1
|
Zhou Q, Jiang L, Su T, Liu G, Yang L. Overview of aristolochic acid nephropathy: an update. Kidney Res Clin Pract 2023; 42:579-590. [PMID: 37448287 PMCID: PMC10565449 DOI: 10.23876/j.krcp.22.211] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/31/2022] [Accepted: 01/16/2023] [Indexed: 07/15/2023] Open
Abstract
Aristolochic acid nephropathy (AAN) is a rapidly progressive renal interstitial fibrosis caused by medical or environmental exposure to aristolochic acid (AA). Since the outbreak of AAN in Belgium was reported nearly 30 years ago, the safety of herbal remedies has drawn considerable attention, and AAN has become a global public health problem. Breakthroughs have been made to better understand the disease, including the toxicity of AAs, the possible mechanisms of AAN, the disease patterns, and the pathological features; however, some critical problems remain unresolved. Because of the insidious onset of the disease, the incidence of AAN and the prevalence of exposure to AAs are unknown and might be largely underestimated. During the past decades, AA-containing herbs have been strictly administrated in many regions and the occurrence of AAN has declined sharply, yet cases of AAN are still sporadically reported. Despite the progress in the understanding of the disease's pathogenesis, there is no effective treatment for delaying or reversing the renal deterioration caused by AAN. Therefore, the risk of exposure to AAs should be taken seriously by public health workers and clinicians. In this review, we updated the latest data on AAN, summarized the advances throughout these years, and put forward some challenges for future research.
Collapse
Affiliation(s)
- Qingqing Zhou
- Division of Renal, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
| | - Lei Jiang
- Division of Renal, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Su
- Division of Renal, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Liu
- Division of Renal, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Yang
- Division of Renal, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2
|
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
|
3
|
Baek S, Jang MG, Kim JW, Ko HC, Nam MH, Hur SP, Park SA, Kim SJ. Polymethoxyflavone-rich Fraction from Citrus sunki Leaves Alleviates Renal Dysfunction in Mice with Unilateral Ureteral Obstruction. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221109412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Polymethoxyflavones (PMFs) are flavonoid compounds present in citrus plants that are proposed to be advantageous to human health. However, the advantageous effects of PMFs in the context of renal dysfunction are unclear. In this study, we made a PMF-rich fraction (PRF) from the leaves of Citrus sunki Hort ex. Tanaka and identified its components using liquid chromatography and mass spectrometry. We then investigated the effect of PRF—comprising 9 types of PMF—on renal dysfunction induced by unilateral ureteral obstruction (UUO) in mice. Animals were divided into four experimental groups ( n = 7 per group): I) sham-operated group (Sham); II) UUO group (UUO); III) UUO + Enalapril 0.1 mg/1 mL (UUO + Enap); IV) UUO + PRF 100 mg/kg/day (UUO + PRF). All mice were orally administered with the drugs once a day from 7 days before UUO to 1 week after UUO. After the experiments were over, serum and tissues were taken for biochemical and histological analysis. PRF promoted the recovery of body weight in the background of UUO. Biochemical and histological analysis revealed that PRF ameliorated UUO-induced renal dysfunction and moderately reversed inflammation and tubulointerstitial fibrosis. Further, PRF inhibited the expression of endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), transforming growth factor-β (TGF-β), collagen I (Col-I), and collagen IV (Col-IV). These results suggest that PRF improves UUO-induced renal dysfunction by regulating the expression of inflammatory and fibrotic response-related genes.
Collapse
Affiliation(s)
- Songyee Baek
- Department of Biology, Jeju National University, Jeju, Korea
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| | - Mi Gyeong Jang
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| | - Jae-Won Kim
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| | - Hee Chul Ko
- Jeju Institute of Korean Medicine, Jeju, Korea
| | - Mi Hyun Nam
- Jeju Institute of Korean Medicine, Jeju, Korea
| | - Sung-Pyo Hur
- Korea Institute of Ocean Science & Technology, Jeju, Korea
| | - Soo Ah Park
- In Vivo Research Center, Central Research Facilities, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Se-Jae Kim
- Department of Biology, Jeju National University, Jeju, Korea
- Biotech Regional Innovation Center, Jeju National University, Jeju, Korea
| |
Collapse
|
4
|
Tsukamoto S, Wakui H, Azushima K, Yamaji T, Urate S, Suzuki T, Abe E, Tanaka S, Taguchi S, Yamada T, Kinguchi S, Kamimura D, Yamashita A, Sano D, Nakano M, Hashimoto T, Tamura K. Tissue-specific expression of the SARS-CoV-2 receptor, angiotensin-converting enzyme 2, in mouse models of chronic kidney disease. Sci Rep 2021; 11:16843. [PMID: 34413390 PMCID: PMC8377123 DOI: 10.1038/s41598-021-96294-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Elevated angiotensin-converting enzyme 2 (ACE2) expression in organs that are potential targets of severe acute respiratory syndrome coronavirus 2 may increase the risk of coronavirus disease 2019 (COVID-19) infection. Previous reports show that ACE2 alter its tissue-specific expression patterns under various pathological conditions, including renal diseases. Here, we examined changes in pulmonary ACE2 expression in two mouse chronic kidney disease (CKD) models: adenine-induced (adenine mice) and aristolochic acid-induced (AA mice). We also investigated changes in pulmonary ACE2 expression due to renin-angiotensin system (RAS) blocker (olmesartan) treatment in these mice. Adenine mice showed significant renal functional decline and elevated blood pressure, compared with controls. AA mice also showed significant renal functional decline, compared with vehicles; blood pressure did not differ between groups. Renal ACE2 expression was significantly reduced in adenine mice and AA mice; pulmonary expression was unaffected. Olmesartan attenuated urinary albumin excretion in adenine mice, but did not affect renal or pulmonary ACE2 expression levels. The results suggest that the risk of COVID-19 infection may not be elevated in patients with CKD because of their stable pulmonary ACE2 expression. Moreover, RAS blockers can be used safely in treatment of COVID-19 patients with CKD.
Collapse
Affiliation(s)
- Shunichiro Tsukamoto
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takahiro Yamaji
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
| | - Shingo Urate
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Toru Suzuki
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Eriko Abe
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Shohei Tanaka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Shinya Taguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Takayuki Yamada
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Department of Medicine, Mount Sinai Beth Israel, New York, NY, USA
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Daisuke Kamimura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Akio Yamashita
- Department of Molecular Biology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Daisuke Sano
- Department of Otorhinolaryngology, Head and Neck Surgery, School of Medicine, Yokohama City University, Yokohama, Japan
| | - Masayuki Nakano
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Tatsuo Hashimoto
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.,Internal Medicine, Kanagawa Dental University, Yokosuka, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
5
|
The transcription factor Twist1 in the distal nephron but not in macrophages propagates aristolochic acid nephropathy. Kidney Int 2019; 97:119-129. [PMID: 31685313 DOI: 10.1016/j.kint.2019.07.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 07/02/2019] [Accepted: 07/11/2019] [Indexed: 11/20/2022]
Abstract
Tubulointerstitial disease in the kidney culminates in renal fibrosis that portents organ failure. Twist1, a basic helix-loop-helix protein 38 transcription factor, regulates several essential biological functions, but inappropriate Twist1 activity in the kidney epithelium can trigger kidney fibrogenesis and chronic kidney disease. By contrast, Twist1 in circulating myeloid cells may constrain inflammatory injury by attenuating cytokine generation. To dissect the effects of Twist1 in kidney tubular versus immune cells on renal inflammation following toxin-induced renal injury, we subjected mice with selective deletion of Twist1 in renal epithelial cells or macrophages to aristolochic acid-induced chronic kidney disease. Ablation of Twist1 in the distal nephron attenuated kidney damage, interstitial fibrosis, and renal inflammation after aristolochic acid exposure. However, macrophage-specific deletion of Twist1 did not impact the development of aristolochic acid-induced nephropathy. In vitro studies confirmed that Twist1 in renal tubular cells underpins their susceptibility to apoptosis and propensity to generate pro-fibrotic mediators in response to aristolochic acid. Moreover, co-culture studies revealed that Twist1 in renal epithelia augmented the recruitment and activation of pro-inflammatory CD64+ macrophages. Thus, Twist1 in the distal nephron rather than in infiltrating macrophages propagates chronic inflammation and fibrogenesis during aristolochic acid-induced nephropathy.
Collapse
|
6
|
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: 134] [Impact Index Per Article: 19.1] [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
|
7
|
Yuen DA, Huang YW, Liu GY, Patel S, Fang F, Zhou J, Thai K, Sidiqi A, Szeto SG, Chan L, Lu M, He X, John R, Gilbert RE, Scholey JW, Robinson LA. Recombinant N-Terminal Slit2 Inhibits TGF-β-Induced Fibroblast Activation and Renal Fibrosis. J Am Soc Nephrol 2016; 27:2609-15. [PMID: 26869008 DOI: 10.1681/asn.2015040356] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 01/12/2016] [Indexed: 12/31/2022] Open
Abstract
Fibrosis and inflammation are closely intertwined injury pathways present in nearly all forms of CKD for which few safe and effective therapies exist. Slit glycoproteins signaling through Roundabout (Robo) receptors have been described to have anti-inflammatory effects through regulation of leukocyte cytoskeletal organization. Notably, cytoskeletal reorganization is also required for fibroblast responses to TGF-β Here, we examined whether Slit2 also controls TGF-β-induced renal fibrosis. In cultured renal fibroblasts, which we found to express Slit2 and Robo-1, the bioactive N-terminal fragment of Slit2 inhibited TGF-β-induced collagen synthesis, actin cytoskeletal reorganization, and Smad2/3 transcriptional activity, but the inactive C-terminal fragment of Slit2 did not. In mouse models of postischemic renal fibrosis and obstructive uropathy, treatment with N-terminal Slit2 before or after injury inhibited the development of renal fibrosis and preserved renal function, whereas the C-terminal Slit2 had no effect. Our data suggest that administration of recombinant Slit2 may be a new treatment strategy to arrest chronic injury progression after ischemic and obstructive renal insults by not only attenuating inflammation but also, directly inhibiting renal fibrosis.
Collapse
Affiliation(s)
- Darren A Yuen
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada; Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada; Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - Yi-Wei Huang
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Guang-Ying Liu
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Sajedabanu Patel
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | | | | | - Kerri Thai
- Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Ahmad Sidiqi
- Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Stephen G Szeto
- Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Lauren Chan
- Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mingliang Lu
- Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Xiaolin He
- Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Rohan John
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto, Ontario, Canada
| | - Richard E Gilbert
- Keenan Research Centre of Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - James W Scholey
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and Division of Nephrology and
| | - Lisa A Robinson
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada; Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
8
|
Antoine MH, Debelle F, Piccirilli J, El Kaddouri F, Declèves AE, De Prez E, Husson C, Mies F, Bourgeade MF, Nortier JL. Human bone morphogenetic protein-7 does not counteract aristolochic acid-induced renal toxicity. J Appl Toxicol 2015; 35:1520-30. [PMID: 25663515 DOI: 10.1002/jat.3116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 11/08/2022]
Abstract
Aristolochic acids (AA) are nephrotoxic and profibrotic agents, leading to chronic kidney disease. As some controversial studies have reported a nephroprotective effect of exogenous recombinant human bone morphogenetic protein (rhBMP)-7 in several models of renal fibrosis, we investigated the putative effect of rhBMP-7 to prevent progressive tubulointerstitial damage after AA intoxication in vitro and in vivo. In vitro, the toxicity of AA on renal tubular cells was demonstrated by an increase in vimentin as well as a decrease in β-catenin expressions, reflecting a dedifferentiation process. Increased fibronectin and interleukin-6 levels were measured in the supernatants. Enhanced α-SMA mRNA levels associated to decreased E-cadherin mRNA levels were also measured. Incubation with rhBMP-7 only prevented the increase in vimentin and the decrease in β-catenin expressions. In vivo, in a rat model of AA nephropathy, severe tubulointerstitial lesions induced by AA after 10 and 35 days (collagen IV deposition and tubular atrophy), were not prevented by the rhBMP-7 treatment. Similarly, rhBMP-7 did not ameliorate the significant increase in urinary concentrations of transforming growth factor-β. In summary, our in vitro data demonstrated a poor beneficial effect of rhBMP-7 to reverse cell toxicity while, in vivo, there was no beneficial effect of rhBMP-7. Therefore, further investigations are needed to confirm the exact role of BMP-7 in progressive chronic kidney disease.
Collapse
Affiliation(s)
- Marie-Hélène Antoine
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Frédéric Debelle
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Julie Piccirilli
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Fadoua El Kaddouri
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Anne-Emilie Declèves
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels.,Laboratory of Molecular Physiology (URPhyM), University of Namur, Namur
| | - Eric De Prez
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Cécile Husson
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| | - Frédérique Mies
- Laboratory of Physiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Joëlle L Nortier
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles, Brussels
| |
Collapse
|
9
|
|
10
|
Tsai KD, Chen W, Wang SH, Hsiao YW, Chi JY, Wu HY, Lee YJ, Wong HY, Tseng MJ, Lin TH. Downregulation of connective tissue growth factor by LPS/IFN-γ-induced nitric oxide is reversed by aristolochic acid treatment in glomerular mesangial cells via STAT-1α and NF-κB signaling. Chem Biol Interact 2014; 210:86-95. [PMID: 24412304 DOI: 10.1016/j.cbi.2013.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 12/09/2013] [Accepted: 12/30/2013] [Indexed: 01/14/2023]
Abstract
Aristolochic acid (AA) is a common cause of Chinese herb nephropathy. The mechanisms involved in the pathogenesis of AA nephropathy (AAN) are intricate. One well-documented effect of AA in the kidney is its pro-fibrotic activity. Nitric oxide (NO), a messenger gas generated from l-arginine, is the product of nitric oxide synthase (NOS). NO is involved in renal hemodynamics and exerts cytoprotective effects against renal injury. In the present study, the role of NO in AAN was investigated in MES-13 cells, a glomerular mesangial cell line. NO endogenously generated by the induction of inducible nitric oxide synthase (iNOS) with lipopolysaccharide (LPS)/interferon-γ (IFN-γ) significantly downregulated connective tissue growth factor (CTGF) protein expression in MES-13 cells. AA significantly suppressed LPS/IFN-γ-induced NO production and reversed CTGF expression that was downregulated by LPS/IFN-γ. AA decreased iNOS gene and protein expressions in a concentration-dependent manner. AA caused declines in LPS/IFN-γ-induced signal transducer and activator of transcription-1α (STAT-1α) phosphorylation and interferon response factor-1 (IRF-1) mRNA expression. Furthermore, AA attenuated IκB phosphorylation and reduced NF-κB translocation to the nuclear fraction. Taken together, our data indicate that AA reversed the CTGF expression inhibited by LPS/IFN-γ treatment via suppression of NO and iNOS expressions in MES-13 cells through inhibition of the JAK/STAT-1α and NF-κB signaling pathways. NO potentially exerts antifibrotic activity by down regulation of CTGF in MES-13 cells and inhibition of the iNOS gene by AA might partially account for the fibrotic effects of AA in nephropathy.
Collapse
Affiliation(s)
- Kuen-Daw Tsai
- Department of Internal Medicine, China Medical University and Beigang Hospital, 123, Sinde Road, Beigang Township, Yunlin County 65152, Taiwan, ROC; Institute of Molecular Biology, National Chung Cheng University, 168 University Road, Minhsiung Township, Chiayi County 62102, Taiwan, ROC
| | - Wei Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chia-Yi Christian Hospital, Chiayi, Taiwan, ROC
| | - Sue-Hong Wang
- Department of Biomedical Sciences, Chung Shan Medical University, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC; Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC
| | - Yu-Wei Hsiao
- Department of Biomedical Sciences, Chung Shan Medical University, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC; Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC
| | - Jhih-Ying Chi
- Department of Biomedical Sciences, Chung Shan Medical University, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC; Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC
| | - Hsing-Yu Wu
- Department of Biomedical Sciences, Chung Shan Medical University, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC; Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC
| | - Yi-Ju Lee
- Institute of Microbiology and Immunology, Chung Shan Medical University, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC
| | - Ho-Yiu Wong
- Department of Internal Medicine, China Medical University and Beigang Hospital, 123, Sinde Road, Beigang Township, Yunlin County 65152, Taiwan, ROC
| | - Min-Jen Tseng
- Institute of Molecular Biology, National Chung Cheng University, 168 University Road, Minhsiung Township, Chiayi County 62102, Taiwan, ROC
| | - Ting-Hui Lin
- Department of Biomedical Sciences, Chung Shan Medical University, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC; Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Jianguo North Road, Section 1, Taichung 40203, Taiwan, ROC.
| |
Collapse
|
11
|
Novitskaya T, McDermott L, Zhang KX, Chiba T, Paueksakon P, Hukriede NA, de Caestecker MP. A PTBA small molecule enhances recovery and reduces postinjury fibrosis after aristolochic acid-induced kidney injury. Am J Physiol Renal Physiol 2013; 306:F496-504. [PMID: 24370591 DOI: 10.1152/ajprenal.00534.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Phenylthiobutanoic acids (PTBAs) are a new class of histone deacetylase (HDAC) inhibitors that accelerate recovery and reduce postinjury fibrosis after ischemia-reperfusion-induced acute kidney injury. However, unlike the more common scenario in which patients present with protracted and less clearly defined onset of renal injury, this model of acute kidney injury gives rise to a clearly defined injury that begins to resolve over a short period of time. In these studies, we show for the first time that treatment with the PTBA analog methyl-4-(phenylthio)butanoate (M4PTB) accelerates recovery and reduces postinjury fibrosis in a progressive model of acute kidney injury and renal fibrosis that occurs after aristolochic acid injection in mice. These effects are apparent when M4PTB treatment is delayed 4 days after the initiating injury and are associated with increased proliferation and decreased G2/M arrest of regenerating renal tubular epithelial cells. In addition, there is reduced peritubular macrophage infiltration and decreased expression of the macrophage chemokines CX3Cl1 and CCL2. Since macrophage infiltration plays a role in promoting kidney injury, and since renal tubular epithelial cells show defective repair and a marked increase in maladaptive G2/M arrest after aristolochic acid injury, these findings suggest M4PTB may be particularly beneficial in reducing injury and enhancing intrinsic cellular repair even when administered days after aristolochic acid ingestion.
Collapse
Affiliation(s)
- Tatiana Novitskaya
- Vanderbilt Univ. Medical Center, Dept. of Medicine, Div. of Nephrology, S3223 Medical Center North, 1161 21st Ave. South, Nashville, TN 37232.
| | | | | | | | | | | | | |
Collapse
|
12
|
Zhou Y, Bian X, Fang L, He W, Dai C, Yang J. Aristolochic acid causes albuminuria by promoting mitochondrial DNA damage and dysfunction in podocyte. PLoS One 2013; 8:e83408. [PMID: 24349506 PMCID: PMC3862730 DOI: 10.1371/journal.pone.0083408] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/12/2013] [Indexed: 02/02/2023] Open
Abstract
Aristolochic acid nephropathy, initially found in patients intaking of slimming herbs containing aristolochic acid (AA), was previously considered as a progressive renal interstitial fibrosis and urothelial malignancy. However, the presence of albuminuria in some patients with AAN suggests that AA may also damage the glomerular filtration barrier. In this study, mice AAN model was generated by daily administration of aristolochic acid I sodium salt intraperitoneally at a dose of 6 mg/kg body weight for 3 days. All of the mice developed heavy albuminuria at day 3 and 7 after receiving AA. In the mice received AA, morphologic change of glomeruli was minor under light microscopy but podocyte foot-process effacement was evident under electron microscopy. In mitochondria isolated from kidney, prominent mitochondrial DNA (mtDNA) damage was accompanied with marked decrease of mtDNA copy number and mitochondrial protein expression level. Similar to those in vivo results, AA treatment impaired the filtration barrier function of cultured podocytes. AA promoted mtDNA damage, decreased mtDNA copy number and mitochondrial protein expression in cultured podocytes. In addition, AA treatment also decreased ATP content, oxygen consumption rate and mitochondrial membrane potential as well as increased cellular reactive oxygen species in cultured podocytes. This study highlighted that AA could induce podocyte damage and albuminuria, which may be mediated by promoting mtDNA damage and mitochondrial dysfunction in podocytes.
Collapse
Affiliation(s)
- Yang Zhou
- Center for Kidney Disease, 2 Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xueqin Bian
- Center for Kidney Disease, 2 Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Fang
- Center for Kidney Disease, 2 Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weichun He
- Center for Kidney Disease, 2 Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chunsun Dai
- Center for Kidney Disease, 2 Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Junwei Yang
- Center for Kidney Disease, 2 Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
- * E-mail:
| |
Collapse
|
13
|
Fuchs TC, Mally A, Wool A, Beiman M, Hewitt P. An Exploratory Evaluation of the Utility of Transcriptional and Urinary Kidney Injury Biomarkers for the Prediction of Aristolochic Acid–Induced Renal Injury in Male Rats. Vet Pathol 2013; 51:680-94. [DOI: 10.1177/0300985813498779] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The predictive value of different urinary and transcriptional biomarkers was evaluated in a proof-of-principle toxicology study in rats using aristolochic acid (AA), a known nephrotoxic agent. Male Wistar rats were orally dosed with 0.1, 1, or 10 mg/kg for 12 days. Urine was collected on days 1, 5, and 12 over 24 hours. Gene expression analysis was also conducted using quantitative real-time polymerase chain reaction and Illumina whole-genome chips. Protein biomarkers (Kim-1, Timp-1, vascular endothelial growth factor, osteopontin, clusterin, cystatin C, calbindin D-28K, β2-microglobulin, α–glutathione S-transferase, GSTY1b, RPA-1, and neutrophil gelatinase-associated lipocalin) were measured in these urine samples. Treatment with AA resulted in a slight dose- and/or time-dependent increase in urinary β2-microglobulin, lipocalin 2, and osteopontin before an increase in serum creatinine or serum urea nitrogen was observed. A strong decrease in urinary calbindin D-28K was also detected. The Compugen Ltd. prediction model scored both the 1- and 10-mg/kg AA dose groups as positive for nephrotoxicity despite the absence of renal histopathological changes. In addition, several previously described transcriptional biomarkers were identified as early predictors of renal toxicity as they were detected before morphological alterations had occurred. Altogether, these findings demonstrated the predictive values of renal biomarkers approved by the Food and Drug Administration, European Medicines Agency, and Pharmaceuticals & Medical Devices Agency in AA-induced renal injury in rats and confirmed the utility of renal transcriptional biomarkers for detecting progression of compound-induced renal injury in rats. In addition, several transcriptional biomarkers identified in this exploratory study could present early predictors of renal tubular epithelium injury in rats.
Collapse
Affiliation(s)
- T. C. Fuchs
- Merck Serono, Non-Clinical Safety, Darmstadt, Germany
| | - A. Mally
- Department of Toxicology, University of Wuerzburg, Wuerzburg, Germany
| | - A. Wool
- Compugen Ltd., Tel Aviv, Israel
| | | | - P. Hewitt
- Merck Serono, Non-Clinical Safety, Darmstadt, Germany
| |
Collapse
|
14
|
Manzetti S, Lu T. The geometry and electronic structure of Aristolochic acid: possible implications for a frozen resonance. J PHYS ORG CHEM 2013. [DOI: 10.1002/poc.3111] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Sergio Manzetti
- Fjordforsk Institute of Science and Technology; Fresvik 6893 Norway
- Science for Life Laboratory, Department of Cell and Molecular Biology; University of Uppsala; Sweden
| | - Tian Lu
- Department of Chemistry and Chemical Engineering, School of Chemical and Biological Engineering; University of Science and Technology Beijing; Beijing 100083 People's Republic of China
| |
Collapse
|
15
|
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: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 06/09/2012] [Indexed: 10/28/2022]
|
16
|
Sun D, Liu CX, Ma YY, Zhang L. Protective effect of prostaglandin E1 on renal microvascular injury in rats of acute aristolochic acid nephropathy. Ren Fail 2011; 33:225-32. [PMID: 21332345 DOI: 10.3109/0886022x.2010.541586] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND To investigate the renal microvascular injury in acute aristolochic acid nephropathy (AAN) and the protective effects of prostaglandin E1 (PGE1) in acute AAN. METHODS Female Sprague-Dawley rats were randomly divided into three groups. The rats in PGE1 group received Caulis Aristolochia manshuriensis (CAM) decoction by gavage for 5 days, and PGE1 was given by vena caudalis before gavage. The rats in model group were gavaged with CAM for 5 days, and the same dose of 0.9% physiologic saline was given by vena caudalis. The rats in control group only received an equal daily volume of saline solution by gavage. Animals were killed at days 3, 5, and 7. Blood urea nitrogen (BUN), serum creatinine, and urinary protein were monitored before killing. Microvascular density was determined by JG12 immunostaining. The expression of angiogenic factor was assessed by vascular endothelial growth factor (VEGF). Tubulointerstitial hypoxia was assessed by hypoxia-inducible factor-1α (HIF-1α) expression. RESULTS CAM induced a significant decrease in VEGF expression and microvascular density in the kidney tissue, accompanied by a significant increase in HIF-1α, which reduced renal function and increased 24-h urinary protein excretion rates. PGE1 lessened the capillary loss, relieved hypoxia, and protected renal function. No significant pathological changes were found in control rats. CONCLUSION The renal microvascular injury in acute AAN is severe. PGE1 can significantly ameliorate the renal microvascular injury, relieve hypoxia, and protect renal function.
Collapse
Affiliation(s)
- Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, PR China.
| | | | | | | |
Collapse
|
17
|
Liu N, Tolbert E, Ponnusamy M, Yan H, Zhuang S. Delayed administration of suramin attenuates the progression of renal fibrosis in obstructive nephropathy. J Pharmacol Exp Ther 2011; 338:758-66. [PMID: 21622732 DOI: 10.1124/jpet.111.181727] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We recently showed that suramin treatment prevents the onset of renal fibrosis in a model of obstructive nephropathy induced by unilateral ureteral obstruction (UUO). In this study, we further assessed the effect of delayed administration of suramin on the progression of tubulointerstitial fibrosis. Mice were given a single dose of suramin at 20 mg/kg starting at day 3 of obstruction, and kidneys were harvested after an additional 7 or 14 days of obstruction. Suramin completely blocked further increase in expression of type I collagen and fibronectin and largely suppressed expression of α-smooth muscle actin (α-SMA) in both treatment groups. UUO injury induced phosphorylation of Smad-3, a key mediator of transforming growth factor-β (TGF-β) signaling, epidermal growth factor receptor, and platelet-derived growth factor receptor after 3 days and further increased at 10 days after UUO injury. When suramin was administered at 3 days after obstruction, phosphorylation of these molecules was not further increased in the obstructed kidney. Suramin treatment also inhibited activation of signal transducer and activator of transcription 3 and extracellular signal-regulated kinase 1 and 2, two signaling pathways associated with renal fibrogenesis. Furthermore, delayed application of suramin suppressed TGF-β1-induced expression of α-SMA and fibronectin in cultured renal interstitial fibroblasts. These results indicate that administration of suramin is effective in attenuating the progression of renal fibrosis after injury and suggest the potential clinical application of suramin as an antifibrotic treatment in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Na Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
18
|
Wu CJ, Chou YC, Cheng YW, Hsiao CJ, Wang CH, Wang HY, Sheu JR, Hsiao G. Aristolochic acid downregulates monocytic matrix metalloproteinase-9 by inhibiting nuclear factor-κB activation. Chem Biol Interact 2011; 192:209-19. [PMID: 21453692 DOI: 10.1016/j.cbi.2011.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 03/15/2011] [Accepted: 03/15/2011] [Indexed: 12/20/2022]
Abstract
Aristolochic acid (AA)-associated nephropathy was described as being characterized by a rapid progressive enhancement of interstitial renal fibrosis. Renal tissue fibrosis occurs because of an imbalance of extracellular matrix (ECM) accumulation and matrix metalloproteinase (MMP) activation. Much evidence indicates that inflammatory renal disease including monocyte and mesangial interactions is linked to the development and progression of renal remodeling. In this study, we found that AA showed concentration-dependent inhibition of tumor necrosis factor (TNF)-α-induced MMP-9 activation with an IC(50) value of 6.4±0.5μM in human monocytic THP-1 cells. A similar effect was also noted with different ratios of AAs (types I and II). However, AA had no inhibitory effect on the intact enzymatic activity of MMP-9 at a concentration of 20μM. On the other hand, the level of tissue inhibitor of metalloproteinase (TIMP)-1 was not induced by AA, but it suppressed TNF-α-induced MMP-9 protein and messenger RNA expressions. AA also significantly inhibited TNF-α-induced IκBα degradation. Furthermore, an electrophoretic mobility shift assay and a reported gene study, respectively, revealed that AA inhibited TNF-α-induced NF-κB translocation and activation. In addition, compared to other NF-κB inhibitors, AA exerted significant inhibition of MMP-9 activation and monocyte chemotactic protein-1-directed invasion. From these results, we concluded that AA, a natural compound, inhibits TNF-α-induced MMP-9 in human monocytic cells possibly through the NF-κB signal pathway. These results also imply that AA may be involved in alteration of matrix homeostasis during renal fibrosis in vivo.
Collapse
Affiliation(s)
- Chih-Jen Wu
- Division of Nephrology, Mackay Memorial Hospital and Mackay Medicine, Nursing and Management College, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Xiao Y, Xue X, Wu YF, Xin GZ, Qian Y, Xie TP, Gong LK, Ren J. beta-Naphthoflavone protects mice from aristolochic acid-I-induced acute kidney injury in a CYP1A dependent mechanism. Acta Pharmacol Sin 2009; 30:1559-65. [PMID: 19890363 DOI: 10.1038/aps.2009.156] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
AIM The role of CYP1A in the protection of aristolochic acid (AA)I-induced nephrotoxicity has been suggested. In the present study we investigated the effects of beta-naphthoflavone (BNF), a non-carcinogen CYP1A inducer, on AAI-induced kidney injury. METHODS Mice were pretreated with 80 mg/kg BNF by daily intraperitoneal injection (ip) for 3 days followed by a single ip of 10 mg/kg AAI. AAI and its major metabolites in blood, liver and kidney, the expression of CYP1A1 and CYP1A2 in microsomes of liver and kidney, as well as the nephrotoxicity were evaluated. RESULTS BNF pretreatment prevented AAI-induced renal damage by facilitating the disposal of AAI in liver. BNF pretreatment induced the expression of CYP1A1 in both liver and kidney; but the induction of CYP1A2 was only observed in liver. CONCLUSION BNF prevents AAI-induced kidney toxicity primarily through CYP1A induction.
Collapse
|
20
|
|
21
|
Ischemic injury underlies the pathogenesis of aristolochic acid-induced acute kidney injury. Transl Res 2008; 152:38-46. [PMID: 18593636 DOI: 10.1016/j.trsl.2008.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 05/14/2008] [Accepted: 05/16/2008] [Indexed: 11/20/2022]
Abstract
Aristolochic acid nephropathy (AAN) is a progressive tubulointerstitial renal disease caused by aristolochic acid intake. To determine the contribution of renal ischemia to the pathogenesis of AAN, we characterized changes in the expression of angiogenic factors and vasoactive substances, and then we evaluated the expression of a marker of hypoxia in an acute AAN rat model. Rats were orally administrated either a decoction of Aristolochiae manshuriensis that contained 20 mg/kg of aristolochic acid-I or an equal volume of distilled water (control group) once daily for 4 days or 7 days. Renal histology and serum creatinine were assessed. Expression of endothelin-1 (ET-1) and hypoxia inducible factor-1 alpha (HIF-1alpha) mRNA within renal cortex were determined by semiquantitative reverse-transcription polymerase chain reaction. Levels of ET-1, nitric oxide (NO), vascular endothelial growth factor (VEGF), and HIF-1alpha in kidneys were determined by radioimmunoassay, Griess method, Western blot, and immunohistochemistry, respectively. Tubular injury scores and ET-1 mRNA expression were increased in the AA-treated rats at both days 4 and 8, whereas serum creatinine level and ET-1 protein expression was increased only at day 4. In contrast, NO production in AA-treated rats was decreased at day 8 compared with the control group. Similarly, VEGF protein expression was reduced in the AA-treated rats at both days 4 and 8. A dramatic increase in nuclear staining for HIF-1alpha was observed mainly in the tubular cells of tubulointerstitial damage area in the AA-treated rats at day 8. The observed increase in HIF-1alpha protein expression, decrease in VEGF protein expression, and imbalance of vasoactive substances after induction of acute kidney injury by AA suggests that ischemic injury contributes to the pathogenesis of AAN.
Collapse
|
22
|
Pozdzik AA, Salmon IJ, Husson CP, Decaestecker C, Rogier E, Bourgeade MF, Deschodt-Lanckman MM, Vanherweghem JL, Nortier JL. Patterns of interstitial inflammation during the evolution of renal injury in experimental aristolochic acid nephropathy. Nephrol Dial Transplant 2008; 23:2480-91. [PMID: 18385385 DOI: 10.1093/ndt/gfn140] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interstitial inflammation is a prominent feature associated with the severity of renal injury and progressive kidney failure. We utilized an animal model of aristolochic acid (AA)-induced nephropathy (AAN) to assess patterns of infiltration and inflammation during the evolution of tubulointerstitial damage and to relate them to the development of fibrosis. METHODS Male Wistar rats receiving sc daily AA or vehicle were sacrificed between Days 1 and 35. Infiltrating mononuclear cells were characterized by immunohistochemistry. The kidney infiltrating T lymphocytes were phenotyped by flow cytometry. Urinary levels of Th-1/ Th-2 cytokines, of monocyte chemoattractant protein-1 and of active transforming growth factor-beta (TGF-beta) were measured. Tissue expression of phosphorylated smad 2/3 protein was used to examine the TGF-beta signalling pathway. RESULTS In AA rats, monocytes/macrophages and T lymphocytes predominantly infiltrated areas of necrotic proximal tubular cells. The coexpressions of ED1 and/or Ki-67/MHCII by infiltrating cells reflected monocyte/macrophage proliferation and their activation, respectively. The accumulation of cytotoxic T lymphocytes was attested by severe signs of CD8+ cell tubulitis. The CD8/E-cadherin costaining confirmed intrarenal homing of CD8+CD103+ cells. Urinary levels of proinflammatory cytokines and of active TGF-beta significantly increased at Days 10 and 35. An early and persistent nuclear overexpression of phosphorylated smad 2/3 protein was detected in tubular and interstitial compartments. CONCLUSION An early and massive interstitial inflammation characterized by activated monocytes/macrophages and cytotoxic CD8+CD103+ T lymphocytes is demonstrated for the first time during the progression of experimental AAN. The involvement in an interstitial fibrosis onset of active TGF-beta is highly suggested, at least via the psmad 2/3 intracellular signalling pathway.
Collapse
Affiliation(s)
- Agnieszka A Pozdzik
- Department of Nephrology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, B-1070 Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Pozdzik AA, Salmon IJ, Debelle FD, Decaestecker C, Van den Branden C, Verbeelen D, Deschodt-Lanckman MM, Vanherweghem JL, Nortier JL. Aristolochic acid induces proximal tubule apoptosis and epithelial to mesenchymal transformation. Kidney Int 2007; 73:595-607. [PMID: 18094681 DOI: 10.1038/sj.ki.5002714] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aristolochic acid contamination in herbal remedies leads to interstitial fibrosis, tubular atrophy, and renal failure in humans. To study the cellular mechanisms contributing to the pathophysiology of this renal disease, we studied Wistar rats treated with aristolochic acid and measured tubular and interstitial cell proliferation, epithelial/mesenchymal cell marker expression, tubular membrane integrity, myofibroblast accumulation, oxidative stress, mitochondrial damage, tubular apoptosis, and fibrosis. Oxidative stress, a loss of cadherin concomitant with vimentin expression, basement membrane denudation with active caspase-3 expression, and mitochondrial injury within tubular cells were evident within 5 days of administration of the toxin. During the chronic phase, interstitial mesenchymal cells accumulated in areas of collagen deposits. Impaired regeneration and apoptosis of proximal tubular cells resulted in tubule atrophy with a near absence of dedifferentiated cell transmembrane migration. We suggest that resident fibroblast activation plays a critical role in the process of renal fibrosis during aristolochic acid toxicity.
Collapse
Affiliation(s)
- A A Pozdzik
- Experimental Nephrology Unit, Faculty of Medicine, Erasme Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lebeau C, Debelle FD, Arlt VM, Pozdzik A, De Prez EG, Phillips DH, Deschodt-Lanckman MM, Vanherweghem JL, Nortier JL. Early proximal tubule injury in experimental aristolochic acid nephropathy: functional and histological studies. Nephrol Dial Transplant 2005; 20:2321-32. [PMID: 16077141 DOI: 10.1093/ndt/gfi042] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Aristolochic acid (AA), the plant extract of Aristolochia species, is involved in the onset of progressive tubulointerstitial renal fibrosis in humans. Clinical and in vitro findings have previously suggested that the proximal tubule was the target of AA. METHODS Using a rat model of AA nephropathy, the proximal tubular lesions induced by daily subcutaneous injections of AA for 35 or 5 days were characterized biochemically and histologically. Urinary excretion of proteins, albumin, low molecular weight proteins, N-acetyl-beta-d-glucosaminidase, alpha-glutathione S-transferase, leucine aminopeptidase and neutral endopeptidase (NEP) was determined and related to histological conventional findings and immunostainings of NEP and megalin. RESULTS In both protocols, an acute phase of release of urinary markers was observed within the first 3 days of AA treatment in parallel with a significant increase of specific AA-related DNA adducts reflecting early tubular intoxication. A dramatic loss of the proximal tubule brush border was histologically confirmed, while the expression of megalin decreased at the damaged apical epithelium (mainly of the S3 segment). CONCLUSION Proximal tubule injury occurs early after AA intoxication in rats, with a link between specific AA-DNA adduct formation, decreased megalin expression and inhibition of receptor-mediated endocytosis of low molecular weight proteins, bringing in vivo confirmation of previous in vitro studies.
Collapse
Affiliation(s)
- Catherine Lebeau
- Laboratory for Research on Peptide Metabolism, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|