1
|
Jin YW, Ma YR, Liu YT, Yang JR, Zhang MK, Ran FL, Chen Y, Wu XA. Identification of a substrate of the renal tubular transporters for detecting drug-induced early acute kidney injury. Toxicol Sci 2024; 201:190-205. [PMID: 39041788 DOI: 10.1093/toxsci/kfae093] [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] [Indexed: 07/24/2024] Open
Abstract
Early identification of drug-induced acute kidney injury (AKI) is essential to prevent renal damage. The renal tubules are typically the first to exhibit damage, frequently accompanied by changes in renal tubular transporters. With this in mind, we have identified an endogenous substrate of the renal tubular transporters that may serve as a biomarker for early detection of drug-induced AKI. Using gentamicin- and vancomycin-induced AKI models, we found that traumatic acid (TA), an end metabolite, was rapidly increased in both AKI models. TA, a highly albumin-bound compound (96% to 100%), could not be filtered by the glomerulus and was predominantly eliminated by renal tubules via the OAT1, OAT3, OATP4C1, and P-gp transporters. Importantly, there is a correlation between elevated serum TA levels and reduced OAT1 and OAT3 levels. A clinical study showed that serum TA levels rose before an increase in serum creatinine in 13 out of 20 AKI patients in an intensive care unit setting. In addition, there was a notable rise in TA levels in the serum of individuals suffering from nephrotic syndrome, chronic renal failure, and acute renal failure. These results indicate that the decrease in renal tubular transporter expression during drug-induced AKI leads to an increase in the serum TA level, and the change in TA may serve as a monitor for renal tubular injury. Acute kidney injury (AKI) has a high clinical incidence, and if patients do not receive timely treatment and intervention, it can lead to severe consequences. During AKI, tubular damage is often the primary issue. Endogenous biomarkers of tubular damage are critical for the early diagnosis and treatment of AKI. However, there is currently a lack of reliable endogenous biomarkers for diagnosing tubular damage in clinical practice. Tubular secretion is primarily mediated by renal tubular transporters (channels), which are also impaired during tubular damage. Therefore, we aim to identify endogenous biomarkers of tubular damage from the perspective of renal tubular transporters, providing support for the early detection and intervention of AKI. TA is a substrate of multiple channels, including OAT1, OAT3, OATP4C1, and P-gp, and is primarily secreted by the renal tubules. In the early stages of rat AKI induced by GEN and VCA, serum TA levels are significantly elevated, occurring earlier than the rise in serum creatinine (SCr). Thus, TA is expected to become a potential endogenous biomarker for the early diagnosis of tubular damage.
Collapse
Affiliation(s)
- Yong-Wen Jin
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Yan-Rong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yu-Ting Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jin-Ru Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Ming-Kang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Feng-Lin Ran
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Xin-An Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
2
|
Li C, Li X, Niu M, Xiao D, Luo Y, Wang Y, Fang ZE, Zhan X, Zhao X, Fang M, Wang J, Xiao X, Bai Z. Unveiling correlations between aristolochic acids and liver cancer: spatiotemporal heterogeneity phenomenon. Chin Med 2024; 19:132. [PMID: 39342223 PMCID: PMC11439320 DOI: 10.1186/s13020-024-01003-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024] Open
Abstract
Aristolochic acids are a class of naturally occurring compounds in Aristolochiaceae that have similar structural skeletons and chemical properties. Exposure to aristolochic acids is a risk factor for severe kidney disease and urinary system cancer. However, the carcinogenicity of aristolochic acids to the liver, which is the main site of aristolochic acid metabolism, is unclear. Although the characteristic fingerprint of aristolochic acid-induced mutations has been detected in the liver and aristolochic acids are known to be hepatotoxic, whether aristolochic acids can directly cause liver cancer is yet to be verified. This review summarizes the findings of long-term carcinogenicity studies of aristolochic acids in experimental animals. We propose that spatiotemporal heterogeneity in the carcinogenicity of these phytochemicals could explain why direct evidence of aristolochic acids causing liver cancer has never been found in adult individuals. We also summarized the reported approaches to mitigate aristolochic acid-induced hepatotoxicity to better address the associated global safety issue and provide directions and recommendations for future investigation.
Collapse
Affiliation(s)
- Chengxian Li
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Xinyu Li
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Ming Niu
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Dake Xiao
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Ye Luo
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yinkang Wang
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhi-E Fang
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, China
| | - Xiaoyan Zhan
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
- National Key Laboratory of Kidney Diseases, Beijing, 100039, China
| | - Xu Zhao
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Mingxia Fang
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jiabo Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Xiaohe Xiao
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- National Key Laboratory of Kidney Diseases, Beijing, 100039, China.
| | - Zhaofang Bai
- Department of Liver Disease, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
- National Key Laboratory of Kidney Diseases, Beijing, 100039, China.
| |
Collapse
|
3
|
Luo P, Chen J, Zhang Q, Xia F, Wang C, Bai Y, Tang H, Liu D, Gu L, Du Q, Xiao W, Yang C, Wang J. Dissection of cellular and molecular mechanisms of aristolochic acid-induced hepatotoxicity via single-cell transcriptomics. PRECISION CLINICAL MEDICINE 2022; 5:pbac023. [PMID: 36349141 PMCID: PMC9635452 DOI: 10.1093/pcmedi/pbac023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
Background Aristolochic acids (AAs), a class of carcinogenic and mutagenic natural products from Aristolochia and Asarum plants, are well-known to be responsible for inducing nephrotoxicity and urothelial carcinoma. Recently, accumulating evidence suggests that exposure to AAs could also induce hepatotoxicity and even hepatocellular carcinoma, though the mechanisms are poorly defined. Methods Here, we aimed to dissect the underlying cellular and molecular mechanisms of aristolochic acid I (AAI)-induced hepatotoxicity by using advanced single-cell RNA sequencing (scRNA-seq) and proteomics techniques. We established the first single-cell atlas of mouse livers in response to AAI. Results In hepatocytes, our results indicated that AAI activated NF-κB and STAT3 signaling pathways, which may contribute to the inflammatory response and apoptosis. In liver sinusoidal endothelial cells (LSECs), AAI activated multiple oxidative stress and inflammatory associated signaling pathways and induced apoptosis. Importantly, AAI induced infiltration of cytotoxic T cells and activation of proinflammatory macrophage and neutrophil cells in the liver to produce inflammatory cytokines to aggravate inflammation. Conclusions Collectively, our study provides novel knowledge of AAs-induced molecular characteristics of hepatotoxicity at a single-cell level and suggests future treatment options for AAs associated hepatotoxicity.
Collapse
Affiliation(s)
- Piao Luo
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jiayun Chen
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qian Zhang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Fei Xia
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chen Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yunmeng Bai
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Huan Tang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Dandan Liu
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liwei Gu
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qingfeng Du
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wei Xiao
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Chuanbin Yang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
| | - Jigang Wang
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China
- Artemisinin Research Center, and Institute of Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
- Center for Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Southern Medical University, Dongguan 523125, China
| |
Collapse
|
4
|
Wang Y, Liu Z, Ma J, Xv Q, Gao H, Yin H, Yan G, Jiang X, Yu W. Lycopene attenuates the inflammation and apoptosis in aristolochic acid nephropathy by targeting the Nrf2 antioxidant system. Redox Biol 2022; 57:102494. [PMID: 36198206 PMCID: PMC9530962 DOI: 10.1016/j.redox.2022.102494] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 10/31/2022] Open
Abstract
Lycopene (LYC) is a carotenoid, has antioxidant properties. This study investigated whether lycopene attenuates aristolochic acids (AAs) -induced chronic kidney disease. In this experiment, lycopene was used to intervene C57BL/6 mice with renal injury induced by aristolochic acid exposure. The histomorphological changes and serological parameters of the kidney were measured in order to assess the alleviating effect of lycopene on renal injury in aristolochic acid nephropathy. In vitro and in vivo experiments were carried out to verify the main mechanism of action and drug targets of lycopene in improving aristolochic acid nephropathy (AAN) and by various experimental methods such as ELISA, immunohistochemistry, immunofluorescence, Western-blot and qRT-PCR. The results showed that oxidative stress injury was induced in the kidney of mice after AAI exposure, resulting in inflammatory response and tubular epithelial cell apoptosis. The results showed that the Nrf2/HO-1 antioxidant signaling pathway was inhibited after AAI exposure. AAI induces oxidative stress injury in the kidney, which ultimately leads to inflammation and tubular epithelial cell apoptosis. After LYC intervened in the body, it activated Nrf2 nuclear translocation and its downstream HO-1 and NQO1 antioxidant signaling pathways. LYC inhibited ROS production by renal tubular epithelial cells, and alleviated mitochondrial damage. LYC further modulated the TNF-α/NF-κB signaling cascade, thereby reduced the accumulation of inflammatory factors in the renal interstitium. Moreover, LYC was able to up-regulate the expression of Bcl-2, down-regulate Bax expression and inhibit the activation of cleaved forms of Caspase-9 and Caspase-3, which finally attenuated the apoptosis of the mitochondrial pathway induced by AAI exposure. It was concluded that lycopene was able to activate the Nrf2 antioxidant signaling pathway to maintain the homeostasis of renal oxidative stress and ultimately attenuated renal inflammatory response and apoptosis. These results suggested that lycopene can be used as a drug to relieve AAN.
Collapse
Affiliation(s)
- Yu Wang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Zhihui Liu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jun Ma
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Qingyang Xv
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hongxin Gao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Hang Yin
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Ge Yan
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Provincial Key Laboratory for Prevention and Control of Common Animal Diseases, Northeast Agricultural University, Harbin, 150030, China; Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
5
|
Navarro Garrido A, Kim YC, Oe Y, Zhang H, Crespo-Masip M, Goodluck HA, Kanoo S, Sanders PW, Bröer S, Vallon V. Aristolochic acid-induced nephropathy is attenuated in mice lacking the neutral amino acid transporter B 0AT1 ( Slc6a19). Am J Physiol Renal Physiol 2022; 323:F455-F467. [PMID: 35979966 PMCID: PMC9484999 DOI: 10.1152/ajprenal.00181.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/10/2022] [Indexed: 01/10/2023] Open
Abstract
B0AT1 (Slc6a19) mediates absorption of neutral amino acids in the small intestine and in the kidneys, where it is primarily expressed in early proximal tubules (S1-S2). To determine the role of B0AT1 in nephropathy induced by aristolochic acid (AA), which targets the proximal tubule, littermate female B0AT1-deficient (Slc6a19-/-), heterozygous (Slc6a19+/-), and wild-type (WT) mice were administered AA (10 mg/kg ip) or vehicle every 3 days for 3 wk, and analyses were performed after the last injection or 3 wk later. Vehicle-treated mice lacking Slc6a19 showed normal body and kidney weight and plasma creatinine versus WT mice. The urinary glucose-to-creatinine ratio (UGCR) and urinary albumin-to-creatinine ratio (UACR) were two to four times higher in vehicle-treated Slc6a19-/- versus WT mice, associated with lesser expression of early proximal transporters Na+-glucose cotransporter 2 and megalin, respectively. AA caused tubular injury independently of B0AT1, including robust increases in cortical mRNA expression of p53, p21, and hepatitis A virus cellular receptor 1 (Havcr1), downregulation of related proximal tubule amino acid transporters B0AT2 (Slc6a15), B0AT3 (Slc6a18), and Slc7a9, and modest histological tubular damage and a rise in plasma creatinine. Absence of B0AT1, however, attenuated AA-induced cortical upregulation of mRNA markers of senescence (p16), inflammation [lipocalin 2 (Lcn2), C-C motif chemokine ligand 2 (Ccl2), and C-C motif chemokine receptor 2 (Ccr2)], and fibrosis [tissue inhibitor of metallopeptidase 1 (Timp1), transforming growth factor-β1 (Tgfb1), and collagen type I-α1 (Col1a1)], associated with lesser fibrosis staining, lesser suppression of proximal tubular organic anion transporter 1, restoration of Na+-glucose cotransporter 2 expression, and prevention of the AA-induced fivefold increase in the urinary albumin-to-creatinine ratio observed in WT mice. The data suggest that proximal tubular B0AT1 is important for the physiology of renal glucose and albumin retention but potentially deleterious for the kidney response following AA-induced kidney injury.NEW & NOTEWORTHY Based on insights from studies manipulating glucose transport, the hypothesis has been proposed that inhibiting intestinal uptake or renal reabsorption of energy substrates has unique therapeutic potential to improve metabolic disease and kidney outcome in response to injury. The present study takes this idea to B0AT1, the major transporter for neutral amino acids in the intestine and kidney, and shows that its absence attenuates aristolochic acid-induced nephropathy.
Collapse
Affiliation(s)
- Aleix Navarro Garrido
- Department of Medicine, University of California-San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Young Chul Kim
- Department of Medicine, University of California-San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Yuji Oe
- Department of Medicine, University of California-San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Haiyan Zhang
- Department of Pathology, University of California-San Diego, San Diego, California
| | - Maria Crespo-Masip
- Department of Medicine, University of California-San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Helen A Goodluck
- Department of Medicine, University of California-San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Sadhana Kanoo
- Department of Medicine, University of California-San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Paul W Sanders
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Veterans Affairs Medical Center, Birmingham, Alabama
| | - Stefan Bröer
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Volker Vallon
- Department of Medicine, University of California-San Diego, La Jolla, California
- Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
6
|
Rao J, Peng T, Li N, Wang Y, Yan C, Wang K, Qiu F. Nephrotoxicity induced by natural compounds from herbal medicines - a challenge for clinical application. Crit Rev Toxicol 2022; 52:757-778. [PMID: 36815678 DOI: 10.1080/10408444.2023.2168178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Herbal medicines (HMs) have long been considered safe and effective without serious toxic and side effects. With the continuous use of HMs, more and more attention has been paid to adverse reactions and toxic events, especially the nephrotoxicity caused by natural compounds in HMs. The composition of HMs is complex and various, especially the mechanism of toxic components has been a difficult and hot topic. This review comprehensively summarizes the kidney toxicity characterization and mechanism of nephrotoxic natural compounds (organic acids, alkaloids, glycosides, terpenoids, phenylpropanoids, flavonoids, anthraquinones, cytotoxic proteins, and minerals) from different sources. Recommendations for the prevention and treatment of HMs-induced kidney injury were provided. In vitro and in vivo models for evaluating nephrotoxicity and the latest biomarkers are also included in this investigation. More broadly, this review may provide theoretical basis for safety evaluation and further comprehensive development and utilization of HMs in the future.
Collapse
Affiliation(s)
- Jinqiu Rao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Ting Peng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Na Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Yuan Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Caiqin Yan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Kai Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| |
Collapse
|
7
|
Chen J, Luo P, Wang C, Yang C, Bai Y, He X, Zhang Q, Zhang J, Yang J, Wang S, Wang J. Integrated single-cell transcriptomics and proteomics reveal cellular-specific response and microenvironment remodeling in aristolochic acid nephropathy. JCI Insight 2022; 7:157360. [PMID: 35852860 PMCID: PMC9462482 DOI: 10.1172/jci.insight.157360] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Aristolochic acid nephropathy (AAN) is characterized by acute proximal tubule necrosis and immune cell infiltration, contributing to the global burden of chronic kidney disease and urothelial cancer. Although the proximal tubule has been defined as the primary target of aristolochic acids I (AAI), the mechanistic underpinning of gross renal deterioration caused by AAI has not been explicitly explained, prohibiting effective therapeutic intervention. To this point, we employed integrated single-cell RNA-Seq, bulk RNA-Seq, and mass spectrometry–based proteomics to analyze the mouse kidney after acute AAI exposure. Our results reveal a dramatic reduction of proximal tubule epithelial cells, associated with apoptotic and inflammatory pathways, indicating permanent damage beyond repair. We found the enriched development pathways in other nephron segments, suggesting activation of reparative programs triggered by AAI. The divergent response may be attributed to the segment-specific distribution of organic anion channels along the nephron, including OAT1 and OAT3. Moreover, we observed dramatic activation and recruitment of cytotoxic T and macrophage M1 cells, highlighting inflammation as a principal contributor to permanent renal injury. Ligand-receptor pairing revealed that critical intercellular crosstalk underpins damage-induced activation of immune cells. These results provide potentially novel insight into the AAI-induced kidney injury and point out possible pathways for future therapeutic intervention.
Collapse
Affiliation(s)
- Jiayun Chen
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Piao Luo
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chen Wang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanbin Yang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yunmeng Bai
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xueling He
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Zhang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junzhe Zhang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Yang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Shuang Wang
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Jigang Wang
- Artemisinin Research Center and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Center for Reproductive Medicine, Dongguan Maternal and Child Health Care Hospital, Southern Medical University, Dongguan, Guangdong, China
| |
Collapse
|
8
|
Askin S, Askin H, Dursun E, Palabiyik E, Uguz H, Cakmak Ö, Koc K. The hepato-renal protective potential of walnut seed skin extract against acute renal ischemia/reperfusion damage. Cytokine 2022; 153:155861. [DOI: 10.1016/j.cyto.2022.155861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/26/2022]
|
9
|
Ribeiro A, Dobosz E, Krill M, Köhler P, Wadowska M, Steiger S, Schmaderer C, Koziel J, Lech M. Macrophage-Specific MCPIP1/Regnase-1 Attenuates Kidney Ischemia-Reperfusion Injury by Shaping the Local Inflammatory Response and Tissue Regeneration. Cells 2022; 11:cells11030397. [PMID: 35159206 PMCID: PMC8834155 DOI: 10.3390/cells11030397] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/27/2023] Open
Abstract
Sterile inflammation either resolves the initial insult or leads to tissue damage. Kidney ischemia/reperfusion injury (IRI) is associated with neutrophilic infiltration, enhanced production of inflammatory mediators, accumulation of necrotic cells and tissue remodeling. Macrophage-dependent microenvironmental changes orchestrate many features of the immune response and tissue regeneration. The activation status of macrophages is influenced by extracellular signals, the duration and intensity of the stimulation, as well as various regulatory molecules. The role of macrophage-derived monocyte chemoattractant protein-induced protein 1 (MCPIP1), also known as Regnase-1, in kidney ischemia-reperfusion injury (IRI) and recovery from sterile inflammation remains unresolved. In this study, we showed that macrophage-specific Mcpip1 deletion significantly affects the kidney phenotype. Macrophage-specific Mcpip1 transgenic mice displayed enhanced inflammation and loss of the tubular compartment upon IRI. We showed that MCPIP1 modulates sterile inflammation by negative regulation of Irf4 expression and accumulation of IRF4+ cells in the tissue and, consequently, suppresses the post-ischemic kidney immune response. Thus, we identified MCPIP1 as an important molecular sentinel of immune homeostasis in experimental acute kidney injury (AKI) and renal fibrosis.
Collapse
Affiliation(s)
- Andrea Ribeiro
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
- Department of Nephrology, Klinikum Rechts der Isar, Technical University Munich, 80336 Munich, Germany;
| | - Ewelina Dobosz
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (E.D.); (M.W.); (J.K.)
| | - Moritz Krill
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
| | - Paulina Köhler
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
| | - Marta Wadowska
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (E.D.); (M.W.); (J.K.)
| | - Stefanie Steiger
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum Rechts der Isar, Technical University Munich, 80336 Munich, Germany;
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland; (E.D.); (M.W.); (J.K.)
| | - Maciej Lech
- LMU Klinikum, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany; (A.R.); (M.K.); (P.K.); (S.S.)
- Correspondence:
| |
Collapse
|
10
|
Sasaki K, Terker AS, Tang J, Cao S, Arroyo JP, Niu A, Wang S, Fan X, Zhang Y, Bennett SR, Zhang MZ, Harris RC. Macrophage interferon regulatory factor 4 deletion ameliorates aristolochic acid nephropathy via reduced migration and increased apoptosis. JCI Insight 2022; 7:150723. [PMID: 35025763 PMCID: PMC8876461 DOI: 10.1172/jci.insight.150723] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
Aristolochic acid (AA) is the causative nephrotoxic alkaloid in aristolochic acid nephropathy, which results in a tubulointerstitial fibrosis. AA causes direct proximal tubule damage. There is also an influx of macrophages, although their role in the pathogenesis is poorly understood. Here we demonstrate that AA directly stimulates migration, inflammation, and reactive oxygen species (ROS) production in macrophages ex vivo. Cells lacking interferon regulatory factor 4 (IRF4), a known regulator of macrophage migration and phenotype, had a reduced migratory response, though effects on ROS production and inflammation were preserved or increased relative to wild-type cells. Macrophage-specific IRF4 knockout mice were protected from both acute and chronic kidney effects of AA administration based on functional and histological analysis. Renal macrophages from kidneys of AA-treated macrophage-specific IRF4 knockout mice demonstrated increased apoptosis and ROS production compared with wildtype controls, indicating that AA directly polarizes macrophages to a promigratory and proinflammatory phenotype. However, knockout mice had reduced renal macrophage abundance following AA administration. While macrophages lacking IRF4 can adopt a proinflammatory phenotype upon AA exposure, their inability to migrate to the kidney and increased rates of apoptosis upon infiltration provide protection from AA in vivo. These results provide evidence of direct AA effects on macrophages in AAN and add to the growing body of evidence that supports a key role of IRF4 in modulating macrophage function in kidney injury.
Collapse
Affiliation(s)
- Kensuke Sasaki
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Andrew S Terker
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Jiaqi Tang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Shirong Cao
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Juan Pablo Arroyo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Aolei Niu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Suwan Wang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Xiaofeng Fan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Yahua Zhang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Stephanie R Bennett
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Ming-Zhi Zhang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States of America
| |
Collapse
|
11
|
Effects of tumor necrosis factor-α inhibition on kidney fibrosis and inflammation in a mouse model of aristolochic acid nephropathy. Sci Rep 2021; 11:23587. [PMID: 34880315 PMCID: PMC8654826 DOI: 10.1038/s41598-021-02864-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor (TNF)-α is a potent mediator of inflammation and is involved in the pathophysiology of chronic kidney disease (CKD). However, the effects of TNF-α inhibition on the progression of kidney fibrosis have not been fully elucidated. We examined the effects of TNF-α inhibition by etanercept (ETN) on kidney inflammation and fibrosis in mice with aristolochic acid (AA) nephropathy as a model of kidney fibrosis. C57BL/6 J mice were administered AA for 4 weeks, followed by a 4-week remodeling period. The mice exhibited kidney fibrosis, functional decline, and albuminuria concomitant with increases in renal mRNA expression of inflammation- and fibrosis-related genes. The 8-week ETN treatment partially but significantly attenuated kidney fibrosis and ameliorated albuminuria without affecting kidney function. These findings were accompanied by significant suppression of interleukin (IL)-1β, IL-6, and collagen types I and III mRNA expression. Moreover, ETN tended to reduce the AA-induced increase in interstitial TUNEL-positive cells with a significant reduction in Bax mRNA expression. Renal phosphorylated p38 MAPK was significantly upregulated by AA but was normalized by ETN. These findings indicate a substantial role for the TNF-α pathway in the pathogenesis of kidney fibrosis and suggest that TNF-α inhibition could become an adjunct therapeutic strategy for CKD with fibrosis.
Collapse
|
12
|
Jiang W, Xu C, Xu S, Su W, Du C, Dong J, Feng R, Huang C, Li J, Ma T. Macrophage-derived, LRG1-enriched extracellular vesicles exacerbate aristolochic acid nephropathy in a TGFβR1-dependent manner. Cell Biol Toxicol 2021; 38:629-648. [PMID: 34677723 DOI: 10.1007/s10565-021-09666-1] [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: 06/24/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022]
Abstract
Aristolochic acid nephropathy (AAN) is a progressive kidney disease caused by some herbal medicines, but treatment remains ineffective. We previously found that leucine-rich α-2-glycoprotein 1 (LRG1), which regulates cellular processes, plays an important role in a kidney injury model. However, the underlying mechanism by which LRG1 regulates AAN is still unknown. In this study, we established an AAN model in vivo, a coculture system of macrophages and TECs, and a macrophage/TEC conditioned media culture model in vitro. We found that macrophage infiltration promoted injury, oxidative stress, and apoptosis in TECs. Furthermore, the role of macrophages in AAN was dependent on macrophage-derived extracellular vesicles (EVs). Importantly, we found that macrophage-derived, LRG1-enriched EVs induced TEC injury and apoptosis via a TGFβR1-dependent process. This study may help design a better therapeutic strategy to treat AAN patients.
Collapse
Affiliation(s)
- Wenjuan Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Chuanting Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Songbing Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Wan Su
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Changlin Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Jiahui Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Rui Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Taotao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
13
|
Chan W, Ham YH. Probing the Hidden Role of Mitochondrial DNA Damage and Dysfunction in the Etiology of Aristolochic Acid Nephropathy. Chem Res Toxicol 2021; 34:1903-1909. [PMID: 34255491 DOI: 10.1021/acs.chemrestox.1c00175] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Aristolochic acid nephropathy (AAN) is a unique type of progressive renal interstitial fibrotic disease caused by prolonged exposure to aristolochic acids (AAs) through AA-containing herbal medicines or AA-tainted food. Despite decades of research and affecting millions of people around the world, the pathophysiology of AAN remains incompletely understood. In this study, we tested the potential causative role of mitochondrial dysfunction in AAN development. Our findings revealed AA exposure induces an exposure concentration and duration dependent lowering of adenosine triphosphate in both cultured human kidney and liver cells, highlighting an AA exposure effect on mitochondrial energy production in the kidney and liver, which both are highly metabolically active and energy-demanding organs. Analysis with liquid chromatography-tandem mass spectrometry coupled with stable isotope dilution method detected high levels of mutagenic 8-oxo-2'-deoxyguanosine and 7-(deoxyadenosine-N6-yl)-aristolactam adduct on mitochondrial DNA isolated from AA-treated cells, unmasking a potentially important causative, but previously unknown role of mitochondrial DNA mutation in the pathophysiology of AAN development.
Collapse
Affiliation(s)
- Wan Chan
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Yat-Hing Ham
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| |
Collapse
|
14
|
Li X, Zou Y, Fu YY, Xing J, Wang KY, Wan PZ, Wang M, Zhai XY. Ibudilast Attenuates Folic Acid-Induced Acute Kidney Injury by Blocking Pyroptosis Through TLR4-Mediated NF-κB and MAPK Signaling Pathways. Front Pharmacol 2021; 12:650283. [PMID: 34025417 PMCID: PMC8139578 DOI: 10.3389/fphar.2021.650283] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
Folic acid (FA)-induced renal tubule damage, which is characterized by extensive inflammation, is a common model of acute kidney injury (AKI). Pyroptosis, a pro-inflammatory form of cell death due to the activation of inflammatory caspases, is involved in AKI progression. Ibudilast, a TLR4 antagonist, has been used in the clinic to exert an anti-inflammatory effect on asthma. However, researchers have not explored whether ibudilast exerts a protective effect on AKI by inhibiting inflammation. In the present study, ibudilast reversed FA-induced AKI in mice, as indicated by the reduced serum creatinine and urea nitrogen levels, and improved renal pathology, as well as the downregulation of kidney injury marker-1. In addition, ibudilast significantly increased the production of the anti-inflammatory factor IL-10 while suppressing the secretion of the pro-inflammatory cytokine TNF-α and macrophage infiltration. Moreover, in the injured kidney, ibudilast reduced the levels of both inflammasome markers (NLRP3) and pyroptosis-related proteins (caspase-1, IL1-β, IL-18, and GSDMD cleavage), and decreased the number of TUNEL-positive cells. Further mechanistic studies showed that ibudilast administration inhibited the FA-induced upregulation of TLR4, blocked NF-κB nuclear translocation, and reduced the phosphorylation of NF-κB and IκBα, p38, ERK, and JNK. Thus, this study substantiates the protective effect of ibudilast on FA-induced AKI in mice and suggests that protection might be achieved by reducing pyroptosis and inflammation, likely through the inhibition of TLR4-mediated NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Xue Li
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China.,Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Zou
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yuan-Yuan Fu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Jia Xing
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Kai-Yue Wang
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Peng-Zhi Wan
- Department of Nephrology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mo Wang
- Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Xiao-Yue Zhai
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China.,Institute of Nephropathology, China Medical University, Shenyang, China
| |
Collapse
|
15
|
Nephrotoxicity of Herbal Products in Europe-A Review of an Underestimated Problem. Int J Mol Sci 2021; 22:ijms22084132. [PMID: 33923686 PMCID: PMC8074082 DOI: 10.3390/ijms22084132] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Currently in Europe, despite the many advances in production technology of synthetic drugs, the interest in natural herbal medicines continues to increase. One of the reasons for their popular use is the assumption that natural equals safe. However, herbal medicines contain pharmacologically active ingredients, some of which have been associated with adverse effects. Kidneys are particularly susceptible to injury induced by toxins, including poisonous constituents from medicinal plants. The most recognized herb-induced kidney injury is aristolochic acid nephropathy connected with misuse of certain Traditional Chinese herbal medicines. Data concerning nephrotoxicity of plant species of European origin are scarce. Here, we critically review significant data of the nephrotoxicity of several plants used in European phytotherapy, including Artemisia herba-alba, Glycyrrhiza glabra, Euphorbia paralias, and Aloe). Causative mechanisms and factors predisposing to intoxications from the use of herbs are discussed. The basic intention of this review is to improve pharmacovigilance of herbal medicine, especially in patients with chronic kidney diseases.
Collapse
|
16
|
Wang L, Man S, Bian Y. Bioinformatics analysis of biomarkers of aristolochic acid-induced early nephrotoxicity in embryonic stem cells. Exp Ther Med 2021; 21:508. [PMID: 33791017 PMCID: PMC8005694 DOI: 10.3892/etm.2021.9939] [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: 05/06/2020] [Accepted: 01/25/2021] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to identify key genes as potential biomarkers for early nephrotoxicity induced by aristolochic acid (AA) in embryonic stem cells (ESCs). An MTT assay was performed to determine the cytotoxicity of AA in ESCs. Differentially expressed genes (DEGs) were identified using the DNA-Chip Analyzer following microarray analysis. Gene Ontology analysis was performed to determine functional terms enriched by the DEGs in the categories biological process, cellular component and molecular function. Furthermore, the DEGs were subjected to Kyoto Encyclopedia of Genes and Genomes analysis to determine pathways they were accumulated in. Furthermore, a protein-protein interaction network was constructed using Cytoscape 3.2 software. Tumor protein 53 apoptosis effector (Perp), cation transport regulator-like 1 (Chac1), adrenoceptor β2 and Wnt6 were selected for confirmation by reverse transcription-quantitative (RT-q) PCR analysis. A total of 72 DEGs (49 upregulated and 23 downregulated) were identified. The DEGs were enriched in functional terms and pathways associated with nephrotoxicity and participated in 92 pathways. A total of two hub genes, fructose-1,6-bisphosphatase (Fbp)1 and Fbp2, were filtered out from the interaction network. Perp and phorbol-12-myristate-13-acetate-induced protein 1 were demonstrated to have vital roles in the p53 signaling pathway which was indicated in the interaction network. The results of the RT-qPCR analysis were consistent with the microarray data. Taken together, the present study suggested that hub genes involved in the p53 pathway, including Fbp1, Fbp2 and Perp, may serve as potential biomarkers for early nephrotoxicity induced by AA.
Collapse
Affiliation(s)
- Li Wang
- Pharmaceutical Sector, Tianjin Second People's Hospital, Tianjin Institute of Liver Disease, Tianjin 300192, P.R. China
| | - Shanshan Man
- Pharmaceutical Sector, Tianjin Second People's Hospital, Tianjin Institute of Liver Disease, Tianjin 300192, P.R. China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| |
Collapse
|
17
|
Koc K, Geyikoglu F, Cakmak O, Koca A, Kutlu Z, Aysin F, Yilmaz A, Aşkın H. The targets of β-sitosterol as a novel therapeutic against cardio-renal complications in acute renal ischemia/reperfusion damage. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:469-479. [PMID: 33048170 DOI: 10.1007/s00210-020-01984-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 09/30/2020] [Indexed: 01/17/2023]
Abstract
This research is the first to use β-sitosterol on myocardial and renal tissues in renal ischemia/reperfusion (IR) damage. Female Wistar rats were randomly divided into three groups: control (sham), renal IR (50 min ischemia - 3 h reperfusion), and renal IR + 150 mg/kg/p.o. β-sitosterol (the rats were treated with β-sitosterol orally once 1 h before the IR procedure). β-Sitosterol pretreatment caused an increase in superoxide dismutase and glutathione activities and a decrease in malondialdehyde levels in the kidney and heart. Moreover, it alleviated histopathological changes and downregulated the levels of tumor necrosis factor-alpha and interleukin-6 and upregulated the levels of endothelial nitric oxide synthase. As conclusion, the potential of β-sitosterol for renal and cardiac necrosis and apoptosis appears to act by limiting inflammatory response and oxidative stress. Thus, the potential of this compound is noteworthy and may serve as a potential therapeutic in the treatment of acute organ damages due to renal IR.
Collapse
Affiliation(s)
- Kubra Koc
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Fatime Geyikoglu
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Ozge Cakmak
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey.
| | - Aynur Koca
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Zerrin Kutlu
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - Ferhunde Aysin
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
- East Anatolian High Technology Research and Application Center (DAYTAM), Ataturk University, Erzurum, Turkey
| | - Asli Yilmaz
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
- East Anatolian High Technology Research and Application Center (DAYTAM), Ataturk University, Erzurum, Turkey
| | - Hakan Aşkın
- Department of Molecular Biology and Genetics, Faculty of Science, Ataturk University, Erzurum, Turkey
| |
Collapse
|
18
|
Rui-Zhi T, Hui D, Jian-Chun L, Xia Z, Xiao-Jia W, Dan W, Jun-Ming F, Li W. Astragalus mongholicus Bunge and Panax Notoginseng Formula (A&P) Combined With Bifidobacterium Contribute a Renoprotective Effect in Chronic Kidney Disease Through Inhibiting Macrophage Inflammatory Response in Kidney and Intestine. Front Physiol 2020; 11:583668. [PMID: 33329031 PMCID: PMC7729014 DOI: 10.3389/fphys.2020.583668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
There is increasing evidence that Chronic Kidney Disease (CKD) can cause intestinal dysfunction, which in turn aggravates the progression of kidney disease. Studies have shown that the immune response of macrophage plays an important role in promoting inflammation in kidney and intestine of CKD. Astragalus mongholicus Bunge and Panax notoginseng formula (A&P) is a widely used traditional medicine for the treatment of CKD in China, however, the underlying mechanism is largely unclear. In this study, we aimed to explore the role of A&P and Bifidobacterium combination treatment in regulation of inflammatory response of macrophage in kidney and intestine of CKD mouse, as well as the potential molecular mechanism. We established a CKD mouse model with 5/6 nephrectomy and a macrophage inflammatory cellular model with LPS and urotoxin in vivo and in vitro. The results showed that A&P combined with Bifidobacterium significantly reduced the expression and secretion of IL-1β, IL-6, TNFα, and MCP-1 in kidney and blood, as well as in inflammatory macrophage. Interestingly, A&P combined with Bifidobacterium strongly improved the intestinal flora and protected the intestinal barrier. Notably, the maintainer of macrophage polarization, Mincle, was activated in kidney and intestine of CKD mouse as well as in urotoxin stimulated macrophage, that was effectively inhibited by the treatment of A&P and Bifidobacterium combination. Overexpression of Mincle by genetic modification can abolish the inhibitory effects of A&P combined with Bifidobacterium on inflammation in urotoxin stimulated RAW264.7 cells. In summary, these findings demonstrated that A&P combined with Bifidobacterium can protect kidney against CKD by down-regulating macrophage inflammatory response in kidney and intestine via suppressing Mincle signaling, which provides a new insight in the treatment of CKD with traditional medicine.
Collapse
Affiliation(s)
- Tan Rui-Zhi
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Diao Hui
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China.,Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li Jian-Chun
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Zhong Xia
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Wang Xiao-Jia
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Wen Dan
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China.,Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fan Jun-Ming
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China.,Department of Nephrology, Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wang Li
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
19
|
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: 5] [Impact Index Per Article: 1.0] [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
|
20
|
Sellmayr M, Hernandez Petzsche MR, Ma Q, Krüger N, Liapis H, Brink A, Lenz B, Angelotti ML, Gnemmi V, Kuppe C, Kim H, Bindels EMJ, Tajti F, Saez-Rodriguez J, Lech M, Kramann R, Romagnani P, Anders HJ, Steiger S. Only Hyperuricemia with Crystalluria, but not Asymptomatic Hyperuricemia, Drives Progression of Chronic Kidney Disease. J Am Soc Nephrol 2020; 31:2773-2792. [PMID: 32938648 DOI: 10.1681/asn.2020040523] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The roles of asymptomatic hyperuricemia or uric acid (UA) crystals in CKD progression are unknown. Hypotheses to explain links between UA deposition and progression of CKD include that (1) asymptomatic hyperuricemia does not promote CKD progression unless UA crystallizes in the kidney; (2) UA crystal granulomas may form due to pre-existing CKD; and (3) proinflammatory granuloma-related M1-like macrophages may drive UA crystal-induced CKD progression. METHODS MALDI-FTICR mass spectrometry, immunohistochemistry, 3D confocal microscopy, and flow cytometry were used to characterize a novel mouse model of hyperuricemia and chronic UA crystal nephropathy with granulomatous nephritis. Interventional studies probed the role of crystal-induced inflammation and macrophages in the pathology of progressive CKD. RESULTS Asymptomatic hyperuricemia alone did not cause CKD or drive the progression of aristolochic acid I-induced CKD. Only hyperuricemia with UA crystalluria due to urinary acidification caused tubular obstruction, inflammation, and interstitial fibrosis. UA crystal granulomas surrounded by proinflammatory M1-like macrophages developed late in this process of chronic UA crystal nephropathy and contributed to the progression of pre-existing CKD. Suppressing M1-like macrophages with adenosine attenuated granulomatous nephritis and the progressive decline in GFR. In contrast, inhibiting the JAK/STAT inflammatory pathway with tofacitinib was not renoprotective. CONCLUSIONS Asymptomatic hyperuricemia does not affect CKD progression unless UA crystallizes in the kidney. UA crystal granulomas develop late in chronic UA crystal nephropathy and contribute to CKD progression because UA crystals trigger M1-like macrophage-related interstitial inflammation and fibrosis. Targeting proinflammatory macrophages, but not JAK/STAT signaling, can attenuate granulomatous interstitial nephritis.
Collapse
Affiliation(s)
- Markus Sellmayr
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilian's-University Hospital, Munich, Germany
| | | | - Qiuyue Ma
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilian's-University Hospital, Munich, Germany
| | - Nils Krüger
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilian's-University Hospital, Munich, Germany
| | - Helen Liapis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri (retired) and Arkana Laboratories, Little Rock, Arkansas
| | - Andreas Brink
- Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Barbara Lenz
- Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Maria Lucia Angelotti
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
| | - Viviane Gnemmi
- Department of Pathology, University Hospital, Centre Hospitalier Régional Universitaire, Lille, France
| | - Christoph Kuppe
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Hyojin Kim
- Faculty of Medicine, Rheinisch-Westfälische Technische Hochschule, Aachen University, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), Aachen, Germany
| | | | - Ferenc Tajti
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany.,Faculty of Medicine, Rheinisch-Westfälische Technische Hochschule, Aachen University, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), Aachen, Germany
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Rheinisch-Westfälische Technische Hochschule, Aachen University, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), Aachen, Germany.,Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University, and Heidelberg University Hospital, Heidelberg, Germany
| | - Maciej Lech
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilian's-University Hospital, Munich, Germany
| | - Rafael Kramann
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany.,Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy.,Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilian's-University Hospital, Munich, Germany
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilian's-University Hospital, Munich, Germany
| |
Collapse
|
21
|
Kashiwazaki D, Maruyama K, Yamamoto S, Saito H, Akioka N, Kuwayama N, Noguchi K, Kuroda S. Unstable Carotid Plaque as a Phenotype of Chronic Systemic Inflammation Enhances Renal Insufficiency. J Stroke Cerebrovasc Dis 2020; 29:104698. [PMID: 32093991 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/11/2020] [Accepted: 01/25/2020] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Chronic systemic inflammation is prevalent in patients with chronic kidney disease (CKD) and is linked to the development of cerebrovascular disease. In this study, we explored the association between the unstable plaques and preoperative CKD in patients who underwent carotid endarterectomy (CEA)/carotid artery stenting (CAS). Furthermore, this study also aimed to explore whether there is a difference in the aggravation of renal function with the presence of stable or unstable plaques. PATIENTS AND METHODS The study included 90 patients who underwent CEA/CAS for carotid artery stenosis. Multivariate analysis was performed to determine the risk factors for CKD. Kaplan-Meier estimation was employed to evaluate the aggravation of renal function following CEA/CAS. RESULTS Multivariate analysis revealed that contralateral carotid occlusive disease (odds ratio [OR], 4.45; 95% confidence interval [CI], 1.36-14.6), and T1 high-intensity carotid plaque (OR, 3.26; 95% CI, 1.2-8.86) were independent factors of CKD. Kaplan-Meier estimation demonstrated a higher aggravation of renal function in the T1 high-intensity group compared to those in the iso-intensity (P =.03). Following CEA/CAS, the time until aggravation of renal insufficiency was 12.0 ± 9.4 months in the T1 high-intensity group and 24.5 ± 9.6 months in the iso-intensity group (P =.03). CONCLUSIONS This study demonstrated that contralateral carotid artery stenosis and T1 high-intensity plaques are more frequently observed in patients with CKD. T1 high-intensity carotid plaque is well linked to CKD development in future.
Collapse
Affiliation(s)
- Daina Kashiwazaki
- Departments of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| | - Kunitaka Maruyama
- Departments of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Shusuke Yamamoto
- Departments of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Hisayasu Saito
- Departments of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Naoki Akioka
- Departments of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Naoya Kuwayama
- Departments of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Kyo Noguchi
- Departments of Radiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Satoshi Kuroda
- Departments of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
22
|
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: 49] [Impact Index Per Article: 9.8] [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
|
23
|
Kim JY, Leem J, Jeon EJ. Protective Effects of Melatonin Against Aristolochic Acid-Induced Nephropathy in Mice. Biomolecules 2019; 10:biom10010011. [PMID: 31861726 PMCID: PMC7023369 DOI: 10.3390/biom10010011] [Citation(s) in RCA: 36] [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: 11/27/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 11/16/2022] Open
Abstract
Melatonin, a pineal hormone, is well known to regulate the sleep–wake cycle. Besides, the hormone has been shown to display pleiotropic effects arising from its powerful anti-oxidant and anti-inflammatory activities. Recent studies have reported that melatonin exerts protective effects in animal models of kidney disease. However, the potential effects of melatonin on aristolochic acid (AA)-induced nephropathy (AAN) have not yet been investigated. Here, we found that the administration of melatonin ameliorated AA-induced renal dysfunction, as evidenced by decreased plasma levels of blood urea nitrogen and creatinine and histopathological abnormalities such as tubular dilatation and cast formation. The upregulation of tubular injury markers after AA injection was reversed by melatonin. Melatonin also suppressed AA-induced oxidative stress, as evidenced by the downregulation of 4-hydroxynonenal and reduced level of malondialdehyde, and modulated expression of pro-oxidant and antioxidant enzymes. In addition, p53-dependent apoptosis of tubular epithelial cells, infiltration of macrophages and CD4+ T cells into damaged kidneys, and renal expression of cytokines and chemokines were inhibited by melatonin. Moreover, melatonin attenuated AA-induced tubulointerstitial fibrosis through suppression of the tumor growth factor-β/Smad signaling pathway. These results suggest that melatonin might be a potential therapeutic agent for AAN.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
- Correspondence: (J.L.); (E.J.J.); Tel.: +82-053-650-3612 (J.L.); +82-053-650-4214 (E.J.J.)
| | - Eon Ju Jeon
- Department of Internal Medicine, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea
- Correspondence: (J.L.); (E.J.J.); Tel.: +82-053-650-3612 (J.L.); +82-053-650-4214 (E.J.J.)
| |
Collapse
|
24
|
Sborchia M, Keun HC, Phillips DH, Arlt VM. The Impact of p53 on Aristolochic Acid I-Induced Gene Expression In Vivo. Int J Mol Sci 2019; 20:ijms20246155. [PMID: 31817608 PMCID: PMC6940885 DOI: 10.3390/ijms20246155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to aristolochic acid (AA) is linked to kidney disease and urothelial cancer in humans. The major carcinogenic component of the AA plant extract is aristolochic acid I (AAI). The tumour suppressor p53 is frequently mutated in AA-induced tumours. We previously showed that p53 protects from AAI-induced renal proximal tubular injury, but the underlying mechanism(s) involved remain to be further explored. In the present study, we investigated the impact of p53 on AAI-induced gene expression by treating Trp53(+/+), Trp53(+/-), and Trp53(-/-) mice with 3.5 mg/kg body weight (bw) AAI daily for six days. The Clariom™ S Assay microarray was used to elucidate gene expression profiles in mouse kidneys after AAI treatment. Analyses in Qlucore Omics Explorer showed that gene expression in AAI-exposed kidneys is treatment-dependent. However, gene expression profiles did not segregate in a clear-cut manner according to Trp53 genotype, hence further investigations were performed by pathway analysis with MetaCore™. Several pathways were significantly altered to varying degrees for AAI-exposed kidneys. Apoptotic pathways were modulated in Trp53(+/+) kidneys; whereas oncogenic and pro-survival pathways were significantly altered for Trp53(+/-) and Trp53(-/-) kidneys, respectively. Alterations of biological processes by AAI in mouse kidneys could explain the mechanisms by which p53 protects from or p53 loss drives AAI-induced renal injury in vivo.
Collapse
Affiliation(s)
- Mateja Sborchia
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
| | - Hector C. Keun
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London W12 0NN, UK;
| | - David H. Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
| | - Volker M. Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King’s College London, London SE1 9NH, UK; (M.S.); (D.H.P.)
- Correspondence:
| |
Collapse
|