yang Zheng Ji Wan Da Huang Qing Wei Wan Dang Gui Si Ni Wan Dao Chi Wan Dieda Wan Fu Ke Fen Quing Wan Guan Xin Su He Wan Ji Sheng Ju He Wan Kat Kit Wan Long Dan Xie Gan Wan Quell Fire Shi Xiang Fan Shen Wan Xin Yi Wan the upper urinary tract.33,65 The cumulative ingested dose of Stephania (in fact, Aristolochia) was demonstrated to be a significant risk for the development of urothelial carcinomas.33 A further follow-up of these CHN/AAN patients was performed in relation with the high risk of bladder carcinoma. Prospective screening cystoscopies were proposed to all renaltransplanted CHN patients from our Nephrology department. Among the 38 kidney recipients who accepted this follow-up (cystoscopy and bladder biopsies every 6 months), bladder urothelial carcinoma was diagnosed in 15 patients, 68–169 months after cessation of AA exposure (cumulative incidence 39.5%): eight urothelial carcinoma in situ, four noninvasive low-grade papillary urothelial carcinoma, and three infiltrating urothelial cancer. Out of 17 patients, 12 patients (71%) with a previous history of upper tract urothelial carcinoma developed bladder cancer during the follow-up, whereas this occurred only in three out of 21 (14%) patients free of upper tract urothelial carcinoma (Po0.01). Despite local and/or systemic chemotherapy, three patients died and two radical cystectomies had to be performed.66 Almost all of the cases of urothelial cancers were detected in CHN/AAN patients with end-stage renal disease. However, the case of a generalized urinary tract cancer without a significant renal failure after the intake of AA-containing Chinese herbal remedies showed that a dissociation between carcinogenicity and nephrotoxicity of AAs may occur.67 Other cases of urothelial carcinoma have been reported outside Belgium: in Taiwan, United Kingdom, France, and Hong Kong.14,16,20,36,68,69 Interestingly, some of these cases, like those described by Laing, were reported after AA had been banned in the respective countries. Taking into account all cases of AAN and urothelial malignancies reported worldwide, the Food and Drug Administration issued warnings to healthcare professionals, industry associations, and consumers regarding the safety of botanical products and dietary complements containing AA. The FDA recommended that all botanical remedies known or suspected to contain AA be discarded.55,56 In 2002, the Kidney International (2008) 74, 158–169 review International Agency for Research on Cancer working group concluded that there was sufficient evidence in humans for the carcinogenicity of herbal remedies containing plant species of the genus Aristolochia (Group 1).57 AA activation, DNA-adducts formation, and carcinogenesis Aristolochic acids I (AAI) and II (AAII), two structurally related nitrophenanthrene carboxylic acids, are the major components of the AA mixture contained in the plant extract of the Aristolochia species. Several enzymes have been demonstrated to metabolize AAI and AAII to a cyclic N-acylnitrenium ion with a delocalized positive charge able to covalently bind to the exocyclic amino groups of purine bases and to form DNA adducts.40 The 7-(deoxyadenosineN6-yl) aristolactam I, 7-(deoxyguanosine-N2-yl) aristolactam II and 7-(deoxy-adenosin-N6-yl) aristolactam II are the main DNA adducts retrieved in AAN patients (Figure 2).33 Based on studies characterizing and quantifying the DNA adducts formed after modulation of metabolic pathways, the nitroreduction of AAs was demonstrated to be a crucial step leading to their ultimate DNA-binding species.40 In human hepatic microsomes, the reductive activation of AA is mainly mediated by cytochrome P450 (CYP) 1A2, and to a minor extent by CYP1A1, whereas in human renal microsomes, NADPH/CYP reductase is more effective in AA biotransformation.70,71 In addition, the NAD(P)H/quinine oxidoreductase and xanthine oxidase, two cytosolic enzymes found in human livers and kidneys, catalyze the activation of AAI to form DNA adducts.72 Several factors such as drugs, smoking habit, environmental chemicals, and genetic polymorphisms affect the expression levels and activities of these enzymes, which could explain variations between individuals in the susceptibility to AA toxicity. The phase II metabolism of AA consists of the presence in the urine and feces of AA metabolites in conjugated forms, such as glucuronides, sulfate, or acetate esters.73,74 However, the precise role of conjugation enzymes in AA activation needs further investigation.75 The predominant 7-(deoxyadenosine-N6-yl) aristolactam I in vivo, which is the most persistent DNA adduct detected in the target tissue, is a mutagenic lesion leading to AT-TA transversions. This specific mutation is retrieved at a high frequency in codon 61 of the H-ras protooncogene in tumors of rodents induced by AAI.76 In AAN patients, an overexpression of P53 protein was observed in urothelial atypia and carcinomas, suggesting that the p53 gene is also mutated.65 Furthermore, DNA-binding studies demonstrated that AAI and AAII preferentially react with purine bases in the human p53 gene, and the adduct distribution was not random.77 A specific AAG-to-TAG mutation in codon 139 (Lys-Stop) of exon 5 in p53 gene was detected in DNA isolated from one AAN-associated urothelial carcinoma.78 These mutations in the p53 gene probably trigger the tumorigenesis in AAN patients in the same way as the mutations in codon 61 of H-ras trigger the tumorigenesis in AA-intoxicated rodents. The metabolic activation of AA and its mediated carcinogenesis 163 review FD Debelle et al.: Aristolochic acid nephropathy Aristolochic acid I (AAI): R=OCH3 Aristolochic acid II (AAII): R=H Aristolactam I (AAI): R=OCH3 Aristolactam II (AAII): R=H O COOH O O N NO2 O O O O NH O + R R R Aristolactam nitrenium ion +DNA O O O R R N dA-AAI dA-AAII NH N NH N C O N O O H O O N N H NH H H H N NH O O O H H O NH H H C H dG-AAI dG-AAII Figure 2 | Metabolic activation and DNA adduct formation of aristolochic acids I (AAI; R ¼ OCH3) and II (AAII; R ¼ H). The 7-(deoxyadenosine-N6-yl) aristolactam I and II and the 7-(deoxyguanosine-N2-yl) aristolactam I and II are formed after the reductive metabolic activation mediated by cytosolic reductases (NAD(P)H/quinone oxidoreductase, xanthine oxidase) as well as enzymes in hepatic (cytochrome P450 1A2 and 1A1, NADPH/CYP reductase) and renal microsomes (prostaglandin H synthase). was the subject of an exhaustive and comprehensive review recently published by Stiborova et al.75 AA TOXICITY: EXPERIMENTAL STUDIES Carcinogenic aspects In the 1980s, Mengs and colleagues devoted several experimental studies to the toxicity of AA, especially to their carcinogenic properties. The administration by gavage of a mixture of AA (77% AAI and 21% AAII) to male and female Wistar rats at the dosage of 0.1, 1, and 10 mg per kg body weight per day for 3–12 months led to the development of forestomach carcinoma and urothelial dysplasia.79 Following their initial observations, they examined the different steps of appearance and development of the AA-induced carcinoma in Wistar rats and NMRI mice.80,81 Just as CHN was frequently associated to urothelial dysplasia and malignancies, injections of AA to New Zealand white rabbits or male Wistar rats led to urothelial atypias.34,35 In the rat model, papillary urothelial carcinoma and fibrohistiocytic sarcoma at the injection site were also retrieved.35 Nephrotoxicity aspects and animal models for AAN The acute toxicity of AA was evaluated in Wistar rats and NMRI mice of both sexes (Table 4).82 The lethal dose 50 (LD50) ranged from 56 to 203 mg kg1 orally or 38 to 83 mg kg1 intravenously, depending on species and sex. The histological evaluation revealed severe tubular necrosis, atrophy of the lymphatic organs, and large areas of superficial 164 ulceration in the forestomach, followed by hyperplasia and hyperkeratosis of the squamous epithelium. According to the extensive tubular necrosis, the authors concluded that the animals died as a result of acute renal failure, although the renal functional parameters were not assessed.82 In a subacute toxicity study, a daily oral administration of 25 mg AA per kg body weight to male Wistar rats induced after 4 weeks a moderate renal tubular necrosis with a significant glucosuria and proteinuria.83 However, neither the proximal tubular atrophy nor the renal interstitial fibrosis, which are the typical histological findings for CHN, were reported in those studies. Following the Belgian outbreak of CHN in 1993 and the etiopathological hypothesis of AA, new experimental studies were therefore undertaken. However, first attempts to experimentally reproduce CHN failed: two groups of seven Wistar rats were orally given either pure AAs (10 mg per kg for 5 days a week for 3 months) or AA-containing herbal powders mixed with fenfluramine. At the time of killing, animals in both groups developed the expected tumors but not renal tubulointerstitial fibrosis.89 On the contrary, typical histological features of CHN/AAN, consisting of tubular atrophy, interstitial fibrosis, and urothelial atypias, were reproduced in 12 female New Zealand white rabbits after 17–21 months of intraperitoneal injections of 0.1 mg AA per kg body weight, 5 days a week.34 In the same time, we developed a short-term model for CHN by administrating subcutaneously 10 mg AA per kg body Kidney International (2008) 74, 158–169 review FD Debelle et al.: Aristolochic acid nephropathy Table 4 | Most relevant studies investigating the renal effects of aristolochic acid in animal models Species Dosage Duration AA components Renal findings Rat/mice 38–86 mg kg1 IV or 150–300 mg kg1 orally 0.2; 1.0; 5.0, or 25 mg kg1 day1 orally 10, 50, or 100 mg kg1 orally 0.1 mg kg1 day1, 5 days a week 1 or 10 mg kg1 day1 s.c. Once AAI (77%)/AAII (21%) 28 days AAI (77%)/AAII (21%) Once AAI (77%)/AAII (21%) 17–21 months 35 days AAI (44%)/AAII (56%) AAI (40%)/AAII (60%) Mice 5 mg kg1 day1 i.p 14 days AAI (44%)/AAII (56%) Rat 10 mg kg1 day1 s.c. 35 days AAI (40%)/AAII (60%) Mice 2.5 mg kg1 day1, 5 days a week 14 days AAI (55%)/AAII (45%) or AAI or AAII or Aristolactam I or AAIV Rat 10 mg kg1 day1 s.c. 35 days AAI (40%)/AAII (60%) Severe PT cells necrosis. LD50 ranged from 56 to 203 mg kg1 orally or from 38 to 83 mg kg1 i.v., depending on species and sex. Proteinuria and glucosuria. PT cells atypia and mild necrosis at highest dosage. Renal interstitial inflammatory cells infiltration. At 100 mg kg1: m sCr and BUN, proteinuria, m gGT and NAG enzymuria. m mitosis and necrosis of PT (pars recta) m sCr, glucosuria and low molecular weight proteinuria. Renal hypocellular interstitial fibrosis. At 10 mg kg1 day1: glucosuria, proteinuria, k LAP enzymuria, and m sCr on days 10 and 35. PT cells necrosis, mononuclear cells infiltrates (day 10), proximal tubular atrophy and interstitial fibrosis (day 35). During the regeneration phase (day 28), circulating transgenederived HGF reduced AA-induced interstitial fibrosis, partially through a k expression of TIMP-1 and m MMP-9 activity. RAS blockade reduced ED-1 + macrophage infiltration but not interstitial fibrosis induced by AA. m sCr, glucosuria, proteinuria, proximal tubule injury, mononuclear cells infiltration, and interstitial fibrosis. Nephrotoxicity depending on strains (C3H/He4Balb/ c4C57Bl6) and AA components (AAI4AAII). No nephrotoxicity of Aristolactam I and AAIV. Defective activation of antioxidative enzymes and mitochondrial damage. Impaired regeneration and apoptosis of PT cells. Interstitial infiltration of monocytes/macrophages and CD8+ lymphocytes. Loss of epithelial markers concomitantly to de novo expression of mesenchymal cell markers and disruption of TBM. Rat Rat NZW rabbit Rat Reference 82 83 84 34 35 85 86 87 88 Abbreviations: AA, aristolochic acid; BUN, blood urea nitrogen; gGT, gamma-glutamyltransferase; HGF, hepatocyte growth factor; i.p., intraperitoneal; i.v., intravenous; LAP, leucine aminopeptidase; LD50, lethal dose 50; MMP, matrix metalloproteinase; NAG, N-acetyl-b-D-glucosaminidase; NZW, New Zeeland white; PT, proximal tubule; RAS, renin–angiotensin system; s.c., subcuatenous; sCr, serum creatinine; TIMP, tissue inhibitor of metalloproteinase; TBM, tubular basement membrane. wt per day to male Wistar rats.35 On day 35, AA-treated rats displayed functional and histological renal impairment as a significant increase of serum creatinine and foci of severe proximal tubular atrophy surrounded by interstitial fibrosis. Nephrotoxicity of different components of AA was also studied in three strains of inbred male mice. The C3H/He mice intraperitoneally injected with 2.5 mg of AA per kg body weight, 5 days a week for 2 weeks, developed foci of proximal tubule cell injury surrounded by mononuclear cell infiltration on day 14.87 Two weeks later, signs of proximal tubule cell proliferation were observed, whereas the inflammatory cell infiltration became more severe and interstitial fibrosis occurred.87 In a CH3/He mice model, AAI exhibited a higher nephrotoxicity than AAII, which was also confirmed in in vitro studies on the proximal tubular LLC-PK1 cell line.87,90,91 Pathogenesis of AAN The pathophysiological mechanisms by which AA induces renal interstitial fibrosis are still largely unknown. The treatment with an angiotensin-converting enzyme inhibitor± angiotensin II receptor blocker did not modify the functional and structural renal impairments in AA-treated Wistar rats, suggesting that pathways leading to interstitial fibrosis seem to be independent of the renin–angiotensin system in this model.86 Kidney International (2008) 74, 158–169 An early phase of acute tubular necrosis preceding the development of tubular atrophy and interstitial fibrosis was also observed in several experimental studies.85,87,92 By using a transgenic mice model, Okada et al.85 demonstrated that hepatocyte growth factor did not interfere with the acute phase but reduced the severity of interstitial fibrosis during the tubular regeneration phase, partially through a decreased expression of tissue inhibitor of metalloproteinase-1 and increased matrix metalloproteinase-9 activity. Pozdzik et al.88 recently showed in the rat AAN model that AA tubulotoxicity resulted in defective activation of antioxidative enzymes and mitochondrial damage. The progressive tubular atrophy was related to impaired regeneration of proximal tubular epithelial cells and apoptosis secondary to caspase-3 activation. The accumulation of vimentin and a-smooth muscle actin-positive cells in the interstitial areas expressing transforming growth factor-b suggested an increase in resident peritubular fibroblasts and their activation into myofibroblasts. These activated resident fibroblasts are proposed as the main source of collagen deposition during experimental AAN.88 As reported in several in vivo and in vitro studies, apoptosis is likely involved in the process of AA-induced proximal tubular atrophy.85,90,93 In an in vitro study, LLC-PK1 cells exposed to AA displayed a rapid increase in their intracellular 165 review calcium content leading to endoplasmic reticulum and mitochondrial stress, which in turn causes activation of the caspase pathway and finally apoptosis.94 It should be noted that the potential impact of the AA–DNA adducts formation on the proximal tubular atrophy, for example, through a defect of DNA repair, is largely unknown and deserves further studies. AAN AND BEN: THE TWO FACES OF JANUS? Balkan-endemic nephropathy is characterized by chronic interstitial fibrosis with slow progression to end-stage renal disease and urothelial malignancy. It was first described about 50 years ago and affects residents of rural areas of Bulgaria, Bosnia, Croatia, Romania, and Serbia along the Danube river basin.95 As the etiology of BEN is currently unknown and different diagnostic criteria have been used in various countries, epidemiological data are difficult to compare. At least 25 000 individuals suffer from BEN or are suspected of having the disease, whereas the total number of people at risk in these countries may exceed 100 000. The significant epidemiologic features of BEN include (1) its focal occurrence in certain farming villages, with unaffected villages located in close vicinity; (2) a familial but not inherited pattern of disease, frequently affected members of the same household; (3) occurrence only in individuals who are older than 18 years, occurrence in o10% of households in endemic villages; and (4) a strong association with upper urinary tract urothelial carcinoma.27 A variety of environmental factors have been explored during the past 50 years, including heavy metals, arsenic, nitrogen species, silica, selenium deficiency, calcium and magnesium deficiency, polycyclic aromatic hydrocarbons in the water originating from Pliocene coal beds, viruses and bacteria, and mycotoxins.96 Of these factors, ochratoxin A (OTA) has been the most investigated target of research.97,98 Its presence in a variety of common foodstuffs, including cereal grains, was recognized more than three decades ago. OTA is classified by the International Agency for Research on Cancer99 as a possible human carcinogen (Group 2B) on the basis on sufficient evidence for carcinogenicity in experimental animals but inadequate evidence in humans. OTA was shown to be a powerful rodent carcinogen, causing liver tumors in mice and renal tumors in mice (males) and rats, in particular adenomas and invasive carcinomas with elevated DNA ploidy distribution.100,101 Nephrotoxicity of OTA is also well recognized in animals. OTA induced the so-called porcine nephropathy after longterm exposure. Initially described in 1976 by Krogh, this renal disease is characterized by lesions compatible with those observed in chronic interstitial nephropathy, including proximal tubule injury and interstitial fibrosis.95 However, as recently well summarized by Mally et al.102, in all the remaining animal species studied (rodents), the most frequent histological observations are nuclear enlargement and polyploidy of proximal tubule cells, reflecting nuclear 166 FD Debelle et al.: Aristolochic acid nephropathy division without cytokinesis. No interstitial fibrosis has been reported associated or not with proteinuria, glucosuria, and increased creatininemia. Except in specific cases of acute tubular necrosis, no similarity could be found in the tubulointerstitial compartment with histological lesions observed in AAN or BEN but, contrasting with BEN, interstitial fibrosis and tubular atrophy are not described.102 Moreover, kidney tumors in OTA-exposed rats developed from the straight segment of the proximal tubule. No upper urinary tract carcinoma has been described, contrasting with its high prevalence in AAN and BEN. Some studies found higher exposure (as measured by the intake of OTA and OTA levels in food stuff, blood, or urine) in individuals from endemic villages as compared with nonendemic villages (as reviewed by Stefanovic et al.; Long and Voice; Pfohl-Leszkowicz and Manderville; and Mally et al.).27,98,102,103 OTA–DNA-related adducts (but also AA–DNA adducts) were detected in kidney tissue from patients with urothelial carcinoma or ureteral stenosis living in endemic areas and in approximately one-third of renal tissue samples from patients suffering from BEN and urothelial carcinoma.27,40,98 The long-term persistence of such OTA–DNA adducts and even the methodological procedures used to detect them are still a matter of debate and may explain some discrepancy between data reported by different research groups.97 Consequently, in the absence of a specific mutation profile related to OTA and direct DNA damage, some researchers like Turesky proposed an indirect mechanism involving oxidative stress and OTA-mediated cytotoxicity rather than direct genotoxic properties to explain OTA carcinogenicity.104 The so-called ‘AA hypothesis’ in BEN was initially formulated by Ivic in 1970.105 He suggested a possible chronic dietary intoxication from bread made from wheat flour contaminated with seeds of Aristolochia clematidis. Aristolochia is, indeed, a common weed in wheat fields in endemic areas, and seeds are mixed with wheat grain during the annual harvest. This hypothesis seemed to be totally forgotten until the late 1990s. In the first report of the Belgian CHN cases, the attention was already pointed out to the clinical similarities between this nephropathy and the Balkan endemic nephropathy.1 In 1994, Cosyns et al.29 underlined the hypocellular pattern of interstitial fibrosis decreasing from outer to inner cortex, which is a typical histopathological feature shared by both nephropathies. More recently, by using an ultrasensitive, quantitative 32P-postlabeling method, in conjunction with HPLC and mass spectroscopic techniques, Grollman et al.23 reported that dA-aristolactam and dG-aristolactam adducts were found in the DNA from the renal cortex of Croatian patients with BEN and in urothelial tumors from residents of BEN endemic villages. In addition, a predominance of A:T-T:A p53 mutations was detected in urothelial cancers from BEN patients (78%). This ‘signature’ mutation is rarely observed in transitional cell cancers (less than 5%).77 Such mutation profile has never been found for OTA. Kidney International (2008) 74, 158–169 review FD Debelle et al.: Aristolochic acid nephropathy Renal interstitial fibrosis and urothelial carcinoma Similarities Balkan endemic nephropathy (1956) Etiological factors: ochratoxin A highly suspected, aristolochic acid evoked Similarities Chinese herb nephropathy (1992) Etiological factors: aristolochic acid highly suspected, ochratoxin A evoked Aristolochic acid Balkan endemic nephropathy (2007–) Etiological factors: aristolochic acid highly suspected, ochratoxin A evoked Aristolochic acid-related specific DNA adducts in renal tissue Aristolochic acid nephropathy (1996–) Etiological factor: role of aristolochic acid proven Similarities Renal interstitial fibrosis, urothelial carcinoma, and aristolochic acid-related specific DNA adducts Figure 3 | Chinese herb/AAN and Balkan endemic nephropathy: the two faces of Janus? Laboratory experiments performed by introducing the human p53 gene into mouse fibroblasts (Hupki cells) and treating them with AA resulted in the observation of a similar mutational A:T-T:A spectrum.106,107 It was found to be similar to that reported in the H-ras gene of rodents treated with AA.40 Taken together, these clinical, histopathological, epidemiological, and recent toxicological data provide evidence that long-term exposure to AA may be the missing link to elucidate the complex multifactorial etiology of BEN (Figure 3).108 CONCLUSION Often considered harmless, the regular use of herbal products may result in dramatic consequences as demonstrated by the ‘Chinese-herb nephropathy’ tragedy occurring in Belgium in the 1990s. The replacement of one substance (Stephania) by another more toxic compound (Aristolochia) was the cause of the outbreak of progressive renal fibrosis and urothelial carcinoma. Such substitution is probably responsible for a higher incidence of AAN than expected because this practice is common and allowed in traditional medicines that are based on a complex nomenclature using vernacular, not botanical, names. In addition, these products are still available legally in many countries and can be bought via the Internet. Finally, AA is proposed as the environmental causal factor for BEN affecting thousands of people living in the Danube basin. ACKNOWLEDGMENTS This work was supported by grants from the Groupement pour I’Etude, le Traitement et la Réhabilitation Sociale des Insuffisants Kidney International (2008) 74, 158–169 Rénaux Chroniques, the Fonds de la Recherche Scientifique Médicale (Belgium), and the Fondation Erasme (Erasme Hospital, Brussels, Belgium). We are indebted to the medical, nursing, and technical staffs of the nephrology and pathology departments for continuous cooperation. REFERENCES 1. Vanherweghem JL, Depierreux M, Tielemans C et al. Rapidly progressive interstitial renal fibrosis in young women: association with slimming regimen including Chinese herbs. Lancet 1993; 341: 387–391. 2. Depierreux M, Van Damme B, Vanden Houte K et al. Pathologic aspects of a newly described nephropathy related to the prolonged use of Chinese herbs. Am J Kidney Dis 1994; 24: 172–180. 3. Vanhaelen M, Vanhaelen-Fastre R, But P et al. Identification of aristolochic acid in Chinese herbs. Lancet 1994; 343: 174. 4. Cosyns JP, Jadoul M, Squifflet JP et al. Urothelial malignancy in nephropathy due to Chinese herbs. Lancet 1994; 344: 188. 5. Wu KM, Farrelly JG, Upton R et al. Complexities of the herbal nomenclature system in traditional Chinese medicine (TCM): lessons learned from the misuse of Aristolochia-related species and the importance of the pharmaceutical name during botanical drug product development. Phytomedicine 2007; 14: 273–279. 6. Pourrat J, Montastruc JL, Lacombe JL et al. Néphropathie associée à des herbes chinoises—2 cas. Presse Med 1994; 23: 1669. 7. Vanherweghem JL. Misuse of herbal remedies: the case of an outbreak of terminal renal failure in Belgium (Chinese herbs nephropathy). J Altern Complement Med 1998; 4: 9–13. 8. Pena JM, Borras M, Ramos J et al. Rapidly progressive interstitial renal fibrosis due to a chronic intake of a herb (Aristolochia pistolochia) infusion. Nephrol Dial Transplant 1996; 11: 1359–1360. 9. Stengel B, Jones E. Insuffisance rénale terminale associée à la consommation d’herbes chinoises en France. Nephrologie 1998; 19: 15–20. 10. Lord GM, Tagore R, Cook T et al. Nephropathy caused by Chinese herbs in the UK. Lancet 1999; 354: 481–482. 11. Tanaka A, Nishida R, Yokoi H et al. The characteristic pattern of aminoaciduria in patients with aristolochic acid-induced Fanconi syndrome: could iminoaciduria be the hallmark of this syndrome? Clin Nephrol 2000; 54: 198–202. 167 review 12. Tanaka A, Nishida R, Maeda K et al. Chinese herb nephropathy in Japan presents adult-onset Fanconi syndrome: could different components of aristolochic acids cause a different type of Chinese herb nephropathy? Clin Nephrol 2000; 53: 301–306. 13. Meyer MM, Chen TP, Bennett WM. Chinese herb nephropathy. Proc (Bayl Univ Med Cent) 2000; 13: 334–337. 14. Yang CS, Lin CH, Chang SH et al. Rapidly progressive fibrosing interstitial nephritis associated with Chinese herbal drugs. Am J Kidney Dis 2000; 35: 313–318. 15. Krumme B, Endmeir R, Vanhaelen M et al. Reversible Fanconi syndrome after ingestion of a Chinese herbal ‘remedy’ containing aristolochic acid. Nephrol Dial Transplant 2001; 16: 400–402. 16. Chang CH, Wang YM, Yang AH et al. Rapidly progressive interstitial renal fibrosis associated with Chinese herbal medications. Am J Nephrol 2001; 21: 441–448. 17. Cronin AJ, Maidment G, Cook T et al. Aristolochic acid as a causative factor in a case of Chinese herbal nephropathy. Nephrol Dial Transplant 2002; 17: 524–525. 18. Lee S, Lee T, Lee B et al. Fanconi’s syndrome and subsequent progressive renal failure caused by a Chinese herb containing aristolochic acid. Nephrology (Carlton) 2004; 9: 126–129. 19. Lo SH, Mo KL, Wong KS et al. Aristolochic acid nephropathy complicating a patient with focal segmental glomerulosclerosis. Nephrol Dial Transplant 2004; 19: 1913–1915. 20. Laing C, Hamour S, Sheaff M et al. Chinese herbal uropathy and nephropathy. Lancet 2006; 368: 338. 21. Yang L, Li X, Wang H. Possible mechanisms explaining the tendency towards interstitial fibrosis in aristolochic acid-induced acute tubular necrosis. Nephrol Dial Transplant 2007; 22: 445–456. 22. Poon WT, Lai CK, Chan A. Aristolochic acid nephropathy: the Hong Kong perspective. Hong Kong J Nephrol 2007; 9: 7–14. 23. Grollman AP, Shibutani S, Moriya M et al. Aristolochic acid and the etiology of endemic (Balkan) nephropathy. Proc Natl Acad Sci USA 2007; 104: 12129–12134. 24. Zhu M, Phillipson JD. Hong Kong samples of the traditional Chinese medicine ‘Fang ji’ contain aristolochic acid toxins. Int J Pharmacognosy 1996; 34: 283–289. 25. Hashimoto K, Higuchi M, Makino B et al. Quantitative analysis of aristolochic acids, toxic compounds, contained in some medicinal plants. J Ethnopharmacol 1999; 64: 185–189. 26. Vanherweghem JL. Aristolochia sp and chronic interstitial nephropathies in Indians. Lancet 1997; 349: 1399. 27. Stefanovic V, Toncheva D, Atanasova S et al. Etiology of Balkan endemic nephropathy and associated urothelial cancer. Am J Nephrol 2006; 26: 1–11. 28. Gold LS, Slone TH. Aristolochic acid, an herbal carcinogen, sold on the Web after FDA alert. N Engl J Med 2003; 349: 1576–1577. 29. Cosyns JP, Jadoul M, Squifflet JP et al. Chinese herbs nephropathy: a clue to Balkan endemic nephropathy? Kidney Int 1994; 45: 1680–1688. 30. Stefanovic V, Polenakovic MH. Balkan nephropathy. Kidney disease beyond the Balkans? Am J Nephrol 1991; 11: 1–11. 31. Muniz Martinez MC, Nortier J, Vereerstraeten P et al. Progression rate of Chinese herb nephropathy: impact of Aristolochia fangchi ingested dose. Nephrol Dial Transplant 2002; 17: 408–412. 32. Schmeiser HH, Bieler CA, Wiessler M et al. Detection of DNA adducts formed by aristolochic acid in renal tissue from patients with Chinese herbs nephropathy. Cancer Res 1996; 56: 2025–2028. 33. Nortier JL, Muniz Martinez MC, Schmeiser HH et al. Urothelial carcinoma associated with the use of a Chinese herb (Aristolochia fangchi). N Engl J Med 2000; 342: 1686–1692. 34. Cosyns JP, Dehoux JP, Guiot Y et al. Chronic aristolochic acid toxicity in rabbits: a model of Chinese herbs nephropathy? Kidney Int 2001; 59: 2164–2173. 35. Debelle FD, Nortier JL, de Prez EG et al. Aristolochic acids induce chronic renal failure with interstitial fibrosis in salt-depleted rats. J Am Soc Nephrol 2002; 13: 431–436. 36. Lo SH, Wong KS, Arlt VM et al. Detection of Herba Aristolochia mollissemae in a patient with unexplained nephropathy. Am J Kidney Dis 2005; 45: 407–410. 37. Tanaka A, Nishida R, Yoshida T et al. Outbreak of Chinese herb nephropathy in Japan: are there any differences from Belgium? Intern Med 2001; 40: 296–300. 38. Tsai CS, Chen YC, Chen HH et al. An unusual cause of hypokalemic paralysis: aristolochic acid nephropathy with Fanconi syndrome. Am J Med Sci 2005; 330: 153–155. 168 FD Debelle et al.: Aristolochic acid nephropathy 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. Yang SS, Chu P, Lin YF et al. Aristolochic acid-induced Fanconi’s syndrome and nephropathy presenting as hypokalemic paralysis. Am J Kidney Dis 2002; 39: E14. Arlt VM, Stiborova M, Schmeiser HH. Aristolochic acid as a probable human cancer hazard in herbal remedies: a review. Mutagenesis 2002; 17: 265–277. Vanherweghem JL, Debelle FD, Muniz-Martinez MC et al. Aristolochic acid nephropathy after Chinese herbal remedies. In: De Broe ME, Porter GA, Bennett WM, Verpooten GA (eds). Clinical Nephrotoxins, 2nd edn, Kluwer Academic Publishers: Dordrecht, 2003, pp 579–586. van Ypersele de Strihou C, Vanherweghem JL. The tragic paradigm of Chinese herbs nephropathy. Nephrol Dial Transplant 1995; 10: 157–160. Reginster F, Jadoul M, van Ypersele de Strihou C. Chinese herbs nephropathy presentation, natural history and fate after transplantation. Nephrol Dial Transplant 1997; 12: 81–86. Kabanda A, Jadoul M, Lauwerys R et al. Low molecular weight proteinuria in Chinese herbs nephropathy. Kidney Int 1995; 48: 1571–1576. Nortier JL, Deschodt-Lanckman MM, Simon S et al. Proximal tubular injury in Chinese herbs nephropathy: monitoring by neutral endopeptidase enzymuria. Kidney Int 1997; 51: 288–293. Lebeau C, Arlt VM, Schmeiser HH et al. Aristolochic acid impedes endocytosis and induces DNA adducts in proximal tubule cells. Kidney Int 2001; 60: 1332–1342. Vanherweghem JL, Abramowicz D, Tielemans C et al. Effects of steroids on the progression of renal failure in chronic interstitial renal fibrosis: a pilot study in Chinese herbs nephropathy. Am J Kidney Dis 1996; 27: 209–215. Muniz Martinez MC, Nortier J, Vereerstraeten P et al. Steroid therapy in chronic interstitial renal fibrosis: the case of Chinese-herb nephropathy. Nephrol Dial Transplant 2002; 17: 2033–2034. De Broe ME. On a nephrotoxic and carcinogenic slimming regimen. Am J Kidney Dis 1999; 33: 1171–1173. Colson CR, De Greef KE, Duymelinck C et al. Role of serotonin in the development of Chinese herbs nephropathy? Nephrol Dial Transplant 1999; 14(Suppl 4): 16. Debelle F, Nortier J, Arlt VM et al. Effects of dexfenfluramine on aristolochic acid nephrotoxicity in a rat model for Chinese-herb nephropathy. Arch Toxicol 2003; 77: 218–226. Vanherweghem JL. Association of valvular heart disease with Chineseherb nephropathy. Lancet 1997; 350: 1858. Unger P, Nortier J, Muniz Martinez MC et al. High prevalence of fenfluramine-related aortic regurgitation in women with end-stage renal disease secondary to Chinese herb nephropathy. Nephrol Dial Transplant 2003; 18: 906–910. Nortier JL, Vanherweghem JL. For patients taking herbal therapy— lessons from aristolochic acid nephropathy. Nephrol Dial Transplant 2007; 22: 1512–1517. Lewis CJ, Alpert S. Letter to Health Care Professionals—FDA Concerned About Botanical Products, Including Dietary Supplements, Containing Aristolochic Acid. US Food and Drug Administration, Center for Food Safety and Applied Nutrition, Office of Natritional Products, Labeling, and Dietary Supplements. May 31, 2000. http://www.cfsan.fda.gov/ Bdms/ds-botl2.html. US Food and Drug Administration. Dietary supplements: aristolochic acid. http://www.cfsan.fda.gov/Bdms/ds-bot.html. World Health Organization. International Agency for research on cancer: IARC monographs on the evaluation of carcinogenic risks to humans— some traditional herbal medicines, some mycotoxins, naphtalene and styrene. IARC Press, Lyon, France 2002; 82: 118. Zhou J, Xie G, Yan X. Traditional Chinese medicines: Molecular Structures, Natural Sources and Applications, 2nd edn, Ashgate Publishing Ltd: Hampshire, 2003. Huang KC. The Pharmacology of Chinese Herbs. CRC Press Inc.: Boca Raton, 2000. Xinhua. Scalpels come out in TCM debate. Shanghai Daily 2007. www.shanghaidaily.com/sp/article/2007-00710/20071018/ article_334911.htm. Hsieh SC, Huang MF, Lin BS et al. Determination of aristolochic acid in Chinese herbal medicine by capillary electrophoresis with laser-induced fluorescence detection. J Chromatogr A 2006; 1105: 127–134. Trujillo WA, Sorenson WR, La LP et al. Determination of aristolochic acid in botanicals and dietary supplements by liquid chromatography with ultraviolet detection and by liquid chromatography/mass spectrometry: single laboratory validation confirmation. J AOAC Int 2006; 89: 942–959. Mani MK. Chronic renal failure in India. Nephrol Dial Transplant 1993; 8: 684–689. Kidney International (2008) 74, 158–169 review FD Debelle et al.: Aristolochic acid nephropathy 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. Vanherweghem JL, Tielemans C, Simon J et al. Chinese herbs nephropathy and renal pelvic carcinoma. Nephrol Dial Transplant 1995; 10: 270–273. Cosyns JP, Jadoul M, Squifflet JP et al. Urothelial lesions in Chinese-herb nephropathy. Am J Kidney Dis 1999; 33: 1011–1017. Lemy A, Wissing KM, Rorive S et al. Late onset of bladder urothelial carcinoma after renal transplantation for end-stage aristolochic acid nephropathy: a case series with 15-year follow-up. Am J Kidney Dis 2008; 51: 471–477. Nortier JL, Schmeiser HH, Muniz Martinez MC et al. Invasive urothelial carcinoma after exposure to Chinese herbal medicine containing aristolochic acid may occur without severe renal failure. Nephrol Dial Transplant 2003; 18: 426–428. Lord GM, Cook T, Arlt VM et al. Urothelial malignant disease and Chinese herbal nephropathy. Lancet 2001; 358: 1515–1516. Arlt VM, Pfohl-Leszkowicz A, Cosyns J et al. Analyses of DNA adducts formed by ochratoxin A and aristolochic acid in patients with Chinese herbs nephropathy. Mutat Res 2001; 494: 143–150. Stiborova M, Frei E, Hodek P et al. 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. Int J Cancer 2005; 113: 189–197. Stiborova M, Frei E, Wiessler M et al. Human enzymes involved in the metabolic activation of carcinogenic aristolochic acids: evidence for reductive activation by cytochromes P450 1A1 and 1A2. Chem Res Toxicol 2001; 14: 1128–1137. Stiborova M, Frei E, Sopko B 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; 24: 1695–1703. Chan W, Cui L, Xu G et al. Study of the phase I and phase II metabolism of nephrotoxin aristolochic acid by liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom 2006; 20: 1755–1760. Krumbiegel G, Hallensleben J, Mennicke WH et al. Studies on the metabolism of aristolochic acids I and II. Xenobiotica 1987; 17: 981–991. Stiborova M, Frei E, Arlt VM et al. Metabolic activation of carcinogenic aristolochic acid, a risk factor for Balkan endemic nephropathy. Mutat Res 2008; 658: 55–67. Arlt VM, Wiessler M, Schmeiser HH. Using polymerase arrest to detect DNA binding specificity of aristolochic acid in the mouse H-ras gene. Carcinogenesis 2000; 21: 235–242. Arlt VM, Schmeiser HH, Pfeifer GP. Sequence-specific detection of aristolochic acid–DNA adducts in the human p53 gene by terminal transferase-dependent PCR. Carcinogenesis 2001; 22: 133–140. Lord GM, Hollstein M, Arlt VM et al. DNA adducts and p53 mutations in a patient with aristolochic acid-associated nephropathy. Am J Kidney Dis 2004; 43: e11–e17. Mengs U, Lang W, Poch J-A. The carcinogenic action of aristolochic acid in rats. Arch Toxicol 1982; 57: 107–119. Mengs U. On the histopathogenesis of rat forestomach carcinoma caused by aristolochic acid. Arch Toxicol 1983; 52: 209–220. Mengs U. Tumour induction in mice following exposure to aristolochic acid. Arch Toxicol 1988; 61: 504–505. Mengs U. Acute toxicity of aristolochic acid in rodents. Arch Toxicol 1987; 59: 328–331. Mengs U, Stotzem CD. Toxicity of aristolochic acid—a subacute study in male rats. Med Sci Res 1992; 20: 223–224. Mengs U, Stotzem CD. Renal toxicity of aristolochic acid in rats as an example of nephrotoxicity testing in routine toxicology. Arch Toxicol 1993; 67: 307–311. Okada H, Watanabe Y, Inoue T et al. Transgene-derived hepatocyte growth factor attenuates reactive renal fibrosis in aristolochic acid nephrotoxicity. Nephrol Dial Transplant 2003; 18: 2515–2523. Kidney International (2008) 74, 158–169 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. Debelle FD, Nortier JL, Husson CP et al. The renin–angiotensin system blockade does not prevent renal interstitial fibrosis induced by aristolochic acids. Kidney Int 2004; 66: 1815–1825. Sato N, Takahashi D, Chen SM et al. Acute nephrotoxicity of aristolochic acids in mice. J Pharm Pharmacol 2004; 56: 221–229. Pozdzik A, Salmon IJ, Debelle FD et al. Aristolochic acid induces proximal tubule apoptosis and epithelial to mesenchymal transformation. Kidney Int 2008; 73: 595–607. Cosyns JP, Goebbels RM, Liberton V et al. Chinese herbs nephropathyassociated slimming regimen induces tumours in the forestomach but no interstitial nephropathy in rats. Arch Toxicol 1998; 72: 738–743. Balachandran P, Wei F, Lin RC et al. Structure activity relationships of aristolochic acid analogues: toxicity in cultured renal epithelial cells. Kidney Int 2005; 67: 1797–1805. Shibutani S, Dong H, Suzuki N et al. Selective toxicity of aristolochic acids I and II. Drug Metab Dispos 2007; 35: 1217–1222. Lebeau C, Debelle FD, Arlt VM et al. Early proximal tubule injury in experimental aristolochic acid nephropathy: functional and histological studies. Nephrol Dial Transplant 2005; 20: 2321–2332. Liu MC, Maruyama S, Mizuno M et al. The nephrotoxicity of Aristolochia manshuriensis in rats is attributable to its aristolochic acids. Clin Exp Nephrol 2003; 7: 186–194. Hsin YH, Cheng CH, Tzen JT et al. Effect of aristolochic acid on intracellular calcium concentration and its links with apoptosis in renal tubular cells. Apoptosis 2006; 11: 2167–2177. Djukanovic L, Radovanovic Z. Balkan endemic nephropathy. In: De Broe ME, Porter GA, Bennett WM, Verpooten GA (eds). Clinical Nephrotoxins, 2nd edn, Kluwer Academic Publishers: Dordrecht, 2003, pp 587–601. Voice TC, Long DT, Radovanovic Z et al. Critical evaluation of environmental exposure agents suspected in the etiology of Balkan endemic nephropathy. Int J Occup Environ Health 2006; 12: 369–376. Pfohl-Leszkowicz A, Tozlovanu M, Manderville R et al. New molecular and field evidences for the implication of mycotoxins but not aristolochic acid in human nephropathy and urinary tract tumor. Mol Nutr Food Res 2007; 51: 1131–1146. Pfohl-Leszkowicz A, Manderville RA. Ochratoxin A: an overview on toxicity and carcinogenicity in animals and humans. Mol Nutr Food Res 2007; 51: 1192. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Some naturally occurring substances: food items and constituents, heterocyclic aromatic amines and mycotoxins. IARC Monogr Eval Carcinog Risks Hum 1993; 56: 489–521. Brown AL, Odell EW, Mantle PG. DNA ploidy distribution in renal tumours induced in male rats by dietary ochratoxin A. Exp Toxicol Pathol 2007; 59: 85–95. Mantle P, Kulinskaya E, Nestler S. Renal tumourigenesis in male rats in response to chronic dietary ochratoxin A. Food Addit Contam 2005; 22(Suppl 1): 58–64. Mally A, Hard GC, Dekant W. Ochratoxin A as a potential etiologic factor in endemic nephropathy: lessons from toxicity studies in rats. Food Chem Toxicol 2007; 45: 2254–2260. Long DT, Voice TC. Role of exposure analysis in solving the mystery of Balkan endemic nephropathy. Croat Med J 2007; 48: 300–311. Turesky RJ. Perspective: ochratoxin A is not a genotoxic carcinogen. Chem Res Toxicol 2005; 18: 1082–1090. Ivic M. Etiology of endemic nephropathy. Lijec Vjesn 1969; 91: 1273–1281. Feldmeyer N, Schmeiser HH, Muehlbauer KR et al. Further studies with a cell immortalization assay to investigate the mutation signature of aristolochic acid in human p53 sequences. Mutat Res 2006; 608: 163–168. Liu Z, Hergenhahn M, Schmeiser HH et al. Human tumor p53 mutations are selected for in mouse embryonic fibroblasts harboring a humanized p53 gene. Proc Natl Acad Sci USA 2004; 101: 2963–2968. Grollman AP, Jelakovic B. Role of environmental toxins in endemic (Balkan) nephropathy. J Am Soc Nephrol 2007; 18: 2817–2823. 169