1
|
Yu Y, Zhang L, Zhang D, Dai Q, Hou M, Chen M, Gao F, Liu XL. The role of ferroptosis in acute kidney injury: mechanisms and potential therapeutic targets. Mol Cell Biochem 2025; 480:759-784. [PMID: 38943027 DOI: 10.1007/s11010-024-05056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Acute kidney injury (AKI) is one of the most common and severe clinical renal syndromes with high morbidity and mortality. Ferroptosis is a form of programmed cell death (PCD), is characterized by iron overload, reactive oxygen species accumulation, and lipid peroxidation. As ferroptosis has been increasingly studied in recent years, it is closely associated with the pathophysiological process of AKI and provides a target for the treatment of AKI. This review offers a comprehensive overview of the regulatory mechanisms of ferroptosis, summarizes its role in various AKI models, and explores its interaction with other forms of cell death, it also presents research on ferroptosis in AKI progression to other diseases. Additionally, the review highlights methods for detecting and assessing AKI through the lens of ferroptosis and describes potential inhibitors of ferroptosis for AKI treatment. Finally, the review presents a perspective on the future of clinical AKI treatment, aiming to stimulate further research on ferroptosis in AKI.
Collapse
Affiliation(s)
- Yanxin Yu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Lei Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Die Zhang
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Qiangfang Dai
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Mingzheng Hou
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Meini Chen
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Feng Gao
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China
| | - Xiao-Long Liu
- Yan'an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan'an University, Yan'an, China.
| |
Collapse
|
2
|
Liu F, Li Y, Li Y, Wang Z, Li X, Liu Y, Zhao Y. Triggering multiple modalities of cell death via dual-responsive nanomedicines to address the narrow therapeutic window of β-lapachone. J Colloid Interface Sci 2025; 678:915-924. [PMID: 39270391 DOI: 10.1016/j.jcis.2024.09.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
The clinical translation of the anticancer drug β-lapachone (LAP) has been limited by the narrow therapeutic window. Stimuli-responsive anticancer drug delivery systems have gained tremendous attention in recent years to alleviate adverse effects and enhance therapeutic efficacy. Here, we report a dual pH- and enzyme-responsive nanocarrier to address the above issue of LAP. In detail, the epigallocatechin gallate (EGCG) and ferric ions were employed to engineer nanoscale ferric ion-polyphenol complexes where LAP was physically encapsulated therein. The coordination bond between Fe3+ and the catechol moiety of EGCG was sensitive to the low pH, enabling the triggered cargo release in the acidic endosomes/lysosomes. Afterward, the released LAP was activated by the overexpressed NAD(P)H: quinone oxidoreductase 1 (NQO1) and ferroptosis suppressor protein 1 (FSP1) in the tumor cells, followed by the generation of reactive oxygen species (ROS), and the induction of oxidative stress and apoptotic cell death. Meanwhile, EGCG could upregulate gasdermin E (GSDME), and ferric ions were involved in the Fenton reaction. Hence, EGCG and ferric ions could sensitize the toxicity of LAP through the induction of multiple cell death pathways (e.g., pyroptosis, ferroptosis, apoptosis, and necroptosis). The current work enlarged the LAP's therapeutic window via controlled cargo release and vehicle sensitization.
Collapse
Affiliation(s)
- Fang Liu
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Yaru Li
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Yao Li
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Zheng Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| | - Xin Li
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | - Yancheng Liu
- Department of Bone and Soft Tissue Oncology, Tianjin Hospital, Tiajin University, 406 Jiefangnan Road, Hexi District, Tianjin 300211, China.
| | - Yanjun Zhao
- Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, School of Pharmaceutical Science & Technology, Faculty of Medicine, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China.
| |
Collapse
|
3
|
Zhou Y, Cui R, Zhang M, Tang F, Ma X, Wu X. Mitochondrial uptake of aristolactam I plays a critical role in its toxicity. Toxicol Lett 2024; 394:76-91. [PMID: 38428544 DOI: 10.1016/j.toxlet.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
Aristolochic acid I (AAI), a component of aristolochic acids, can be converted to the toxic metabolite Aristolactam I (ALI) in vivo which forms aristolactam-nitrenium with delocalized positive charges. It is widely accepted that delocalized lipophilic cations can accumulate in mitochondria due to the highly negatively charged microenvironment of the mitochondrial matrix, but the uptake of ALI by mitochondria is not known. In this study, the cell uptake and mitochondrial localization of ALI, and its subsequent impact on mitochondrial function were investigated. Results show that ALI can rapidly penetrate HK-2 cells without relying on organic anion transporters 1/3 (OAT1/3). The cellular distribution of ALI was found to align with the observed distribution of a mitochondria-selective dye in HK-2 cells. Furthermore, the cell uptake and mitochondrial uptake of ALI were both inhibited by carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone, which induces mitochondrial membrane depolarization. These results suggest that ALI is selectively taken up by mitochondria. Consequently, mitochondrial dysfunction was observed after treatment with ALI. It should be noted that inhibiting OAT1/3 could result in an increased exposure of ALI in vivo and cause more seriously nephrotoxicity. In conclusion, this research reports the mitochondrial uptake of ALI and provides new insight on potential strategies for protection against AAI-induced nephrotoxicity.
Collapse
Affiliation(s)
- Yan Zhou
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Ruirui Cui
- College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Mingkang Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, China; College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Fabing Tang
- Pathology Department, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaohua Ma
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China
| | - Xin'an Wu
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Gansu Province, China.
| |
Collapse
|
4
|
Oe Y, Kim YC, Sidorenko VS, Zhang H, Kanoo S, Lopez N, Goodluck HA, Crespo-Masip M, Vallon V. SGLT2 inhibitor dapagliflozin protects the kidney in a murine model of Balkan nephropathy. Am J Physiol Renal Physiol 2024; 326:F227-F240. [PMID: 38031729 PMCID: PMC11198975 DOI: 10.1152/ajprenal.00228.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
Proximal tubular uptake of aristolochic acid (AA) forms aristolactam (AL)-DNA adducts, which cause a p53/p21-mediated DNA damage response and acute tubular injury. Recurrent AA exposure causes kidney function loss and fibrosis in humans (Balkan endemic nephropathy) and mice and is a model of (acute kidney injury) AKI to chronic kidney disease (CKD) transition. Inhibitors of the proximal tubule sodium-glucose transporter SGLT2 can protect against CKD progression, but their effect on AA-induced kidney injury remains unknown. C57BL/6J mice (15-wk-old) were administered vehicle or AA every 3 days for 3 wk (10 and 3 mg/kg ip in females and males, respectively). Dapagliflozin (dapa, 0.01 g/kg diet) or vehicle was initiated 7 days prior to AA injections. All dapa effects were sex independent, including a robust glycosuria. Dapa lowered urinary kidney-injury molecule 1 (KIM-1) and albumin (both normalized to creatinine) after the last AA injection and kidney mRNA expression of early DNA damage response markers (p53 and p21) 3 wk later at the study end. Dapa also attenuated AA-induced increases in plasma creatinine as well as AA-induced up-regulation of renal pro-senescence, pro-inflammatory and pro-fibrotic genes, and kidney collagen staining. When assessed 1 day after a single AA injection, dapa pretreatment attenuated AL-DNA adduct formation by 10 and 20% in kidney and liver, respectively, associated with reduced p21 expression. Initiating dapa application after the last AA injection also improved kidney outcome but in a less robust manner. In conclusion, the first evidence is presented that pretreatment with an SGLT2 inhibitor can attenuate the AA-induced DNA damage response and subsequent nephropathy.NEW & NOTEWORTHY Recurrent exposure to aristolochic acid (AA) causes kidney function loss and fibrosis in mice and in humans, e.g., in the form of the endemic Balkan nephropathy. Inhibitors of the proximal tubule sodium-glucose transporter SGLT2 can protect against CKD progression, but their effect on AA-induced kidney injury remains unknown. Here we provide the first evidence in a murine model that pretreatment with an SGLT2 inhibitor can attenuate the AA-induced DNA damage response and subsequent nephropathy.
Collapse
Affiliation(s)
- Yuji Oe
- Department of Medicine, University of California-San Diego, La Jolla, California, United States
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States
| | - Young Chul Kim
- Department of Medicine, University of California-San Diego, La Jolla, California, United States
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States
| | - Viktoriya S Sidorenko
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, United States
| | - Haiyan Zhang
- Department of Pathology, University of California-San Diego, San Diego, California, United States
| | - Sadhana Kanoo
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States
| | - Natalia Lopez
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States
| | - Helen A Goodluck
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States
| | - Maria Crespo-Masip
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States
| | - Volker Vallon
- Department of Medicine, University of California-San Diego, La Jolla, California, United States
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States
| |
Collapse
|
5
|
Renal angiotensin I-converting enzyme-deficient mice are protected against aristolochic acid nephropathy. Pflugers Arch 2023; 475:391-403. [PMID: 36520238 PMCID: PMC9908662 DOI: 10.1007/s00424-022-02779-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
The renal renin-angiotensin system (RAS) is involved in the development of chronic kidney disease. Here, we investigated whether mice with reduced renal angiotensin I-converting enzyme (ACE-/-) are protected against aristolochic acid nephropathy (AAN). To further elucidate potential molecular mechanisms, we assessed the renal abundances of several major RAS components. AAN was induced using aristolochic acid I (AAI). Glomerular filtration rate (GFR) was determined using inulin clearance and renal protein abundances of renin, angiotensinogen, angiotensin I-converting enzyme (ACE) 2, and Mas receptor (Mas) were determined in ACE-/- and C57BL/6J control mice by Western blot analyses. Renal ACE activity was determined using a colorimetric assay and renal angiotensin (Ang) (1-7) concentration was determined by ELISA. GFR was similar in vehicle-treated mice of both strains. AAI decreased GFR in controls but not in ACE-/- mice. Furthermore, AAI decreased renal ACE activity in controls but not in ACE-/- mice. Vehicle-treated ACE-/- mice had significantly higher renal ACE2 and Mas protein abundances than controls. AAI decreased renal ACE2 protein abundance in both strains. Furthermore, AAI increased renal Mas protein abundance, although the latter effect did not reach statistical significance in the ACE-/- mice. Renal Ang(1-7) concentration was similar in vehicle-treated mice of both strains. AAI increased renal Ang(1-7) concentration in the ACE-/- mice but not in the controls. Mice with reduced renal ACE are protected against AAN. Our data suggest that in the face of renal ACE deficiency, AAI may activate the ACE2/Ang(1-7)/Mas axis, which in turn may deploy its reno-protective effects.
Collapse
|
6
|
Ma L, Tang J, Cai G, Chen F, Liu Q, Zhou Z, Zhang S, Liu X, Hou N, Yi W. Structure-based screening and biological validation of the anti-thrombotic drug-dicoumarol as a novel and potent PPARγ-modulating ligand. Bioorg Chem 2022; 129:106191. [DOI: 10.1016/j.bioorg.2022.106191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/17/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022]
|
7
|
Feng C, Anger EE, Zhang X, Su S, Su C, Zhao S, Yu F, Li J. Protective Effects of Mitochondrial Uncoupling Protein 2 against Aristolochic Acid I-Induced Toxicity in HK-2 Cells. Int J Mol Sci 2022; 23:ijms23073674. [PMID: 35409033 PMCID: PMC8998172 DOI: 10.3390/ijms23073674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 02/04/2023] Open
Abstract
Aristolochic acid I (AA I) is one of the most abundant and toxic aristolochic acids that is reported to cause Aristolochic acid nephropathy (AAN). This paper was designed to assess whether mitochondrial Uncoupling Protein 2 (UCP2), which plays an antioxidative and antiapoptotic role, could protect human renal proximal tubular epithelial (HK-2) cells from toxicity induced by AA I. In this study, HK-2 cells were treated with different concentrations of AA I with or without UCP2 inhibitor (genipin). To upregulate the expression of UCP2 in HK-2 cells, UCP2-DNA transfection was performed. The cell viability was evaluated by colorimetric method using MTT. A series of related biological events such as Reactive Oxygen Species (ROS), Glutathione peroxidase (GSH-Px), and Malondialdehyde (MDA) were evaluated. The results showed that the cytotoxicity of AA I with genipin group was much higher than that of AA I alone. Genipin dramatically boosted oxidative stress and exacerbated AA I-induced apoptosis. Furthermore, the increased expression of UCP2 can reduce the toxicity of AA I on HK-2 cells and upregulation of UCP2 expression can reduce AA I-induced oxidative stress and apoptosis. In conclusion, UCP2 might be a potential target for alleviating AA I-induced nephrotoxicity.
Collapse
|
8
|
Rodriguez-Esteban R. The speed of information propagation in the scientific network distorts biomedical research. PeerJ 2022; 10:e12764. [PMID: 35070506 PMCID: PMC8759377 DOI: 10.7717/peerj.12764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/17/2021] [Indexed: 01/07/2023] Open
Abstract
Delays in the propagation of scientific discoveries across scientific communities have been an oft-maligned feature of scientific research for introducing a bias towards knowledge that is produced within a scientist's closest community. The vastness of the scientific literature has been commonly blamed for this phenomenon, despite recent improvements in information retrieval and text mining. Its actual negative impact on scientific progress, however, has never been quantified. This analysis attempts to do so by exploring its effects on biomedical discovery, particularly in the discovery of relations between diseases, genes and chemical compounds. Results indicate that the probability that two scientific facts will enable the discovery of a new fact depends on how far apart these two facts were originally within the scientific landscape. In particular, the probability decreases exponentially with the citation distance. Thus, the direction of scientific progress is distorted based on the location in which each scientific fact is published, representing a path-dependent bias in which originally closely-located discoveries drive the sequence of future discoveries. To counter this bias, scientists should open the scope of their scientific work with modern information retrieval and extraction approaches.
Collapse
Affiliation(s)
- Raul Rodriguez-Esteban
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
9
|
Liu YZ, Lu HL, Qi XM, Xing GZ, Wang X, Yu P, Liu L, Yang FF, Ding XL, Zhang ZA, Deng ZP, Gong LK, Ren J. Aristolochic acid I promoted clonal expansion but did not induce hepatocellular carcinoma in adult rats. Acta Pharmacol Sin 2021; 42:2094-2105. [PMID: 33686245 PMCID: PMC8633323 DOI: 10.1038/s41401-021-00622-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/06/2021] [Indexed: 12/31/2022]
Abstract
Aristolochic acid I (AAI) is a well-known nephrotoxic carcinogen, which is currently reported to be also associated with hepatocellular carcinoma (HCC). Whether AAI is a direct hepatocarcinogen remains controversial. In this study we investigated the association between AAI exposure and HCC in adult rats using a sensitive rat liver bioassay with several cofactors. Formation of glutathione S-transferase placental form-positive (GST-P+) foci was used as the marker for preneoplastic lesions/clonal expansion. We first conducted a medium-term (8 weeks) study to investigate whether AAI had any tumor-initiating or -promoting activity. Then a long-term (52 weeks) study was conducted to determine whether AAI can directly induce HCC. We showed that oral administration of single dose of AAI (20, 50, or 100 mg/kg) in combination with partial hepatectomy (PH) to stimulate liver proliferation did not induce typical GST-P+ foci in liver. In the 8-week study, only high dose of AAI (10 mg · kg-1 · d-1, 5 days a week for 6 weeks) in combination with PH significantly increased the number and area of GST-P+ foci initiated by diethylnitrosamine (DEN) in liver. Similarly, only high dose of AAI (10 mg· kg-1· d-1, 5 days a week for 52 weeks) in combination with PH significantly increased the number and area of hepatic GST-P+ foci in the 52-week study. No any nodules or HCC were observed in liver of any AAI-treated groups. In contrast, long-term administration of AAI (0.1, 1, 10 mg· kg-1· d-1) time- and dose-dependently caused death due to the occurrence of cancers in the forestomach, intestine, and/or kidney. Besides, AAI-DNA adducts accumulated in the forestomach, kidney, and liver in a time- and dose-dependent manner. Taken together, AAI promotes clonal expansion only in the high-dose group but did not induce any nodules or HCC in liver of adult rats till their deaths caused by cancers developed in the forestomach, intestine, and/or kidney. Findings from our animal studies will pave the way for further large-scale epidemiological investigation of the associations between AA and HCC.
Collapse
Affiliation(s)
- Yong-Zhen Liu
- Center for Drug Safety Evaluation and Research, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Heng-Lei Lu
- Center for Drug Safety Evaluation and Research, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin-Ming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guo-Zhen Xing
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xin Wang
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Pan Yu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu Liu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Fang-Fang Yang
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Lan Ding
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ze-An Zhang
- Center for Drug Safety Evaluation and Research, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhong-Ping Deng
- Center for Drug Safety Evaluation and Research, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Li-Kun Gong
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Institutes of Drug Discovery and Development, CAS, Zhongshan, 528400, China.
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
10
|
Maier L, Goemans CV, Wirbel J, Kuhn M, Eberl C, Pruteanu M, Müller P, Garcia-Santamarina S, Cacace E, Zhang B, Gekeler C, Banerjee T, Anderson EE, Milanese A, Löber U, Forslund SK, Patil KR, Zimmermann M, Stecher B, Zeller G, Bork P, Typas A. Unravelling the collateral damage of antibiotics on gut bacteria. Nature 2021; 599:120-124. [PMID: 34646011 PMCID: PMC7612847 DOI: 10.1038/s41586-021-03986-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
Antibiotics are used to fight pathogens but also target commensal bacteria, disturbing the composition of gut microbiota and causing dysbiosis and disease1. Despite this well-known collateral damage, the activity spectrum of different antibiotic classes on gut bacteria remains poorly characterized. Here we characterize further 144 antibiotics from a previous screen of more than 1,000 drugs on 38 representative human gut microbiome species2. Antibiotic classes exhibited distinct inhibition spectra, including generation dependence for quinolones and phylogeny independence for β-lactams. Macrolides and tetracyclines, both prototypic bacteriostatic protein synthesis inhibitors, inhibited nearly all commensals tested but also killed several species. Killed bacteria were more readily eliminated from in vitro communities than those inhibited. This species-specific killing activity challenges the long-standing distinction between bactericidal and bacteriostatic antibiotic classes and provides a possible explanation for the strong effect of macrolides on animal3-5 and human6,7 gut microbiomes. To mitigate this collateral damage of macrolides and tetracyclines, we screened for drugs that specifically antagonized the antibiotic activity against abundant Bacteroides species but not against relevant pathogens. Such antidotes selectively protected Bacteroides species from erythromycin treatment in human-stool-derived communities and gnotobiotic mice. These findings illluminate the activity spectra of antibiotics in commensal bacteria and suggest strategies to circumvent their adverse effects on the gut microbiota.
Collapse
Affiliation(s)
- Lisa Maier
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany.
| | - Camille V Goemans
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jakob Wirbel
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Kuhn
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Claudia Eberl
- Max-von-Pettenkofer Institute, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU Munich, Munich, Germany
| | - Mihaela Pruteanu
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Biology, Humboldt University Berlin, Berlin, Germany
| | - Patrick Müller
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
| | | | - Elisabetta Cacace
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Boyao Zhang
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Cordula Gekeler
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
| | - Tisya Banerjee
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Chemistry, TU Munich, Munich, Germany
| | - Exene Erin Anderson
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- NYU School of Medicine, New York, NY, USA
| | - Alessio Milanese
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Ulrike Löber
- Experimental and Clinical Research Center, a cooperation of Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Sofia K Forslund
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Experimental and Clinical Research Center, a cooperation of Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Kiran Raosaheb Patil
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- The Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Michael Zimmermann
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Bärbel Stecher
- Max-von-Pettenkofer Institute, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU Munich, Munich, Germany
| | - Georg Zeller
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, South Korea
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Athanasios Typas
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
11
|
Wang Y, Wang Z, Wu Z, Chen M, Dong D, Yu P, Lu D, Wu B. Involvement of REV-ERBα dysregulation and ferroptosis in aristolochic acid I-induced renal injury. Biochem Pharmacol 2021; 193:114807. [PMID: 34673015 DOI: 10.1016/j.bcp.2021.114807] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/23/2021] [Accepted: 10/11/2021] [Indexed: 10/20/2022]
Abstract
The molecular events underlying aristolochic acid (AA) nephropathy are poorly understood, and specific therapies for treatment of AA nephropathy are still lacking. Here we aimed to investigate a potential role of REV-ERBα and ferroptosis in renal injury induced by aristolochic acid I (AAI), a typical AA. The regulatory effects of REV-ERBα on AAI-induced renal injury were determined using kidney-specific Rev-erbα knockout mice. Ferroptosis was assessed based on measurements of iron, GSH, and GPX4. Targeted antagonism of REV-ERBα to alleviate AAI-induced renal injury and ferroptosis was assessed using the small molecule antagonist SR8278. mRNAs and proteins were quantified by qPCR and Western blotting, respectively. We first showed that REV-ERBα was upregulated and its target BMAL1 was downregulated in the kidney of mice with AAI nephropathy. Upregulation of REV-ERBα protein was confirmed in aristolactam I (ALI, a nephrotoxic metabolite of AAI)-treated mRTECs. We also observed enhanced ferroptosis (known to be regulated by REV-ERBα) in mice with AAI nephropathy and in ALI-treated mRTECs. Kidney-specific knockout of Rev-erbα reduced the sensitivity of mice to AAI-induced ferroptosis and renal injury. Furthermore, knockdown of Rev-erbα by siRNA or SR8278 (a REV-ERBα antagonist) treatment attenuated ALI-induced ferroptosis in mRTECs. Moreover, REV-ERBα antagonism by SR8278 alleviated ferroptosis and renal injury caused by AAI in mice. In conclusion, we identify REV-ERBα as a regulator of AAI-induced renal injury via promoting ferroptosis. Targeting REV-ERBα may represent a promising approach for management of AAI nephropathy.
Collapse
Affiliation(s)
- Yi Wang
- College of Pharmacy, Jinan University, Guangzhou, China; Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhigang Wang
- Department of Intensive Care Unit, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhengping Wu
- School of Medicine, Yichun University, Yichun, Jiangxi, China
| | - Menglin Chen
- College of Pharmacy, Jinan University, Guangzhou, China; Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dong Dong
- School of Medicine, Jinan University, Guangzhou, China
| | - Pei Yu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Danyi Lu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Baojian Wu
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
12
|
Bárta F, Dedíková A, Bebová M, Dušková Š, Mráz J, Schmeiser HH, Arlt VM, Hodek P, Stiborová M. Co-Exposure to Aristolochic Acids I and II Increases DNA Adduct Formation Responsible for Aristolochic Acid I-Mediated Carcinogenicity in Rats. Int J Mol Sci 2021; 22:ijms221910479. [PMID: 34638820 PMCID: PMC8509051 DOI: 10.3390/ijms221910479] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
The plant extract aristolochic acid (AA), containing aristolochic acids I (AAI) and II (AAII) as major components, causes aristolochic acid nephropathy (AAN) and Balkan endemic nephropathy (BEN), unique renal diseases associated with upper urothelial cancer. Recently (Chemical Research in Toxicology 33(11), 2804–2818, 2020), we showed that the in vivo metabolism of AAI and AAII in Wistar rats is influenced by their co-exposure (i.e., AAI/AAII mixture). Using the same rat model, we investigated how exposure to the AAI/AAII mixture can influence AAI and AAII DNA adduct formation (i.e., AA-mediated genotoxicity). Using 32P-postlabelling, we found that AA-DNA adduct formation was increased in the livers and kidneys of rats treated with AAI/AAII mixture compared to rats treated with AAI or AAII alone. Measuring the activity of enzymes involved in AA metabolism, we showed that enhanced AA-DNA adduct formation might be caused partially by both decreased AAI detoxification as a result of hepatic CYP2C11 inhibition during treatment with AAI/AAII mixture and by hepatic or renal NQO1 induction, the key enzyme predominantly activating AA to DNA adducts. Moreover, our results indicate that AAII might act as an inhibitor of AAI detoxification in vivo. Consequently, higher amounts of AAI might remain in liver and kidney tissues, which can be reductively activated, resulting in enhanced AAI DNA adduct formation. Collectively, these results indicate that AAII present in the plant extract AA enhances the genotoxic properties of AAI (i.e., AAI DNA adduct formation). As patients suffering from AAN and BEN are always exposed to the plant extract (i.e., AAI/AAII mixture), our findings are crucial to better understanding host factors critical for AAN- and BEN-associated urothelial malignancy.
Collapse
Affiliation(s)
- František Bárta
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic; (F.B.); (A.D.); (M.B.); (P.H.); (M.S.)
| | - Alena Dedíková
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic; (F.B.); (A.D.); (M.B.); (P.H.); (M.S.)
| | - Michaela Bebová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic; (F.B.); (A.D.); (M.B.); (P.H.); (M.S.)
| | - Šárka Dušková
- Centre of Occupational Health, National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic; (Š.D.); (J.M.)
| | - Jaroslav Mráz
- Centre of Occupational Health, National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic; (Š.D.); (J.M.)
| | - Heinz H. Schmeiser
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany;
| | - Volker M. Arlt
- Department of Analytical, Environmental and Forensic Sciences Division, King’s College London, 150 Stamford Street, London SE1 9NH, UK
- Toxicology Department, GAB Consulting GmbH, Heinrich-Fuchs-Str. 96, 69126 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-432018-0
| | - Petr Hodek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic; (F.B.); (A.D.); (M.B.); (P.H.); (M.S.)
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic; (F.B.); (A.D.); (M.B.); (P.H.); (M.S.)
| |
Collapse
|
13
|
Bellamri M, Brandt K, Brown CV, Wu MT, Turesky RJ. Cytotoxicity and genotoxicity of the carcinogen aristolochic acid I (AA-I) in human bladder RT4 cells. Arch Toxicol 2021; 95:2189-2199. [PMID: 33938965 PMCID: PMC8284306 DOI: 10.1007/s00204-021-03059-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/21/2021] [Indexed: 11/28/2022]
Abstract
Aristolochic acid (AA-I) induces upper urothelial tract cancer (UUTC) and bladder cancer (BC) in humans. AA-I forms the 7-(2'-deoxyadenosin-N6-yl)aristolactam I (dA-AL-I) adduct, which induces multiple A:T-to-T:A transversion mutations in TP53 of AA-I exposed UTUC patients. This mutation is rarely reported in TP53 of other transitional cell carcinomas and thus recognized as an AA-I mutational signature. A:T-to-T:A transversion mutations were recently detected in bladder tumors of patients in Asia with known AA-I-exposure, implying that AA-I contributes to BC. Mechanistic studies on AA-I genotoxicity have not been reported in human bladder. In this study, we examined AA-I DNA adduct formation and mechanisms of toxicity in the human RT4 bladder cell line. The biological potencies of AA-I were compared to 4-aminobiphenyl, a recognized human bladder carcinogen, and several structurally related carcinogenic heterocyclic aromatic amines (HAA), which are present in urine of smokers and omnivores. AA-I (0.05-10 µM) induced a concentration- and time-dependent cytotoxicity. AA-I (100 nM) DNA adduct formation occurred at over a thousand higher levels than the principal DNA adducts formed with 4-ABP or HAAs (1 µM). dA-AL-I adduct formation was detected down to a 1 nM concentration. Studies with selective chemical inhibitors provided evidence that NQO1 is the major enzyme involved in AA-I bio-activation in RT4 cells, whereas CYP1A1, another enzyme implicated in AA-I toxicity, had a lesser role in bio-activation or detoxification of AA-I. AA-I DNA damage also induced genotoxic stress leading to p53-dependent apoptosis. These biochemical data support the human mutation data and a role for AA-I in BC.
Collapse
Affiliation(s)
- Medjda Bellamri
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, MN, 55455, USA
| | - Kyle Brandt
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, MN, 55455, USA
| | - Christina V Brown
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, MN, 55455, USA
| | - Ming-Tsang Wu
- Department of Environmental and Occupational Medicine, Kaohsiung Medical University, CS Building, 100 Shih-Chuan 1st Road, Kaohsiung, Taiwan
| | - Robert J Turesky
- Masonic Cancer Center and Department of Medicinal Chemistry, Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th Street, Minneapolis, MN, 55455, USA.
| |
Collapse
|
14
|
Abdullah R, Wesseling S, Spenkelink B, Louisse J, Punt A, Rietjens IM. Defining in vivo dose-response curves for kidney DNA adduct formation of aristolochic acid I in rat, mouse and human by an in vitro and physiologically based kinetic modeling approach. J Appl Toxicol 2020; 40:1647-1660. [PMID: 33034907 PMCID: PMC7689901 DOI: 10.1002/jat.4024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/14/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022]
Abstract
Aristolochic acid I (AAI) is a well-known genotoxic kidney carcinogen. Metabolic conversion of AAI into the DNA-reactive aristolactam-nitrenium ion is involved in the mode of action of tumor formation. This study aims to predict in vivo AAI-DNA adduct formation in the kidney of rat, mouse and human by translating the in vitro concentration-response curves for AAI-DNA adduct formation to the in vivo situation using physiologically based kinetic (PBK) modeling-based reverse dosimetry. DNA adduct formation in kidney proximal tubular LLC-PK1 cells exposed to AAI was quantified by liquid chromatography-electrospray ionization-tandem mass spectrometry. Subsequently, the in vitro concentration-response curves were converted to predicted in vivo dose-response curves in rat, mouse and human kidney using PBK models. Results obtained revealed a dose-dependent increase in AAI-DNA adduct formation in the rat, mouse and human kidney and the predicted DNA adduct levels were generally within an order of magnitude compared with values reported in the literature. It is concluded that the combined in vitro PBK modeling approach provides a novel way to define in vivo dose-response curves for kidney DNA adduct formation in rat, mouse and human and contributes to the reduction, refinement and replacement of animal testing.
Collapse
Affiliation(s)
- Rozaini Abdullah
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
- Department of Environmental & Occupational Health, Faculty of Medicine and Health SciencesUniversiti Putra MalaysiaSelangorMalaysia
| | | | - Bert Spenkelink
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
| | - Jochem Louisse
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
| | - Ans Punt
- Division of ToxicologyWageningen UniversityWageningenThe Netherlands
| | | |
Collapse
|
15
|
Dedı Ková A, Bárta F, Martínek V, Kotalík K, Dušková Š, Mráz J, Arlt VM, Stiborová M, Hodek P. In Vivo Metabolism of Aristolochic Acid I and II in Rats Is Influenced by Their Coexposure. Chem Res Toxicol 2020; 33:2804-2818. [PMID: 32894017 DOI: 10.1021/acs.chemrestox.0c00198] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The plant extract aristolochic acid (AA), containing aristolochic acid I (AAI) and II (AAII) as major components, causes aristolochic acid nephropathy and Balkan endemic nephropathy, unique renal diseases associated with upper urothelial cancer. Differences in the metabolic activation and detoxification of AAI and AAII and their effects on the metabolism of AAI/AAII mixture in the plant extract might be of great importance for an individual's susceptibility in the development of AA-mediated nephropathies and malignancies. Here, we investigated in vivo metabolism of AAI and AAII after ip administration to Wistar rats as individual compounds and as AAI/AAII mixture using high performance liquid chromatography/electrospray ionization mass spectrometry. Experimental findings were supported by theoretical calculations using density functional theory. We found that exposure to AAI/AAII mixture affected the generation of their oxidative and reductive metabolites formed during Phase I biotransformation and excreted in rat urine. Several Phase II metabolites of AAI and AAII found in the urine of exposed rats were also analyzed. Our results indicate that AAI is more efficiently metabolized in rats in vivo than AAII. Whereas AAI is predominantly oxidized during in vivo metabolism, its reduction is the minor metabolic pathway. In contrast, AAII is mainly metabolized by reduction. The oxidative reaction only occurs if aristolactam II, the major reductive metabolite of AAII, is enzymatically hydroxylated, forming aristolactam Ia. In AAI/AAII mixture, the metabolism of AAI and AAII is influenced by the presence of both AAs. For instance, the reductive metabolism of AAI is increased in the presence of AAII while the presence of AAI decreased the reductive metabolism of AAII. These results suggest that increased bioactivation of AAI in the presence of AAII also leads to increased AAI genotoxicity, which may critically impact AAI-mediated carcinogenesis. Future studies are needed to explain the underlying mechanism(s) for this phenomenon.
Collapse
Affiliation(s)
- Alena Dedı Ková
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 12840 Prague, Czech Republic
| | - František Bárta
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 12840 Prague, Czech Republic
| | - Václav Martínek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 12840 Prague, Czech Republic
| | - Kevin Kotalík
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 12840 Prague, Czech Republic
| | - Šárka Dušková
- Centre of Occupational Health, National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic
| | - Jaroslav Mráz
- Centre of Occupational Health, National Institute of Public Health, Šrobárova 48, 100 42 Prague 10, Czech Republic
| | - Volker Manfred Arlt
- Department of Analytical, Environmental and Forensic Sciences Division, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 12840 Prague, Czech Republic
| | - Petr Hodek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 12840 Prague, Czech Republic
| |
Collapse
|
16
|
Sun C, Zhao W, Wang X, Sun Y, Chen X. A pharmacological review of dicoumarol: An old natural anticoagulant agent. Pharmacol Res 2020; 160:105193. [PMID: 32911072 DOI: 10.1016/j.phrs.2020.105193] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022]
Abstract
Dicoumarol is an oral anticoagulant agent prescribed in clinical for decades. It is a natural hydroxycoumarin discovered from the spoilage of Melilotus officinalis (L.) Pall and is originally discovered as a rodenticide. Due to its structural similarity to that of vitamin K, it significantly inhibits vitamin K epoxide reductase and acts as a vitamin K antagonist. Dicoumarol is mainly used as an anticoagulant to prevent thrombogenesis and to cure vascular thrombosis. Other biological activities besides anticoagulants such as anticancer, antimicrobial, antiviral, etc., have also been documented. The side effects of dicoumarol raise safety concerns for clinical application. In this review, the physicochemical property, the pharmacological activities, the side effects, and the pharmacokinetics of dicoumarol were summarized, aiming to provide a whole picture of the "old" anticoagulant.
Collapse
Affiliation(s)
- Chong Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Wenwen Zhao
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Xumei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Yinxiang Sun
- Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, China.
| | - Xiuping Chen
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| |
Collapse
|
17
|
Li XL, Guo XQ, Wang HR, Chen T, Mei N. Aristolochic Acid-Induced Genotoxicity and Toxicogenomic Changes in Rodents. WORLD JOURNAL OF TRADITIONAL CHINESE MEDICINE 2020; 6:12-25. [PMID: 32258091 PMCID: PMC7110418 DOI: 10.4103/wjtcm.wjtcm_33_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aristolochic acid (AA) is a group of structurally related nitrophenanthrene carboxylic acids found in many plants that are widely used by many cultures as traditional herbal medicines. AA is a causative agent for Chinese herbs nephropathy, a term replaced later by AA nephropathy. Evidence indicates that AA is nephrotoxic, genotoxic, and carcinogenic in humans; and it also induces tumors in the forestomach, kidney, renal pelvis, urinary bladder, and lung of rats and mice. Therefore, plants containing AA have been classified as carcinogenic to humans (Group 1) by the International Agency for Research on Cancer. In our laboratories, we have conducted a series of genotoxicity and toxicogenomic studies in the rats exposed to AA of 0.1–10 mg/kg for 12 weeks. Our results demonstrated that AA treatments induced DNA adducts and mutations in the kidney, liver, and spleen of rats, as well as significant alteration of gene expression in both its target and nontarget tissues. AA treatments altered mutagenesis- or carcinogenesis-related microRNA expression in rat kidney and resulted in significant changes in protein expression profiling. We also applied benchmark dose (BMD) modeling to the 3-month AA-induced genotoxicity data. The obtained BMDL10 (the lower 95% confidence interval of the BMD10 that is a 10% increase over the background level) for AA-induced mutations in the kidney of rats was about 7 μg/kg body weight per day. This review constitutes an overview of our investigations on AA-induced genotoxicity and toxicogenomic changes including gene expression, microRNA expression, and proteomics; and presents updated information focused on AA-induced genotoxicity in rodents.
Collapse
Affiliation(s)
- Xi-Lin Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| | - Xiao-Qing Guo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| | - Hai-Rong Wang
- Tianjin Center for New Drug Safety Assessment and Research, Tianjin, China
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR, USA
| |
Collapse
|
18
|
Anger EE, Yu F, Li J. Aristolochic Acid-Induced Nephrotoxicity: Molecular Mechanisms and Potential Protective Approaches. Int J Mol Sci 2020; 21:E1157. [PMID: 32050524 PMCID: PMC7043226 DOI: 10.3390/ijms21031157] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/21/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Aristolochic acid (AA) is a generic term that describes a group of structurally related compounds found in the Aristolochiaceae plants family. These plants have been used for decades to treat various diseases. However, the consumption of products derived from plants containing AA has been associated with the development of nephropathy and carcinoma, mainly the upper urothelial carcinoma (UUC). AA has been identified as the causative agent of these pathologies. Several studies on mechanisms of action of AA nephrotoxicity have been conducted, but the comprehensive mechanisms of AA-induced nephrotoxicity and carcinogenesis have not yet fully been elucidated, and therapeutic measures are therefore limited. This review aimed to summarize the molecular mechanisms underlying AA-induced nephrotoxicity with an emphasis on its enzymatic bioactivation, and to discuss some agents and their modes of action to reduce AA nephrotoxicity. By addressing these two aspects, including mechanisms of action of AA nephrotoxicity and protective approaches against the latter, and especially by covering the whole range of these protective agents, this review provides an overview on AA nephrotoxicity. It also reports new knowledge on mechanisms of AA-mediated nephrotoxicity recently published in the literature and provides suggestions for future studies.
Collapse
Affiliation(s)
| | | | - Ji Li
- Department of Clinical Pharmacy, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (E.E.A.); (F.Y.)
| |
Collapse
|
19
|
Xu C, Song R, Lu P, Chen J, Zhou Y, Shen G, Jiang M, Zhang W. A pH-Responsive Charge-Reversal Drug Delivery System with Tumor-Specific Drug Release and ROS Generation for Cancer Therapy. Int J Nanomedicine 2020; 15:65-80. [PMID: 32021165 PMCID: PMC6955620 DOI: 10.2147/ijn.s230237] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/10/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Poor cell uptake and incomplete intracellular drug release are the two major challenges for polymeric prodrug-based drug delivery systems (PPDDSs) in cancer treatment. METHODS Herein, a PPDDS with pH-induced surface charge-reversal and reactive oxygen species (ROS) amplification for ROS-triggered self-accelerating drug release was developed, which was formed by encapsulating a ROS generation agent (vitamin K3 (VK3)) in pH/ROS dual-sensitive polymetric prodrug (PEG-b-P(LL-g-TK-PTX)-(LL-g-DMA)) based micelle nanoparticles (denoted as PVD-NPs). RESULTS The surface charge of the PVD-NPs can change from negative to positive for enhanced cell uptake in response to tumor extracellular acidity pH. After internalization by cancer cells, PVD-NPs demonstrate dual drug release in response to intracellular ROS-rich conditions. In addition, the released VK3 can produce ROS under the catalysis by NAD(P)H:quinone oxidoreductase-1, which facilitates tumor-specific ROS amplification and drug release selectively in cancer cells to enhance chemotherapy. CONCLUSION Both in vitro and in vivo experiments demonstrated that the PVD-NPs showed significant antitumor activity in human prostate cancer.
Collapse
Affiliation(s)
- Chen Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing210029, People’s Republic of China
- Department of Urology, Affiliated Wujiang Hospital of Nantong Univerisity, Suzhou215200, People’s Republic of China
| | - Rijin Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Pei Lu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Jianchun Chen
- Department of Urology, Affiliated Wujiang Hospital of Nantong Univerisity, Suzhou215200, People’s Republic of China
| | - Yongqiang Zhou
- Department of Urology, Affiliated Wujiang Hospital of Nantong Univerisity, Suzhou215200, People’s Republic of China
| | - Gang Shen
- Department of Urology, Affiliated Wujiang Hospital of Nantong Univerisity, Suzhou215200, People’s Republic of China
| | - Minjun Jiang
- Department of Urology, Affiliated Wujiang Hospital of Nantong Univerisity, Suzhou215200, People’s Republic of China
| | - Wei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing210029, People’s Republic of China
| |
Collapse
|
20
|
Sidorenko VS. Biotransformation and Toxicities of Aristolochic Acids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1241:139-166. [PMID: 32383120 DOI: 10.1007/978-3-030-41283-8_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Environmental and iatrogenic exposures contribute significantly to human diseases, including cancer. The list of known human carcinogens has recently been extended by the addition of aristolochic acids (AAs). AAs occur primarily in Aristolochia herbs, which are used extensively in folk medicines, including Traditional Chinese Medicine. Ingestion of AAs results in chronic renal disease and cancer. Despite importation bans imposed by certain countries, herbal remedies containing AAs are readily available for purchase through the internet. With recent advancements in mass spectrometry, next generation sequencing, and the development of integrated organs-on-chips, our knowledge of cancers associated with AA exposure, and of the mechanisms involved in AA toxicities, has significantly improved. DNA adduction plays a central role in AA-induced cancers; however, significant gaps remain in our knowledge as to how cellular enzymes promote activation of AAs and how the reactive species selectively bind to DNA and kidney proteins. In this review, I describe pathways for AAs biotransformation, adduction, and mutagenesis, emphasizing novel methods and ideas contributing to our present understanding of AA toxicities in humans.
Collapse
Affiliation(s)
- Viktoriya S Sidorenko
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
21
|
NQO1 potentiates apoptosis evasion and upregulates XIAP via inhibiting proteasome-mediated degradation SIRT6 in hepatocellular carcinoma. Cell Commun Signal 2019; 17:168. [PMID: 31842909 PMCID: PMC6915971 DOI: 10.1186/s12964-019-0491-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/26/2019] [Indexed: 12/11/2022] Open
Abstract
Background Our previous study has demonstrated that NAD(P)H: quinone oxidoreductase 1 (NQO1) is significantly upregulated in human liver cancer where it potentiates the apoptosis evasion of liver cancer cell. However, the underlying mechanisms of the oncogenic function of NQO1 in HCC have not been fully elucidated. Methods Expression of NQO1, SIRT6, AKT and X-linked inhibitor of apoptosis protein (XIAP) protein were measured by western blotting and immunohistochemistry. Additionally, the interaction between NQO1 and potential proteins were determined by immunoprecipitation assays. Furthermore, the effect of NQO1 and SIRT6 on tumor growth was determined in cell model and orthotopic tumor implantation model. Results We found that NQO1 overexpression in HCC enhanced SIRT6 protein stability via inhibiting ubiquitin-mediated 26S proteasome degradation. High level of SIRT6 reduced acetylation of AKT which resulted in increased phosphorylation and activity of AKT. Activated AKT subsequently phosphorylated anti-apoptotic protein XIAP at Ser87 which determined its protein stability. Reintroduction of SIRT6 or AKT efficiently rescued NQO1 knock-out-mediated inhibition of growth and induction of apoptosis. In orthotopic mouse model, NQO1 knock-out inhibited tumor growth and induced apoptosis while this effect was effectively rescued by SIRT6 overexpression or MG132 treatment partially. Conclusions Collectively, these results reveal an oncogenic function of NQO1 in sustaining HCC cell proliferation through SIRT6/AKT/XIAP signaling pathway.
Collapse
|
22
|
Wen B, Gorycki P. Bioactivation of herbal constituents: mechanisms and toxicological relevance. Drug Metab Rev 2019; 51:453-497. [DOI: 10.1080/03602532.2019.1655570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bo Wen
- Department of Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Collegeville, PA, USA
| | - Peter Gorycki
- Department of Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Collegeville, PA, USA
| |
Collapse
|
23
|
Aristolochic Acids: Newly Identified Exposure Pathways of this Class of Environmental and Food-Borne Contaminants and its Potential Link to Chronic Kidney Diseases. TOXICS 2019; 7:toxics7010014. [PMID: 30893813 PMCID: PMC6468885 DOI: 10.3390/toxics7010014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 11/17/2022]
Abstract
Aristolochic acids (AAs) are nitrophenanthrene carboxylic acids naturally produced by Aristolochia plants. These plants were widely used to prepare herbal remedies until AAs were observed to be highly nephrotoxic and carcinogenic to humans. Although the use of AA-containing Aristolochia plants in herbal medicine is prohibited in countries worldwide, emerging evidence nevertheless has indicated that AAs are the causative agents of Balkan endemic nephropathy (BEN), an environmentally derived disease threatening numerous residents of rural farming villages along the Danube River in countries of the Balkan Peninsula. This perspective updates recent findings on the identification of AAs in food as a result of the root uptake of free AAs released from the decayed seeds of Aristolochia clematitis L., in combination with their presence and fate in the environment. The potential link between AAs and the high prevalence of chronic kidney diseases in China is also discussed.
Collapse
|
24
|
Srijiwangsa P, Ponnikorn S, Na-Bangchang K. Effect of β-Eudesmol on NQO1 suppression-enhanced sensitivity of cholangiocarcinoma cells to chemotherapeutic agents. BMC Pharmacol Toxicol 2018; 19:32. [PMID: 29914576 PMCID: PMC6006851 DOI: 10.1186/s40360-018-0223-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/10/2018] [Indexed: 01/17/2023] Open
Abstract
Background Cholangiocarcinoma (CCA), an epithelial malignancy of the biliary tree, is one of the aggressive cancers with poor prognosis and unsatisfactory response to chemotherapy with acquired resistance. NAD(P)H-quinone oxidoreductase 1 (NQO1), an antioxidant/detoxifying enzyme, plays important roles in chemo-resistance and proliferation in several cancer cells. The study aimed to investigate the inhibitory effect of β-eudesmol on NQO1 enhanced chemotherapeutic effects of 5-fluorouracil (5-FU) and doxorubicin (DOX) in the high NQO1-expressing human CCA cell line, NQO1-KKU-100. In addition, the molecular events associated with the inhibition of the cell proliferation, cell migration, and induction of apoptosis were investigated. Methods Human CCA KKU-100 cells were exposed to β-eudesmol at various concentrations. NQO1 enzyme activity and protein expression were measured by enzymatic assay and Western blot analysis, respectively. Sulforhodamine B (SRB) assay and wound healing assay were performed to detect the inhibitory effect of β-eudesmol on cell proliferation, cell migration, and sensitivity to 5-FU and DOX. Apoptotic induction was detected by flow cytometry with annexin V/PI and DAPI nuclear staining. Caspase 3/7 activation was determined by fluorescence microscopy. The mechanism of enhanced chemo-sensitivity was evaluated by Western blot analysis. Results β-Eudesmol significantly suppressed NQO1 enzyme activity (both in KKU-100 cells and cell lysates) and protein expression in KKU-100 cells in a concentration-dependent manner. β-Eudesmol exhibited potent cytotoxicity on KKU-100 cells with mean ± SD IC50 values of 47.62 ± 9.54 and 37.46 ± 12.58 μM at 24 and 48 h, respectively. In addition, it also potentiated the cytotoxic activities and inhibitory activities of 5-FU and DOX on cell migration through induction of cell apoptosis and activation of caspase 3/7. Western blot analysis suggested that β-eudesmol enhanced chemosensitivity was associated with the suppression of NQO1 protein and activation of Bax/Bcl-2 protein expression ratio in CCA cells. Conclusions β-Eudesmol may serve as a potential anti-CCA candidate particularly when used in combination with conventional chemotherapeutics. The mechanisms involved may be mediated via NQO1 suppression-related apoptosis pathway.
Collapse
Affiliation(s)
- Pimradasiri Srijiwangsa
- Chulabhorn International College of Medicine, Thammasat University, (Rangsit Campus), Pathum Thani, 12121, Thailand
| | - Saranyoo Ponnikorn
- Chulabhorn International College of Medicine, Thammasat University, (Rangsit Campus), Pathum Thani, 12121, Thailand
| | - Kesara Na-Bangchang
- Chulabhorn International College of Medicine, Thammasat University, (Rangsit Campus), Pathum Thani, 12121, Thailand. .,Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand.
| |
Collapse
|
25
|
Chang MM, Lin CN, Fang CC, Chen M, Liang PI, Li WM, Yeh BW, Cheng HC, Huang BM, Wu WJ, Chen YMA. Glycine N-methyltransferase inhibits aristolochic acid nephropathy by increasing CYP3A44 and decreasing NQO1 expression in female mouse hepatocytes. Sci Rep 2018; 8:6960. [PMID: 29725048 PMCID: PMC5934382 DOI: 10.1038/s41598-018-22298-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/19/2018] [Indexed: 12/12/2022] Open
Abstract
Plants containing aristolochic acids (AA) are nephrotoxins. Glycine N-methyltransferase (GNMT) acts to bind environmental toxins such as benzo(a)pyrene and aflatoxin B1, translocate into nucleus, and alter hepatic metabolism. This study aims to determine the role of GNMT in AA-induced nephropathy. We established an AA nephropathy mouse model and found that AA type I (AAI)-induced nephropathy at a lower concentration in male than in female mice, implying sex differences in AAI resistance. Microarray analysis and AAI-treated mouse models showed that GNMT moderately reduced AAI-induced nephropathy by lowering the upregulated level of NQO1 in male, but significantly improved the nephropathy additionally by increasing Cyp3A44/3A41 in female. The protective effects of GNMT were absent in female GNMT knockout mice, in which re-expression of hepatic GNMT significantly decreased AAI-induced nephropathy. Mechanism-wise, AAI enhanced GNMT nuclear translocation, resulting in GNMT interaction with the promoter region of the genes encoding Nrf2 and CAR/PXR, the transcription factors for NQO1 and CYP3A44/3A41, respectively. Unlike the preference for Nrf2/NQO1 transcriptions at lower levels of GNMT, overexpression of GNMT preferred CAR/PXR/CYP3A44/3A41 transcriptions and alleviated kidney injury upon AAI treatment. In summary, hepatic GNMT protected mice from AAI nephropathy by enhancing CAR/PXR/CYP3A44/3A41 transcriptions and reducing Nrf2/NQO1 transcriptions.
Collapse
Affiliation(s)
- Ming-Min Chang
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chang-Ni Lin
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Chieh Fang
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Marcelo Chen
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Urology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Cosmetic Applications and Management, Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Peir-In Liang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Ming Li
- Pingtung Hospital, Ministry of Health and Welfare, Executive Yuan, Pingtung, Taiwan.,Department of Urology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Bi-Wen Yeh
- Department of Urology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hung-Chi Cheng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bu-Miin Huang
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Jeng Wu
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Urology, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ming Arthur Chen
- Center for Infectious Disease and Cancer Research (CICAR), Kaohsiung Medical University, Kaohsiung, Taiwan. .,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
26
|
Chang SY, Weber EJ, Sidorenko VS, Chapron A, Yeung CK, Gao C, Mao Q, Shen D, Wang J, Rosenquist TA, Dickman KG, Neumann T, Grollman AP, Kelly EJ, Himmelfarb J, Eaton DL. Human liver-kidney model elucidates the mechanisms of aristolochic acid nephrotoxicity. JCI Insight 2017; 2:95978. [PMID: 29202460 DOI: 10.1172/jci.insight.95978] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022] Open
Abstract
Environmental exposures pose a significant threat to human health. However, it is often difficult to study toxicological mechanisms in human subjects due to ethical concerns. Plant-derived aristolochic acids are among the most potent nephrotoxins and carcinogens discovered to date, yet the mechanism of bioactivation in humans remains poorly understood. Microphysiological systems (organs-on-chips) provide an approach to examining the complex, species-specific toxicological effects of pharmaceutical and environmental chemicals using human cells. We microfluidically linked a kidney-on-a-chip with a liver-on-a-chip to determine the mechanisms of bioactivation and transport of aristolochic acid I (AA-I), an established nephrotoxin and human carcinogen. We demonstrate that human hepatocyte-specific metabolism of AA-I substantially increases its cytotoxicity toward human kidney proximal tubular epithelial cells, including formation of aristolactam adducts and release of kidney injury biomarkers. Hepatic biotransformation of AA-I to a nephrotoxic metabolite involves nitroreduction, followed by sulfate conjugation. Here, we identify, in a human tissue-based system, that the sulfate conjugate of the hepatic NQO1-generated aristolactam product of AA-I (AL-I-NOSO3) is the nephrotoxic form of AA-I. This conjugate can be transported out of liver via MRP membrane transporters and then actively transported into kidney tissue via one or more organic anionic membrane transporters. This integrated microphysiological system provides an ex vivo approach for investigating organ-organ interactions, whereby the metabolism of a drug or other xenobiotic by one tissue may influence its toxicity toward another, and represents an experimental approach for studying chemical toxicity related to environmental and other toxic exposures.
Collapse
Affiliation(s)
- Shih-Yu Chang
- Department of Environmental and Occupational Health Sciences and
| | - Elijah J Weber
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Viktoriya S Sidorenko
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | - Alenka Chapron
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Catherine K Yeung
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.,Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - Chunying Gao
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Qingcheng Mao
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Danny Shen
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Thomas A Rosenquist
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | - Kathleen G Dickman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | | | - Arthur P Grollman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA.,Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Jonathan Himmelfarb
- Department of Medicine, Division of Nephrology, Kidney Research Institute, University of Washington, Seattle, Washington, USA
| | - David L Eaton
- Department of Environmental and Occupational Health Sciences and
| |
Collapse
|
27
|
DNA Adducts Formed by Aristolochic Acid Are Unique Biomarkers of Exposure and Explain the Initiation Phase of Upper Urothelial Cancer. Int J Mol Sci 2017; 18:ijms18102144. [PMID: 29036902 PMCID: PMC5666826 DOI: 10.3390/ijms18102144] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 02/06/2023] Open
Abstract
Aristolochic acid (AA) is a plant alkaloid that causes aristolochic acid nephropathy (AAN) and Balkan endemic nephropathy (BEN), unique renal diseases frequently associated with upper urothelial cancer (UUC). This review summarizes the significance of AA-derived DNA adducts in the aetiology of UUC leading to specific A:T to T:A transversion mutations (mutational signature) in AAN/BEN-associated tumours, which are otherwise rare in individuals with UCC not exposed to AA. Therefore, such DNA damage produced by AA-DNA adducts is one rare example of the direct association of exposure and cancer development (UUC) in humans, confirming that the covalent binding of carcinogens to DNA is causally related to tumourigenesis. Although aristolochic acid I (AAI), the major component of the natural plant extract AA, might directly cause interstitial nephropathy, enzymatic activation of AAI to reactive intermediates capable of binding to DNA is a necessary step leading to the formation of AA-DNA adducts and subsequently AA-induced malignant transformation. Therefore, AA-DNA adducts can not only be utilized as biomarkers for the assessment of AA exposure and markers of AA-induced UUC, but also be used for the mechanistic evaluation of its enzymatic activation and detoxification. Differences in AA metabolism might be one of the reasons for an individual’s susceptibility in the multi-step process of AA carcinogenesis and studying associations between activities and/or polymorphisms of the enzymes metabolising AA is an important determinant to identify individuals having a high risk of developing AA-mediated UUC.
Collapse
|
28
|
Jadot I, Declèves AE, Nortier J, Caron N. An Integrated View of Aristolochic Acid Nephropathy: Update of the Literature. Int J Mol Sci 2017; 18:ijms18020297. [PMID: 28146082 PMCID: PMC5343833 DOI: 10.3390/ijms18020297] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 01/09/2023] Open
Abstract
The term “aristolochic acid nephropathy” (AAN) is used to include any form of toxic interstitial nephropathy that is caused either by ingestion of plants containing aristolochic acids (AA) as part of traditional phytotherapies (formerly known as “Chinese herbs nephropathy”), or by the environmental contaminants in food (Balkan endemic nephropathy). It is frequently associated with urothelial malignancies. Although products containing AA have been banned in most of countries, AAN cases remain regularly reported all over the world. Moreover, AAN incidence is probably highly underestimated given the presence of AA in traditional herbal remedies worldwide and the weak awareness of the disease. During these two past decades, animal models for AAN have been developed to investigate underlying molecular and cellular mechanisms involved in AAN pathogenesis. Indeed, a more-in-depth understanding of these processes is essential to develop therapeutic strategies aimed to reduce the global and underestimated burden of this disease. In this regard, our purpose was to build a broad overview of what is currently known about AAN. To achieve this goal, we aimed to summarize the latest data available about underlying pathophysiological mechanisms leading to AAN development with a particular emphasis on the imbalance between vasoactive factors as well as a focus on the vascular events often not considered in AAN.
Collapse
Affiliation(s)
- Inès Jadot
- Molecular Physiology Research Unit-URPhyM, Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), Namur 5000, Belgium.
| | - Anne-Emilie Declèves
- Laboratory of Molecular Biology, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMons), Mons 7000, Belgium.
| | - Joëlle Nortier
- Nephrology Department, Erasme Academic Hospital and Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium.
| | - Nathalie Caron
- Molecular Physiology Research Unit-URPhyM, Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), Namur 5000, Belgium.
| |
Collapse
|
29
|
Stiborová M, Arlt VM, Schmeiser HH. Balkan endemic nephropathy: an update on its aetiology. Arch Toxicol 2016; 90:2595-2615. [PMID: 27538407 PMCID: PMC5065591 DOI: 10.1007/s00204-016-1819-3] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/04/2016] [Indexed: 02/02/2023]
Abstract
Balkan endemic nephropathy (BEN) is a unique, chronic renal disease frequently associated with upper urothelial cancer (UUC). It only affects residents of specific farming villages located along tributaries of the Danube River in Bosnia-Herzegovina, Croatia, Macedonia, Serbia, Bulgaria, and Romania where it is estimated that ~100,000 individuals are at risk of BEN, while ~25,000 have the disease. This review summarises current findings on the aetiology of BEN. Over the last 50 years, several hypotheses on the cause of BEN have been formulated, including mycotoxins, heavy metals, viruses, and trace-element insufficiencies. However, recent molecular epidemiological studies provide a strong case that chronic dietary exposure to aristolochic acid (AA) a principal component of Aristolochia clematitis which grows as a weed in the wheat fields of the endemic regions is the cause of BEN and associated UUC. One of the still enigmatic features of BEN that need to be resolved is why the prevalence of BEN is only 3-7 %. This suggests that individual genetic susceptibilities to AA exist in humans. In fact dietary ingestion of AA along with individual genetic susceptibility provides a scenario that plausibly can explain all the peculiarities of BEN such as geographical distribution and high risk of urothelial cancer. For the countries harbouring BEN implementing public health measures to avoid AA exposure is of the utmost importance because this seems to be the best way to eradicate this once mysterious disease to which the residents of BEN villages have been completely and utterly at mercy for so long.
Collapse
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40, Prague 2, Czech Republic.
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environmental and Health, King's College London, 150 Stamford Street, London, SE1 9NH, UK
- NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Heinz H Schmeiser
- Division of Radiopharmaceutical Chemistry (E030), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| |
Collapse
|
30
|
Hashimoto K, Zaitseva IN, Bonala R, Attaluri S, Ozga K, Iden CR, Johnson F, Moriya M, Grollman AP, Sidorenko VS. Sulfotransferase-1A1-dependent bioactivation of aristolochic acid I and N-hydroxyaristolactam I in human cells. Carcinogenesis 2016; 37:647-655. [PMID: 27207664 DOI: 10.1093/carcin/bgw045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 04/12/2016] [Indexed: 11/14/2022] Open
Abstract
Aristolochic acids (AA) are implicated in the development of chronic renal disease and upper urinary tract carcinoma in humans. Using in vitro approaches, we demonstrated that N-hydroxyaristolactams, metabolites derived from partial nitroreduction of AA, require sulfotransferase (SULT)-catalyzed conjugation with a sulfonyl group to form aristolactam-DNA adducts. Following up on this observation, bioactivation of AA-I and N-hydroxyaristolactam I (AL-I-NOH) was studied in human kidney (HK-2) and skin fibroblast (GM00637) cell lines. Pentachlorophenol, a known SULT inhibitor, significantly reduced cell death and aristolactam-DNA adduct levels in HK-2 cells following exposure to AA-I and AL-I-NOH, suggesting a role for Phase II metabolism in AA activation. A gene knockdown, siRNA approach was employed to establish the involvement of selected SULTs and nitroreductases in AA-I bioactivation. Silencing of SULT1A1 and PAPSS2 led to a significant decrease in aristolactam-DNA levels in both cell lines following exposure to AA-I, indicating the critical role for sulfonation in the activation of AA-I in vivo Since HK-2 cells proved relatively resistant to knockdown with siRNAs, gene silencing of xanthine oxidoreductase, cytochrome P450 oxidoreductase and NADPH:quinone oxidoreductase was conducted in GM00637 cells, showing a significant increase, decrease and no effect on aristolactam-DNA levels, respectively. In GM00637 cells exposed to AL-I-NOH, suppressing the SULT pathway led to a significant decrease in aristolactam-DNA formation, mirroring data obtained for AA-I. We conclude from these studies that SULT1A1 is involved in the bioactivation of AA-I through the sulfonation of AL-I-NOH, contributing significantly to the toxicities of AA observed in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francis Johnson
- Department of Pharmacological Sciences.,Department of Chemistry and
| | | | - Arthur P Grollman
- Department of Pharmacological Sciences.,Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | | |
Collapse
|
31
|
Abdullah R, Alhusainy W, Woutersen J, Rietjens IMCM, 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 Chem Toxicol 2016; 92:104-16. [PMID: 27016491 DOI: 10.1016/j.fct.2016.03.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 02/27/2016] [Accepted: 03/21/2016] [Indexed: 12/31/2022]
Abstract
Aristolochic acids are naturally occurring nephrotoxins. This study aims to investigate whether physiologically based kinetic (PBK) model-based reverse dosimetry could convert in vitro concentration-response curves of aristolochic acid I (AAI) to in vivo dose response-curves for nephrotoxicity in rat, mouse and human. To achieve this extrapolation, PBK models were developed for AAI in these different species. Subsequently, concentration-response curves obtained from in vitro cytotoxicity models were translated to in vivo dose-response curves using PBK model-based reverse dosimetry. From the predicted in vivo dose-response curves, points of departure (PODs) for risk assessment could be derived. The PBK models elucidated species differences in the kinetics of AAI with the overall catalytic efficiency for metabolic conversion of AAI to aristolochic acid Ia (AAIa) being 2-fold higher for rat and 64-fold higher for mouse than human. Results show that the predicted PODs generally fall within the range of PODs derived from the available in vivo studies. This study provides proof of principle for a new method to predict a POD for in vivo nephrotoxicity by integrating in vitro toxicity testing with in silico PBK model-based reverse dosimetry.
Collapse
Affiliation(s)
- Rozaini Abdullah
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands; Department of Environmental & Occupational Health, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| | - Wasma Alhusainy
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands
| | - Jasper Woutersen
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands
| | - Ans Punt
- Division of Toxicology, Wageningen University, Tuinlaan 5, 6703 HE Wageningen, The Netherlands
| |
Collapse
|
32
|
Dračínská H, Bárta F, Levová K, Hudecová A, Moserová M, Schmeiser HH, Kopka K, Frei E, Arlt VM, Stiborová M. Induction of cytochromes P450 1A1 and 1A2 suppresses formation of DNA adducts by carcinogenic aristolochic acid I in rats in vivo. Toxicology 2016; 344-346:7-18. [PMID: 26845733 PMCID: PMC4804751 DOI: 10.1016/j.tox.2016.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 01/21/2016] [Accepted: 01/29/2016] [Indexed: 02/05/2023]
Abstract
Aristolochic acid I (AAI) is a natural plant alkaloid causing aristolochic acid nephropathy, Balkan endemic nephropathy and their associated urothelial malignancies. One of the most efficient enzymes reductively activating AAI to species forming AAI-DNA adducts is cytosolic NAD(P)H quinone oxidoreductase 1. AAI is also either reductively activated or oxidatively detoxified to 8-hydroxyaristolochic acid (AAIa) by microsomal cytochrome P450 (CYP) 1A1 and 1A2. Here, we investigated which of these two opposing CYP1A1/2-catalyzed reactions prevails in AAI metabolism in vivo. The formation of AAI-DNA adducts was analyzed in liver, kidney and lung of rats treated with AAI, Sudan I, a potent inducer of CYP1A1/2, or AAI after pretreatment with Sudan I. Compared to rats treated with AAI alone, levels of AAI-DNA adducts determined by the (32)P-postlabeling method were lower in liver, kidney and lung of rats treated with AAI after Sudan I. The induction of CYP1A1/2 by Sudan I increased AAI detoxification to its O-demethylated metabolite AAIa, thereby reducing the actual amount of AAI available for reductive activation. This subsequently resulted in lower AAI-DNA adduct levels in the rat in vivo. Our results demonstrate that CYP1A1/2-mediated oxidative detoxification of AAI is the predominant role of these enzymes in rats in vivo, thereby suppressing levels of AAI-DNA adducts.
Collapse
Affiliation(s)
- Helena Dračínská
- Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - František Bárta
- Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Kateřina Levová
- Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Alena Hudecová
- Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Michaela Moserová
- Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Heinz H Schmeiser
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Eva Frei
- Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment & Health, King's College London, London, United Kingdom
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University in Prague, Prague, Czech Republic.
| |
Collapse
|
33
|
Milichovský J, Bárta F, Schmeiser HH, Arlt VM, Frei E, Stiborová M, Martínek V. Active Site Mutations as a Suitable Tool Contributing to Explain a Mechanism of Aristolochic Acid I Nitroreduction by Cytochromes P450 1A1, 1A2 and 1B1. Int J Mol Sci 2016; 17:213. [PMID: 26861298 PMCID: PMC4783945 DOI: 10.3390/ijms17020213] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/13/2016] [Accepted: 01/21/2016] [Indexed: 12/21/2022] Open
Abstract
Aristolochic acid I (AAI) is a plant drug found in Aristolochia species that causes aristolochic acid nephropathy, Balkan endemic nephropathy and their associated urothelial malignancies. AAI is activated via nitroreduction producing genotoxic N-hydroxyaristolactam, which forms DNA adducts. The major enzymes responsible for the reductive bioactivation of AAI are NAD(P)H quinone oxidoreductase and cytochromes P450 (CYP) 1A1 and 1A2. Using site-directed mutagenesis we investigated the possible mechanisms of CYP1A1/1A2/1B1-catalyzed AAI nitroreduction. Molecular modelling predicted that the hydroxyl groups of serine122/threonine124 (Ser122/Thr124) amino acids in the CYP1A1/1A2-AAI binary complexes located near to the nitro group of AAI, are mechanistically important as they provide the proton required for the stepwise reduction reaction. In contrast, the closely related CYP1B1 with no hydroxyl group containing residues in its active site is ineffective in catalyzing AAI nitroreduction. In order to construct an experimental model, mutant forms of CYP1A1 and 1A2 were prepared, where Ser122 and Thr124 were replaced by Ala (CYP1A1-S122A) and Val (CYP1A2-T124V), respectively. Similarly, a CYP1B1 mutant was prepared in which Ala133 was replaced by Ser (CYP1B1-A133S). Site-directed mutagenesis was performed using a quickchange approach. Wild and mutated forms of these enzymes were heterologously expressed in Escherichia coli and isolated enzymes characterized using UV-vis spectroscopy to verify correct protein folding. Their catalytic activity was confirmed with CYP1A1, 1A2 and 1B1 marker substrates. Using (32)P-postlabelling we determined the efficiency of wild-type and mutant forms of CYP1A1, 1A2, and 1B1 reconstituted with NADPH:CYP oxidoreductase to bioactivate AAI to reactive intermediates forming covalent DNA adducts. The S122A and T124V mutations in CYP1A1 and 1A2, respectively, abolished the efficiency of CYP1A1 and 1A2 enzymes to generate AAI-DNA adducts. In contrast, the formation of AAI-DNA adducts was catalyzed by CYP1B1 with the A133S mutation. Our experimental model confirms the importance of the hydroxyl group possessing amino acids in the active center of CYP1A1 and 1A2 for AAI nitroreduction.
Collapse
Affiliation(s)
- Jan Milichovský
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, CZ-12843 Prague 2, Czech Republic.
| | - František Bárta
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, CZ-12843 Prague 2, Czech Republic.
| | - Heinz H Schmeiser
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment and Health, King's College London, London SE1 9NH, UK.
| | - Eva Frei
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, CZ-12843 Prague 2, Czech Republic.
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, CZ-12843 Prague 2, Czech Republic.
| | - Václav Martínek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, CZ-12843 Prague 2, Czech Republic.
| |
Collapse
|
34
|
Stiborová M, Bárta F, Levová K, Hodek P, Schmeiser HH, Arlt VM, Martínek V. 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. Int J Mol Sci 2015; 16:27561-75. [PMID: 26593908 PMCID: PMC4661905 DOI: 10.3390/ijms161126047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 01/30/2023] Open
Abstract
Aristolochic acid I (AAI) is a plant alkaloid causing aristolochic acid nephropathy, Balkan endemic nephropathy and their associated urothelial malignancies. AAI is detoxified by cytochrome P450 (CYP)-mediated O-demethylation to 8-hydroxyaristolochic acid I (aristolochic acid Ia, AAIa). We previously investigated the efficiencies of human and rat CYPs in the presence of two other components of the mixed-functions-oxidase system, NADPH:CYP oxidoreductase and cytochrome b₅, to oxidize AAI. Human and rat CYP1A are the major enzymes oxidizing AAI. Other CYPs such as CYP2C, 3A4, 2D6, 2E1, and 1B1, also form AAIa, but with much lower efficiency than CYP1A. Based on velocities of AAIa formation by examined CYPs and their expression levels in human and rat livers, here we determined the contributions of individual CYPs to AAI oxidation in these organs. Human CYP1A2 followed by CYP2C9, 3A4 and 1A1 were the major enzymes contributing to AAI oxidation in human liver, while CYP2C and 1A were most important in rat liver. We employed flexible in silico docking methods to explain the differences in AAI oxidation in the liver by human CYP1A1, 1A2, 2C9, and 3A4, the enzymes that all O-demethylate AAI, but with different effectiveness. We found that the binding orientations of the methoxy group of AAI in binding centers of the CYP enzymes and the energies of AAI binding to the CYP active sites dictate the efficiency of AAI oxidation. Our results indicate that utilization of experimental and theoretical methods is an appropriate study design to examine the CYP-catalyzed reaction mechanisms of AAI oxidation and contributions of human hepatic CYPs to this metabolism.
Collapse
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2 CZ-12843, Czech Republic.
| | - František Bárta
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2 CZ-12843, Czech Republic.
| | - Kateřina Levová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2 CZ-12843, Czech Republic.
| | - Petr Hodek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2 CZ-12843, Czech Republic.
| | - Heinz H Schmeiser
- Division of Radiopharmaceutical Chemistry, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany.
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environment and Health, King's College London, London SE1 9NH, UK.
| | - Václav Martínek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, Prague 2 CZ-12843, Czech Republic.
| |
Collapse
|
35
|
|
36
|
Stiborová M, Bárta F, Levová K, Hodek P, Frei E, Arlt VM, Schmeiser HH. The influence of ochratoxin A on DNA adduct formation by the carcinogen aristolochic acid in rats. Arch Toxicol 2014; 89:2141-58. [PMID: 25209566 DOI: 10.1007/s00204-014-1360-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/28/2014] [Indexed: 11/28/2022]
Abstract
UNLABELLED Exposure to the plant nephrotoxin and carcinogen aristolochic acid (AA) leads to the development of AA nephropathy, Balkan endemic nephropathy (BEN) and upper urothelial carcinoma (UUC) in humans. Beside AA, exposure to ochratoxin A (OTA) was linked to BEN. Although OTA was rejected as a factor for BEN/UUC, there is still no information whether the development of AA-induced BEN/UUC is influenced by OTA exposure. Therefore, we studied the influence of OTA on the genotoxicity of AA (AA-DNA adduct formation) in vivo. AA-DNA adducts were formed in liver and kidney of rats treated with AA or AA combined with OTA, but no OTA-related DNA adducts were detectable in rats treated with OTA alone or OTA combined with AA. Compared to rats treated with AA alone, AA-DNA adduct levels were 5.4- and 1.6-fold higher in liver and kidney, respectively, of rats treated with AA combined with OTA. Although AA and OTA induced NAD(P)H quinone oxidoreductase (NQO1) activating AA to DNA adducts, their combined treatment did not lead to either higher NQO1 enzyme activity or higher AA-DNA adduct levels in ex vivo incubations. Oxidation of AA I (8-methoxy-6-nitrophenanthro[3,4-d]-1,3-dioxole-5-carboxylic acid) to its detoxification metabolite, 8-hydroxyaristolochic acid, was lower in microsomes from rats treated with AA and OTA, and this was paralleled by lower activities of cytochromes P450 1A1/2 and/or 2C11 in these microsomes. Our results indicate that a decrease in AA detoxification after combined exposure to AA and OTA leads to an increase in AA-DNA adduct formation in liver and kidney of rats.
Collapse
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40, Prague 2, Czech Republic.
| | - František Bárta
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40, Prague 2, Czech Republic
| | - Kateřina Levová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40, Prague 2, Czech Republic
| | - Petr Hodek
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40, Prague 2, Czech Republic
| | - Eva Frei
- Division of Preventive Oncology, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environmental and Health, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Heinz H Schmeiser
- Division of Radiopharmaceutical Chemistry (E030), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| |
Collapse
|
37
|
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; 768:1-7. [PMID: 24769487 DOI: 10.1016/j.mrgentox.2014.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 12/22/2013] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
Abstract
Aristolochic acid is the cause of aristolochic acid nephropathy (AAN) and Balkan endemic nephropathy (BEN) and their associated urothelial malignancies. Using Western blotting, we investigated the expression of NAD(P)H quinone oxidoreductase (NQO1), the most efficient cytosolic enzyme that reductively activates aristolochic acid I (AAI) in mice and rats. In addition, the effect of AAI on the expression of the NQO1 protein and its enzymatic activity in these experimental animal models was examined. We found that NQO1 protein levels in cytosolic fractions isolated from liver, kidney and lung of mice differed from those expressed in these organs of rats. In mice, the highest levels of NQO1 protein and NQO1 activity were found in the kidney, followed by lung and liver. In contrast, the NQO1 protein levels and enzyme activity were lowest in rat-kidney cytosol, whereas the highest amounts of NQO1 protein and activity were found in lung cytosols, followed by those of liver. NQO1 protein and enzyme activity were induced in liver and kidney of AAI-pretreated mice compared with those of untreated mice. NQO1 protein and enzyme activity were also induced in rat kidney by AAI. Furthermore, the increase in hepatic and renal NQO1 enzyme activity was associated with AAI bio-activation and elevated AAI-DNA adduct levels were found in ex vivo incubations of cytosolic fractions with DNA and AAI. In conclusion, our results indicate that AAI can increase its own metabolic activation by inducing NQO1, thereby enhancing its own genotoxic potential.
Collapse
Affiliation(s)
- František Bárta
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Kateřina Levová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Eva Frei
- Division of Preventive Oncology, National Center for Tumour Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Heinz H Schmeiser
- Research Group Genetic Alteration in Carcinogenesis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, King's College London, MRC-PHE Centre for Environmental & Health, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic.
| |
Collapse
|
38
|
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. Int J Mol Sci 2014; 15:10271-95. [PMID: 24918288 PMCID: PMC4100152 DOI: 10.3390/ijms150610271] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 01/14/2023] Open
Abstract
This review summarizes the results found in studies investigating the enzymatic activation of two genotoxic nitro-aromatics, an environmental pollutant and carcinogen 3-nitrobenzanthrone (3-NBA) and a natural plant nephrotoxin and carcinogen aristolochic acid I (AAI), to reactive species forming covalent DNA adducts. Experimental and theoretical approaches determined the reasons why human NAD(P)H quinone oxidoreductase (NQO1) and cytochromes P450 (CYP) 1A1 and 1A2 have the potential to reductively activate both nitro-aromatics. The results also contributed to the elucidation of the molecular mechanisms of these reactions. The contribution of conjugation enzymes such as N,O-acetyltransferases (NATs) and sulfotransferases (SULTs) to the activation of 3-NBA and AAI was also examined. The results indicated differences in the abilities of 3-NBA and AAI metabolites to be further activated by these conjugation enzymes. The formation of DNA adducts generated by both carcinogens during their reductive activation by the NOQ1 and CYP1A1/2 enzymes was investigated with pure enzymes, enzymes present in subcellular cytosolic and microsomal fractions, selective inhibitors, and animal models (including knock-out and humanized animals). For the theoretical approaches, flexible in silico docking methods as well as ab initio calculations were employed. The results summarized in this review demonstrate that a combination of experimental and theoretical approaches is a useful tool to study the enzyme-mediated reaction mechanisms of 3-NBA and AAI reduction.
Collapse
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, CZ-12843, Prague 2, Czech Republic.
| | - Eva Frei
- Division of Preventive Oncology, National Center for Tumor Diseases, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Heinz H Schmeiser
- Radiopharmaceutical Chemistry E030, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Volker M Arlt
- Analytical and Environmental Sciences Division, MRC-PHE Centre for Environmental & Health, King's College London, 150 Stamford Street, London SE1 9NH, UK.
| | - Václav Martínek
- Department of Biochemistry, Faculty of Science, Charles University, Hlavova 2030, CZ-12843, Prague 2, Czech Republic.
| |
Collapse
|
39
|
Romanov V, Whyard TC, Waltzer WC, Grollman AP, Rosenquist T. Aristolochic acid-induced apoptosis and G2 cell cycle arrest depends on ROS generation and MAP kinases activation. Arch Toxicol 2014; 89:47-56. [DOI: 10.1007/s00204-014-1249-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 04/15/2014] [Indexed: 12/13/2022]
|
40
|
Stiborová M, Levová K, Bárta F, Šulc M, Frei E, Arlt VM, Schmeiser HH. The influence of dicoumarol on the bioactivation of the carcinogen aristolochic acid I in rats. Mutagenesis 2014; 29:189-200. [PMID: 24598128 DOI: 10.1093/mutage/geu004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aristolochic acid I (AAI) is the major toxic component of the plant extract AA, which leads to the development of nephropathy and urothelial cancer in human. Individual susceptibility to AAI-induced disease might reflect variability in enzymes that metabolise AAI. In vitro NAD(P)H quinone oxidoreductase (NQO1) is the most potent enzyme that activates AAI by catalyzing formation of AAI-DNA adducts, which are found in kidneys of patients exposed to AAI. Inhibition of renal NQO1 activity by dicoumarol has been shown in mice. Here, we studied the influence of dicoumarol on metabolic activation of AAI in Wistar rats in vivo. In contrast to previous in vitro findings, dicoumarol did not inhibit AAI-DNA adduct formation in rats. Compared with rats treated with AAI alone, 11- and 5.4-fold higher AAI-DNA adduct levels were detected in liver and kidney, respectively, of rats pretreated with dicoumarol prior to exposure to AAI. Cytosols and microsomes isolated from liver and kidney of these rats were analysed for activity and protein levels of enzymes known to be involved in AAI metabolism. The combination of dicoumarol with AAI induced NQO1 protein level and activity in both organs. This was paralleled by an increase in AAI-DNA adduct levels found in ex vivo incubations with cytosols from rats pretreated with dicoumarol compared to cytosols from untreated rats. Microsomal ex vivo incubations showed a lower AAI detoxication to its oxidative metabolite, 8-hydroxyaristolochic acid (AAIa), although cytochrome P450 (CYP) 1A was practically unchanged. Because of these unexpected results, we examined CYP2C activity in microsomes and found that treatment of rats with dicoumarol alone and in combination with AAI inhibited CYP2C6/11 in liver. Therefore, these results indicate that CYP2C enzymes might contribute to AAI detoxication.
Collapse
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
41
|
Sidorenko VS, Attaluri S, Zaitseva I, Iden CR, Dickman KG, Johnson F, Grollman AP. Bioactivation of the human carcinogen aristolochic acid. Carcinogenesis 2014; 35:1814-22. [PMID: 24743514 DOI: 10.1093/carcin/bgu095] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Aristolochic acids are potent human carcinogens; the role of phase II metabolism in their bioactivation is unclear. Accordingly, we tested the ability of the partially reduced metabolites, N-hydroxyaristolactams (AL-NOHs), and their N-O-sulfonated and N-O-acetylated derivatives to react with DNA to form aristolactam-DNA adducts. AL-NOHs displayed little or no activity in this regard while the sulfo- and acetyl compounds readily form DNA adducts, as detected by (32)P-post-labeling analysis. Mouse hepatic and renal cytosols stimulated binding of AL-NOHs to DNA in the presence of adenosine 3'-phosphate 5'-phosphosulfate (PAPS) but not of acetyl-CoA. Using Time of Flight liquid chromatography/mass spectrometry, N-hydroxyaristolactam I formed the sulfated compound in the presence of PAPS and certain human sulfotransferases, SULT1B1 >>> SULT1A2 > SULT1A1 >>> SULT1A3. The same pattern of SULT reactivity was observed when N-hydroxyaristolactam I was incubated with these enzymes and PAPS and the reaction was monitored by formation of aristolactam-DNA adducts. In the presence of human NAD(P)H quinone oxidoreductase, the ability of aristolochic acid I to bind DNA covalently was increased significantly by addition of PAPS and SULT1B1. We conclude from these studies that AL-NOHs, formed following partial nitroreduction of aristolochic acids, serve as substrates for SULT1B1, producing N-sulfated esters, which, in turn, are converted to highly active species that react with DNA and, potentially, cellular proteins, resulting in the genotoxicity and nephrotoxicity associated with ingestion of aristolochic acids by humans.
Collapse
Affiliation(s)
| | | | | | | | | | - Francis Johnson
- Department of Pharmacological Sciences, Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, USA
| | | |
Collapse
|
42
|
Michl J, Ingrouille MJ, Simmonds MSJ, Heinrich M. Naturally occurring aristolochic acid analogues and their toxicities. Nat Prod Rep 2014; 31:676-93. [DOI: 10.1039/c3np70114j] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
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; 44:135-45. [PMID: 24152141 DOI: 10.3109/00498254.2013.848310] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
1. Aristolochic acid I (AAI) is the predominant component in plant extract of Aristolochia genus that is involved in development of aristolochic acid nephropathy, Balkan endemic nephropathy and urothelial cancer. The diseases do not develop in all individuals exposed to AAI and patients exhibit different clinical outcomes. Differences in the activities of enzymes catalyzing the metabolism of AAI might be one of the reasons for this individual susceptibility. 2. Understanding which human enzymes are involved in reductive activation of AAI generating AAI-DNA adducts, and/or its detoxication to the O-demethylated metabolite, aristolochic acid Ia (AAIa), is necessary in the assessment of the susceptibility to this compound. 3. This review summarizes the results of the latest studies utilizing genetically engineered mouse models to identify which human and rodent enzymes catalyze the reductive activation of AAI to AAI-DNA adducts and its oxidative detoxication to AAIa in vivo. 4. The use of hepatic cytochrome P450 (Cyp) reductase null (HRN) mice, in which NADPH:Cyp oxidoreductase (Por) is deleted in hepatocytes, Cyp1a1((-/-)), Cyp1a2((-/-)) single-knockout, Cyp1a1/1a2((-/-)) double-knockout and CYP1A-humanized mice revealed that mouse and human CYP1A1 and 1A2, besides mouse NAD(P)H quinone oxidoreductase, were involved in the activation of AAI but CYP1A1 and 1A2 also oxidatively detoxified AAI.
Collapse
Affiliation(s)
- Marie Stiborová
- Department of Biochemistry, Faculty of Science, Charles University , Prague , Czech Republic
| | | | | | | |
Collapse
|
44
|
Radford R, Frain H, Ryan MP, Slattery C, McMorrow T. Mechanisms of chemical carcinogenesis in the kidneys. Int J Mol Sci 2013; 14:19416-33. [PMID: 24071941 PMCID: PMC3821564 DOI: 10.3390/ijms141019416] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 09/05/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
Chemical carcinogens are substances which induce malignant tumours, increase their incidence or decrease the time taken for tumour formation. Often, exposure to chemical carcinogens results in tissue specific patterns of tumorigenicity. The very same anatomical, biochemical and physiological specialisations which permit the kidney to perform its vital roles in maintaining tissue homeostasis may in fact increase the risk of carcinogen exposure and contribute to the organ specific carcinogenicity observed with numerous kidney carcinogens. This review will address the numerous mechanisms which play a role in the concentration, bioactivation, and uptake of substances from both the urine and blood which significantly increase the risk of cancer in the kidney.
Collapse
Affiliation(s)
- Robert Radford
- UCD School of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Dublin 4, Ireland; E-Mails: (R.R.); (H.F.); (M.P.R.); (C.S.)
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Helena Frain
- UCD School of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Dublin 4, Ireland; E-Mails: (R.R.); (H.F.); (M.P.R.); (C.S.)
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Michael P. Ryan
- UCD School of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Dublin 4, Ireland; E-Mails: (R.R.); (H.F.); (M.P.R.); (C.S.)
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Craig Slattery
- UCD School of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Dublin 4, Ireland; E-Mails: (R.R.); (H.F.); (M.P.R.); (C.S.)
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Tara McMorrow
- UCD School of Biomolecular and Biomedical Research, Conway Institute, University College Dublin, Dublin, Dublin 4, Ireland; E-Mails: (R.R.); (H.F.); (M.P.R.); (C.S.)
- Renal Disease Research Group, School of Biomolecular and Biomedical Science, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
45
|
Ziarani GM, Badiei A, Azizi M, Lashgari N. Efficient One‐Pot Synthesis of Bis(4‐hydroxycoumarin)methanes in the Presence of Sulfonic Acid Functionalized Nanoporous Silica (SBA‐Pr‐SO 3H). J CHIN CHEM SOC-TAIP 2013; 60:499-502. [DOI: 10.1002/jccs.201200530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
AbstractAn efficient and simple method for the synthesis of bis(4‐hydroxycoumarin)methanes has been achieved through a one‐pot condensation of aryl aldehydes and 4‐hydroxycoumarin in the presence of nanoporous solid acid catalyst of SBA‐Pr‐SO3H with pore size of 6 nm. Excellent yields, short reaction times, reusability of the catalyst and simple workup are advantages of this synthetic procedure.
Collapse
|
46
|
Madeo J, Zubair A, Marianne F. A review on the role of quinones in renal disorders. SPRINGERPLUS 2013; 2:139. [PMID: 23577302 PMCID: PMC3618882 DOI: 10.1186/2193-1801-2-139] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/10/2013] [Indexed: 12/12/2022]
Abstract
Quinones are electron and proton carriers that play a primary role in the aerobic metabolism of virtually every cell in nature. Most physiological quinones are benzoquinones. They undergo highly regulated redox reactions in the mitochondria, Golgi apparatus, plasma membrane and endoplasmic reticulum. Important consequences of these electron transfer reactions are the production of and protection against reactive oxygen species (ROS). Quinones have been extensively studied for both their cytotoxic as well as cellular protective properties and they have been particularly useful in rational drug design. The role of quinones in medicine is explored in this literature review with a particular focus on renal diseases. Due to their high basal metabolism and detoxification role, the kidneys are particularly sensitive to oxidative stress. Regardless of the underlying etiology, ROS plays an important role in both acute kidney injury (AKI) and chronic kidney diseases (CKD). Depending on the oxidative state of the kidney, quinones can be nephrotoxoic or nephro-protective. Many factors play a role in the interaction between quinones and the kidney and the consequences of this are just beginning to be explored.
Collapse
Affiliation(s)
- Jennifer Madeo
- Department of Medicine, Nassau University Medical Center, 2201 Hempstead Turnpike, East Meadow, NY 11554 USA
| | | | | |
Collapse
|
47
|
Levova K, Moserova M, Nebert DW, Phillips DH, Frei E, Schmeiser HH, Arlt VM, Stiborova M. 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. Toxicol Appl Pharmacol 2012; 265:360-7. [PMID: 22982977 DOI: 10.1016/j.taap.2012.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/05/2012] [Accepted: 09/06/2012] [Indexed: 01/01/2023]
Abstract
Aristolochic acid causes a specific nephropathy (AAN), Balkan endemic nephropathy, and urothelial malignancies. Using Western blotting suitable to determine protein expression, we investigated in several transgenic mouse lines expression of NAD(P)H:quinone oxidoreductase (NQO1)-the most efficient cytosolic enzyme that reductively activates aristolochic acid I (AAI). The mouse tissues used were from previous studies [Arlt et al., Chem. Res. Toxicol. 24 (2011) 1710; Stiborova et al., Toxicol. Sci. 125 (2012) 345], in which the role of microsomal cytochrome P450 (CYP) enzymes in AAI metabolism in vivo had been determined. We found that NQO1 levels in liver, kidney and lung of Cyp1a1⁻/⁻, Cyp1a2⁻/⁻ and Cyp1a1/1a2⁻/⁻ knockout mouse lines, as well as in two CYP1A-humanized mouse lines harboring functional human CYP1A1 and CYP1A2 and lacking the mouse Cyp1a1/1a2 orthologs, differed from NQO1 levels in wild-type mice. NQO1 protein and enzymic activity were induced in hepatic and renal cytosolic fractions isolated from AAI-pretreated mice, compared with those in untreated mice. Furthermore, this increase in hepatic NQO1 enzyme activity was associated with bioactivation of AAI and elevated AAI-DNA adduct levels in ex vivo incubations of cytosolic fractions with DNA and AAI. In conclusion, AAI appears to increase its own metabolic activation by inducing NQO1, thereby enhancing its own genotoxic potential.
Collapse
Affiliation(s)
- Katerina Levova
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Li YC, Tsai SH, Chen SM, Chang YM, Huang TC, Huang YP, Chang CT, Lee JA. Aristolochic acid-induced accumulation of methylglyoxal and Nε-(carboxymethyl)lysine: An important and novel pathway in the pathogenic mechanism for aristolochic acid nephropathy. Biochem Biophys Res Commun 2012; 423:832-7. [DOI: 10.1016/j.bbrc.2012.06.049] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 06/09/2012] [Indexed: 10/28/2022]
|