1
|
Guo J, Zhou S, Wang H, Qiu X, Dong F, Jiang S, Xu N, Cui Y, Liu R, Li P, Ma Z, Zhao L, Lai EY. ADAMTS13 attenuates renal fibrosis by suppressing thrombospondin 1 mediated TGF-β1/Smad3 activation. Toxicol Appl Pharmacol 2025; 496:117260. [PMID: 39929281 PMCID: PMC11877307 DOI: 10.1016/j.taap.2025.117260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Renal fibrosis is a common pathologic pathway for the progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD). Its mechanisms are unclear and it lacks effective therapy. Thrombospondin 1 (TSP1) mediated transforming growth factor-β1 (TGF-β1) activation was confirmed to promote renal fibrosis. Recently, a disintegrin and metalloprotease with thrombospondin type 1 repeats, member 13 (ADAMTS13), was reported to inhibit Thrombospondin 1 (TSP1) mediated Ca2+ signaling in the myocardial cell, besides its cleavage of von Willebrand factor (VWF). Therefore, we hypothesized that ADAMTS13 might protect against renal fibrosis by inhibiting TSP1-mediated TGF-β1 activation. In this study, clinical data on renal fibrosis and healthy controls were collected. Renal fibrosis models were established both in vivo and in vitro. In vivo, mice underwent unilateral ureteral obstruction (UUO) for 14 days. In vitro, human proximal tubular epithelial cells (HK-2) were exposed to TGF-β1. The results showed that the expression of ADAMTS13 was decreased accompanied by the increased expression of TSP1 in patients with renal fibrosis and renal fibrosis models in vivo and in vitro. The administration of rhADAMTS13 reduced proteinuria and renal fibrosis in UUO mice. rhADAMTS13 inhibited the expression of TSP1 and the activation of TGF-β1/Smad signaling pathway. The knockdown of ADAMTS13 exhibited a contrary result. The regulation of TSP1 directly affected the protective role of ADAMTS13 in renal fibrosis. Moreover, rhADAMTS13 attenuated inflammation induced by UUO. In conclusion, ADAMTS13 attenuates renal fibrosis induced by UUO. ADAMTS13 exerts its protective role by inhibiting TGF-β1 /Smad signaling via TSP1. NEW AND NOTEWORTHY: ADAMTS13 may be used as a novel molecular marker and a new therapeutic target for renal fibrosis. In this paper, ADAMTS13 was found to have an antifibrotic effect independent of its cleavage of VWF.
Collapse
Affiliation(s)
- Jie Guo
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Suhan Zhou
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Honghong Wang
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingyu Qiu
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Dong
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Shan Jiang
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Nan Xu
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China; Department of Pathophysiology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yu Cui
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruisheng Liu
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, FL, USA
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| | - Zufu Ma
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Liang Zhao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, and Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China.
| | - En Yin Lai
- Kidney Disease Center of the First Affiliated Hospital, and Department of Physiology, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
2
|
Shu L, Quan L, Wang Y, Chen Y, Yong C, Tian F, Gao K, Zhou E. Suyin Detoxification Prescription Regulates the Klotho and ERK/NF-κB Signaling Pathways to Alleviate Renal Injury. Cell Biochem Biophys 2025:10.1007/s12013-025-01695-5. [PMID: 39966333 DOI: 10.1007/s12013-025-01695-5] [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] [Accepted: 02/04/2025] [Indexed: 02/20/2025]
Abstract
Chronic Kidney Disease (CKD) is an irreversible pathological condition resulting from the gradual accumulation of nephrotoxic substances. The application of the "Kidney Toxicity" theory in CKD treatment holds significant research potential and promising clinical prospects. We established a murine model of unilateral ureteral obstruction (UUO) and administered the traditional Chinese medicine prescription Suyin detoxification prescription (SDP). The efficacy of SDP was assessed by comparing the expression levels of Klotho, NF-κB, pNF-κB, VEGF, α-SMA, pERK1/2, and ERK1/2 proteins with those in the Sham operation group using immunohistochemistry, RT-qPCR, and Western Blot techniques. The study revealed a significant reduction in the expression of Klotho and VEGF proteins during the progression of renal fibrosis in mice, while there was a marked increase in the levels of pNF-κB, α-SMA, and pERK1/2 proteins, demonstrating statistical significance (p < 0.05). In UUO mice treated with a high dose of SDP, these proteins exhibited an opposite expression trend compared to that observed in pure operated model mice, with statistically significant differences (p < 0.05). Subsequently, we investigated the relationship between Klotho protein and the ERK/NF-κB signaling pathway-related proteins in human umbilical vein endothelial cells (HUVECs). Knockdown of Klotho protein in HUVECs confirmed its potential as a target for SDP's renal protective effects in vivo by regulating ERK/NF-κB signaling pathway-related proteins to some extent. The renoprotective effect of SDP is mediated through modulation of Klotho protein expression in renal tissues, thereby influencing the ERK/NF-κB signaling pathway and ameliorating the inflammatory processes associated with renal fibrosis. The present study has significantly contributed to the advancement and refinement of the pathogenesis of "Kidney Toxicity" as well as the therapeutic approach of "Kidney Detoxification".
Collapse
Affiliation(s)
- Lianghui Shu
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
- Department of Nephrology, Wuxi Ninth People's Hospital Affiliated of Soochow University, Wuxi, People's Republic of China
| | - Li Quan
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Yanping Wang
- Department of Nephrology, Afffliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yanlin Chen
- No.1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Chen Yong
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Fang Tian
- Research Center of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Kun Gao
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
- Nanjing University of Chinese Medicine, Jiangsu University Key Laboratory of Tonifying Kidney and Anti-senescenc, Nanjing, People's Republic of China
| | - Enchao Zhou
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China.
- Nanjing University of Chinese Medicine, Jiangsu University Key Laboratory of Tonifying Kidney and Anti-senescenc, Nanjing, People's Republic of China.
| |
Collapse
|
3
|
Di X, Li Y, Wei J, Li T, Liao B. Targeting Fibrosis: From Molecular Mechanisms to Advanced Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410416. [PMID: 39665319 PMCID: PMC11744640 DOI: 10.1002/advs.202410416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/27/2024] [Indexed: 12/13/2024]
Abstract
As the final stage of disease-related tissue injury and repair, fibrosis is characterized by excessive accumulation of the extracellular matrix. Unrestricted accumulation of stromal cells and matrix during fibrosis impairs the structure and function of organs, ultimately leading to organ failure. The major etiology of fibrosis is an injury caused by genetic heterogeneity, trauma, virus infection, alcohol, mechanical stimuli, and drug. Persistent abnormal activation of "quiescent" fibroblasts that interact with or do not interact with the immune system via complicated signaling cascades, in which parenchymal cells are also triggered, is identified as the main mechanism involved in the initiation and progression of fibrosis. Although the mechanisms of fibrosis are still largely unknown, multiple therapeutic strategies targeting identified molecular mechanisms have greatly attenuated fibrotic lesions in clinical trials. In this review, the organ-specific molecular mechanisms of fibrosis is systematically summarized, including cardiac fibrosis, hepatic fibrosis, renal fibrosis, and pulmonary fibrosis. Some important signaling pathways associated with fibrosis are also introduced. Finally, the current antifibrotic strategies based on therapeutic targets and clinical trials are discussed. A comprehensive interpretation of the current mechanisms and therapeutic strategies targeting fibrosis will provide the fundamental theoretical basis not only for fibrosis but also for the development of antifibrotic therapies.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
4
|
Huang H, Huang S, Li C, Zhang C, Wang R, Wei L, Wu J, Mo P, Li Z, Li S, Chen J. Jian-Pi-Yi-Shen formula ameliorates renal fibrosis-induced anemia in rats with chronic kidney disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118607. [PMID: 39069029 DOI: 10.1016/j.jep.2024.118607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jian-Pi-Yi-Shen (JPYS) formula is an effective herbal therapy against renal injury, and JPYS has been clinically applied to ameliorate chronic kidney disease (CKD) and CKD-associated anemia. Increasing evidence supports the link between renal fibrosis and anemia in CKD. JPYS possessed anti-fibrosis effects in experimental CKD. Nevertheless, research on the mechanisms of JPYS in ameliorating renal anemia (RA) through suppressing renal fibrosis remains to be clarified. AIM OF THE STUDY Our study here was carried out to investigate the mechanisms of JPYS in protecting against RA. MATERIALS AND METHODS An adenine-induced anemia model in rats with CKD at three different time points was established, and bio-samples taken from each group were analyzed. Biochemical analysis was employed to detect kidney function and hematological parameters. Masson staining was used to evaluate renal fibrosis of rats. Western blot and immunohistochemistry were utilized to evaluate the expressions of fibrotic markers, erythropoietin (EPO) and hypoxia inducible factor-2α (HIF-2α) in the kidneys of rats. Subsequently, transcriptomic analysis was conducted to disclose the possible mechanisms of JPYS in treating RA. Finally, the expression levels of key targets were analyzed and validated by using Western blot and enzyme-linked immunosorbent assay (ELISA). RESULTS JPYS treatment improved kidney function, suppressed renal fibrosis and enhanced hematological parameters in CKD rats. Moreover, JPYS treatment restored the increased expression levels of fibrotic markers and the declined EPO with time dependence. In parallel, data indicated JPYS treatment stimulated the translocation of HIF-2α into nucleus in the renal interstitium and thus promoted the expression of EPO. Transcriptomic profiling disclosed that activations of both nuclear factor kappa B (NF-κB) and transforming growth factor-β (TGF-β)/Smad pathways were closely associated with RA. Ultimately, experimental validation results presented that the increased expressions of target proteins from the above-mentioned two pathways in the kidneys were decreased significantly after JPYS treatment. CONCLUSION Our findings suggest that JPYS may improve RA by alleviating renal fibrosis, and the mechanisms of which involve in inhibiting the NF-κB and TGF-β/Smad pathways.
Collapse
Affiliation(s)
- Haipiao Huang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Shiying Huang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Changhui Li
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Chi Zhang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Rui Wang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Lifang Wei
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Jinru Wu
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Pingli Mo
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Zhonggui Li
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Shunmin Li
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
| | - Jianping Chen
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
| |
Collapse
|
5
|
Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol 2024; 327:F788-F805. [PMID: 39298548 DOI: 10.1152/ajprenal.00184.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are increasingly recognized as interconnected conditions with overlapping pathophysiological mechanisms. This review examines the transition from AKI to CKD, focusing on the molecular mechanisms, animal models, and biomarkers essential for understanding and managing this progression. AKI often progresses to CKD due to maladaptive repair processes, persistent inflammation, and fibrosis, with both conditions sharing common pathways involving cell death, inflammation, and extracellular matrix (ECM) deposition. Current animal models, including ischemia-reperfusion injury (IRI) and nephrotoxic damage, help elucidate these mechanisms but have limitations in replicating the complexity of human disease. Emerging biomarkers such as kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and soluble tumor necrosis factor receptors (TNFRs) show promise in early detection and monitoring of disease progression. This review highlights the need for improved animal models and biomarker validation to better mimic human disease and enhance clinical translation. Advancing our understanding of the AKI-to-CKD transition through targeted therapies and refined research approaches holds the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Tingfang Zhang
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
6
|
Rende U, Guller A, Goldys EM, Pollock C, Saad S. Diagnostic and prognostic biomarkers for tubulointerstitial fibrosis. J Physiol 2023; 601:2801-2826. [PMID: 37227074 DOI: 10.1113/jp284289] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/23/2023] [Indexed: 05/26/2023] Open
Abstract
Renal fibrosis is the final common pathophysiological pathway in chronic kidney disease (CKD) regardless of the underlying cause of kidney injury. Tubulointerstitial fibrosis (TIF) is considered to be the key pathological predictor of CKD progression. Currently, the gold-standard tool to identify TIF is kidney biopsy, an invasive method that carries risks. Non-invasive diagnostics rely on an estimation of glomerular filtration rate and albuminuria to assess kidney function, but these fail to diagnose early CKD accurately or to predict progressive decline in kidney function. In this review, we summarize the current and emerging molecular biomarkers that have been studied in various clinical settings and in animal models of kidney disease and that are correlated with the degree of TIF. We examine the potential of these biomarkers to diagnose TIF non-invasively and to predict disease progression. We also examine the potential of new technologies and non-invasive diagnostic approaches in assessing TIF. Limitations of current and potential biomarkers are discussed and knowledge gaps identified.
Collapse
Affiliation(s)
- Umut Rende
- School of Biomedical Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - Anna Guller
- Macquarie Medical School, Faculty of Medicine, Health & Human Sciences, Macquarie University, NSW, Australia
| | - Ewa M Goldys
- School of Biomedical Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - Carol Pollock
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Sonia Saad
- Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
7
|
Ume AC, Wenegieme TY, Shelby JN, Paul-Onyia CDB, Waite AMJ, Kamau JK, Adams DN, Susuki K, Bennett ES, Ren H, Williams CR. Tacrolimus induces fibroblast-to-myofibroblast transition via a TGF-β-dependent mechanism to contribute to renal fibrosis. Am J Physiol Renal Physiol 2023; 324:F433-F445. [PMID: 36927118 PMCID: PMC10085566 DOI: 10.1152/ajprenal.00226.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 02/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Use of immunosuppressant calcineurin inhibitors (CNIs) is limited by irreversible kidney damage, hallmarked by renal fibrosis. CNIs directly damage many renal cell types. Given the diverse renal cell populations, additional targeted cell types and signaling mechanisms warrant further investigation. We hypothesized that fibroblasts contribute to CNI-induced renal fibrosis and propagate profibrotic effects via the transforming growth factor-β (TGF-β)/Smad signaling axis. To test this, kidney damage-resistant mice (C57BL/6) received tacrolimus (10 mg/kg) or vehicle for 21 days. Renal damage markers and signaling mediators were assessed. To investigate their role in renal damage, mouse renal fibroblasts were exposed to tacrolimus (1 nM) or vehicle for 24 h. Morphological and functional changes in addition to downstream signaling events were assessed. Tacrolimus-treated kidneys displayed evidence of renal fibrosis. Moreover, α-smooth muscle actin expression was significantly increased, suggesting the presence of fibroblast activation. TGF-β receptor activation and downstream Smad2/3 signaling were also upregulated. Consistent with in vivo findings, tacrolimus-treated renal fibroblasts displayed a phenotypic switch known as fibroblast-to-myofibroblast transition (FMT), as α-smooth muscle actin, actin stress fibers, cell motility, and collagen type IV expression were significantly increased. These findings were accompanied by concomitant induction of TGF-β signaling. Pharmacological inhibition of the downstream TGF-β effector Smad3 attenuated tacrolimus-induced phenotypic changes. Collectively, these findings suggest that 1) tacrolimus inhibits the calcineurin/nuclear factor of activated T cells axis while inducing TGF-β1 ligand secretion and receptor activation in renal fibroblasts; 2) aberrant TGF-β receptor activation stimulates Smad-mediated production of myofibroblast markers, notable features of FMT; and 3) FMT contributes to extracellular matrix expansion in tacrolimus-induced renal fibrosis. These results incorporate renal fibroblasts into the growing list of CNI-targeted cell types and identify renal FMT as a process mediated via a TGF-β-dependent mechanism.NEW & NOTEWORTHY Renal fibrosis, a detrimental feature of irreversible kidney damage, remains a sinister consequence of long-term calcineurin inhibitor (CNI) immunosuppressive therapy. Our study not only incorporates renal fibroblasts into the growing list of cell types negatively impacted by CNIs but also identifies renal fibroblast-to-myofibroblast transition as a process mediated via a TGF-β-dependent mechanism. This insight will direct future studies investigating the feasibility of inhibiting TGF-β signaling to maintain CNI-mediated immunosuppression while ultimately preserving kidney health.
Collapse
Affiliation(s)
- Adaku C Ume
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Tara Y Wenegieme
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Jennae N Shelby
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Chiagozie D B Paul-Onyia
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Aston M J Waite
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - John K Kamau
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Danielle N Adams
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Eric S Bennett
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Hongmei Ren
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Clintoria R Williams
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| |
Collapse
|
8
|
Fuchs MAA, Schrankl J, Wagner C, Daniel C, Kurtz A, Broeker KAE. Localization and characterization of proenkephalin-A as a potential biomarker for kidney disease in murine and human kidneys. Biomarkers 2023; 28:76-86. [PMID: 36354355 DOI: 10.1080/1354750x.2022.2146196] [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: 11/12/2022]
Abstract
INTRODUCTION Exact measurement of renal function is essential for the treatment of patients. Elevated serum-creatinine levels, while established, are influenced by other parameters and show a significant time-lag. This drives the search for novel biomarkers of renal function and injury. Beside Lipocalin-2 and kidney-injury-molecule-1 (KIM-1), the endogenous opioid precursor proenkephalin-A (Penk) has recently emerged as a promising marker for renal function. But the cellular origin and regulation of Penk outside the brain has not yet been investigated in depth. MATERIALS AND METHODS This study characterizes the cellular origin of Penk expression with high-resolution in situ hybridization in two models of renal fibrosis in mice and human tissue. RESULTS Interstitial cells are the main expression site for renal Penk. This classifies Penk as biomarker for interstitial damage as opposed to tubular damage markers like Lipocalin-2 and KIM-1. Furthermore, our data indicate that renal Penk expression is not regulated by classical profibrotic pathways. DISCUSSION This study characterizes changing Penk expression in the kidneys. The similarity of Penk expression across species gives rise to further investigations into the function of Penk in healthy and injured kidneys. CONCLUSION Penk is a promising biomarker for interstitial renal damage that warrants further studies to utilize its predictive potential.Clinical significanceKnowledge of real-time renal function is essential for proper treatment of critically ill patients and in early diagnosis of acute kidney injury (AKI). Proenkephalin-A has been measured in a number of patient cohorts as a highly accurate and predictive biomarker of renal damage.The present study identifies Penk as a biomarker for interstitial damage in contrast to the tubular biomarkers such as Lipocalin-2 or KIM-1.Our data show that Penk is regulated independently of classical profibrotic or proinflammatory pathways, indicating it might be more robust against extra-renal influences.Data presented in this study provide fundamental information about cell type-specific localization and regulation of the potential new biomarker Penk across species as foundation for further research.
Collapse
Affiliation(s)
| | - Julia Schrankl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Christoph Daniel
- Department of Nephropathology, Universitätsklinikum Erlangen-Nürnberg, Erlangen, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | | |
Collapse
|
9
|
Jia T, Xu T, Smeets B, Buhl EM, Moeller MJ, Floege J, Klinkhammer BM, Boor P. The Role of Platelet-Derived Growth Factor in Focal Segmental Glomerulosclerosis. J Am Soc Nephrol 2023; 34:241-257. [PMID: 36351762 PMCID: PMC10103089 DOI: 10.1681/asn.2022040491] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND FSGS is the final common pathway to nephron loss in most forms of severe or progressive glomerular injury. Although podocyte injury initiates FSGS, parietal epithelial cells (PECs) are the main effectors. Because PDGF takes part in fibrotic processes, we hypothesized that the ligand PDGF-B and its receptor PDGFR- β participate in the origin and progression of FSGS. METHODS We challenged Thy1.1 transgenic mice, which express Thy1.1 in the podocytes, with anti-Thy1.1 antibody to study the progression of FSGS. We investigated the role of PDGF in FSGS using challenged Thy1.1 mice, 5/6 nephrectomized mice, Col4 -/- (Alport) mice, patient kidney biopsies, and primary murine PECs, and challenged Thy1.1 mice treated with neutralizing anti-PDGF-B antibody therapy. RESULTS The unchallenged Thy1.1 mice developed only mild spontaneous FSGS, whereas challenged mice developed progressive FSGS accompanied by a decline in kidney function. PEC activation, proliferation, and profibrotic phenotypic switch drove the FSGS. During disease, PDGF-B was upregulated in podocytes, whereas PDGFR- β was upregulated in PECs from both mice and patients with FSGS. Short- and long-term treatment with PDGF-B neutralizing antibody improved kidney function and reduced FSGS, PEC proliferation, and profibrotic activation. In vitro , stimulation of primary murine PECs with PDGF-B recapitulated in vivo findings with PEC activation and proliferation, which was inhibited by PDGF-B antibody or imatinib. CONCLUSION PDGF-B-PDGFR- β molecular crosstalk between podocytes and PECs drives glomerulosclerosis and the progression of FSGS. PODCAST This article contains a podcast at.
Collapse
Affiliation(s)
- Ting Jia
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Tong Xu
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Eva Miriam Buhl
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
| | - Marcus Johannes Moeller
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
- Heisenberg Chair for Preventive and Translational Nephrology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Barbara Mara Klinkhammer
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
10
|
Soomro A, Khajehei M, Li R, O’Neil K, Zhang D, Gao B, MacDonald M, Kakoki M, Krepinsky JC. A therapeutic target for CKD: activin A facilitates TGFβ1 profibrotic signaling. Cell Mol Biol Lett 2023; 28:10. [PMID: 36717814 PMCID: PMC9885651 DOI: 10.1186/s11658-023-00424-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND TGFβ1 is a major profibrotic mediator in chronic kidney disease (CKD). Its direct inhibition, however, is limited by adverse effects. Inhibition of activins, also members of the TGFβ superfamily, blocks TGFβ1 profibrotic effects, but the mechanism underlying this and the specific activin(s) involved are unknown. METHODS Cells were treated with TGFβ1 or activins A/B. Activins were inhibited generally with follistatin, or specifically with neutralizing antibodies or type I receptor downregulation. Cytokine levels, signaling and profibrotic responses were assessed with ELISA, immunofluorescence, immunoblotting and promoter luciferase reporters. Wild-type or TGFβ1-overexpressing mice with unilateral ureteral obstruction (UUO) were treated with an activin A neutralizing antibody. RESULTS In primary mesangial cells, TGFβ1 induces secretion primarily of activin A, which enables longer-term profibrotic effects by enhancing Smad3 phosphorylation and transcriptional activity. This results from lack of cell refractoriness to activin A, unlike that for TGFβ1, and promotion of TGFβ type II receptor expression. Activin A also supports transcription through regulating non-canonical MRTF-A activation. TGFβ1 additionally induces secretion of activin A, but not B, from tubular cells, and activin A neutralization prevents the TGFβ1 profibrotic response in renal fibroblasts. Fibrosis induced by UUO is inhibited by activin A neutralization in wild-type mice. Worsened fibrosis in TGFβ1-overexpressing mice is associated with increased renal activin A expression and is inhibited to wild-type levels with activin A neutralization. CONCLUSIONS Activin A facilitates TGFβ1 profibrotic effects through regulation of both canonical (Smad3) and non-canonical (MRTF-A) signaling, suggesting it may be a novel therapeutic target for preventing fibrosis in CKD.
Collapse
Affiliation(s)
- Asfia Soomro
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Mohammad Khajehei
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Renzhong Li
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Kian O’Neil
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Dan Zhang
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Bo Gao
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Melissa MacDonald
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Masao Kakoki
- grid.410711.20000 0001 1034 1720Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Joan C. Krepinsky
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada ,grid.416721.70000 0001 0742 7355St. Joseph’s Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON L8N 4A6 Canada
| |
Collapse
|
11
|
Ruby M, Gifford CC, Pandey R, Raj VS, Sabbisetti VS, Ajay AK. Autophagy as a Therapeutic Target for Chronic Kidney Disease and the Roles of TGF-β1 in Autophagy and Kidney Fibrosis. Cells 2023; 12:412. [PMID: 36766754 PMCID: PMC9913737 DOI: 10.3390/cells12030412] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Autophagy is a lysosomal protein degradation system that eliminates cytoplasmic components such as protein aggregates, damaged organelles, and even invading pathogens. Autophagy is an evolutionarily conserved homoeostatic strategy for cell survival in stressful conditions and has been linked to a variety of biological processes and disorders. It is vital for the homeostasis and survival of renal cells such as podocytes and tubular epithelial cells, as well as immune cells in the healthy kidney. Autophagy activation protects renal cells under stressed conditions, whereas autophagy deficiency increases the vulnerability of the kidney to injury, resulting in several aberrant processes that ultimately lead to renal failure. Renal fibrosis is a condition that, if chronic, will progress to end-stage kidney disease, which at this point is incurable. Chronic Kidney Disease (CKD) is linked to significant alterations in cell signaling such as the activation of the pleiotropic cytokine transforming growth factor-β1 (TGF-β1). While the expression of TGF-β1 can promote fibrogenesis, it can also activate autophagy, which suppresses renal tubulointerstitial fibrosis. Autophagy has a complex variety of impacts depending on the context, cell types, and pathological circumstances, and can be profibrotic or antifibrotic. Induction of autophagy in tubular cells, particularly in the proximal tubular epithelial cells (PTECs) protects cells against stresses such as proteinuria-induced apoptosis and ischemia-induced acute kidney injury (AKI), whereas the loss of autophagy in renal cells scores a significant increase in sensitivity to several renal diseases. In this review, we discuss new findings that emphasize the various functions of TGF-β1 in producing not just renal fibrosis but also the beneficial TGF-β1 signaling mechanisms in autophagy.
Collapse
Affiliation(s)
- Miss Ruby
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat 131029, Haryana, India
| | - Cody C. Gifford
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - RamendraPati Pandey
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat 131029, Haryana, India
| | - V. Samuel Raj
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat 131029, Haryana, India
| | - Venkata S. Sabbisetti
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Amrendra K. Ajay
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Zhang J, Shen R, Lin H, Pan J, Feng X, Lin L, Niu D, Hou Y, Su X, Wang C, Wang L, Qiao X. Effects of contralateral nephrectomy timing and ischemic conditions on kidney fibrosis after unilateral kidney ischemia-reperfusion injury. Ren Fail 2022; 44:1568-1584. [PMID: 36154902 PMCID: PMC9543178 DOI: 10.1080/0886022x.2022.2126790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Acute kidney injury (AKI) is an important cause of chronic kidney disease (CKD), but the underlying mechanisms are unclear. Animal models are tools for studying the AKI-CKD progression. Kidney ischemia-reperfusion injury (IRI) models, especially the unilateral IRI (uIRI) model with delayed contralateral kidney resection, are commonly used to induce fibrotic progression to CKD after AKI. However, in previous studies, we found that details of the operation had a significant impact on the long-term outcomes of the kidney in this uIRI model. In this study, we investigated the effects of resection timing of the contralateral intact kidney, core body temperatures during ischemia, and time length of kidney ischemia on kidney function, histological injury and kidney fibrosis after AKI, using a mouse uIRI model with delayed contralateral nephrectomy. The results showed that all these parameters significantly affected the AKI-CKD transition. The post-AKI fibrosis worsened and the survival rate declined with a longer interval between contralateral nephrectomy and uIRI, higher ischemic body temperature, or longer ischemic duration when the other two variables were fixed. In conclusion, in the uIRI model with delayed contralateral nephrectomy, kidney fibrosis after AKI is influenced by many factors. Strictly controlling the experimental conditions is very important for the stability and consistency of the model.
Collapse
Affiliation(s)
- Junhua Zhang
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ruihua Shen
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hui Lin
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Juan Pan
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xinyuan Feng
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Ling Lin
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Dan Niu
- Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Department of Pathology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yanjuan Hou
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xiaole Su
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Chen Wang
- Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Department of Pathology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Lihua Wang
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xi Qiao
- Department of Nephrology, Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.,Shanxi Kidney Disease Institute, Taiyuan, People's Republic of China.,Institute of Nephrology, Shanxi Medical University, Taiyuan, People's Republic of China
| |
Collapse
|
13
|
Kobayashi H, Davidoff O, Pujari-Palmer S, Drevin M, Haase VH. EPO synthesis induced by HIF-PHD inhibition is dependent on myofibroblast transdifferentiation and colocalizes with non-injured nephron segments in murine kidney fibrosis. Acta Physiol (Oxf) 2022; 235:e13826. [PMID: 35491502 PMCID: PMC9329237 DOI: 10.1111/apha.13826] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/14/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022]
Abstract
AIM Erythropoietin (EPO) is regulated by hypoxia-inducible factor (HIF)-2. In the kidney, it is produced by cortico-medullary perivascular interstitial cells, which transdifferentiate into collagen-producing myofibroblasts in response to injury. Inhibitors of prolyl hydroxylase domain (PHD) dioxygenases (HIF-PHIs) activate HIF-2 and stimulate kidney and liver EPO synthesis in patients with anemia of chronic kidney disease (CKD). We examined whether HIF-PHIs can reactivate EPO synthesis in interstitial cells that have undergone myofibroblast transdifferentiation in established kidney fibrosis. METHODS We investigated Epo transcription in myofibroblasts and characterized the histological distribution of kidney Epo transcripts by RNA in situ hybridization combined with immunofluorescence in mice with adenine nephropathy (AN) treated with HIF-PHI molidustat. Lectin absorption chromatography was used to assess liver-derived EPO. In addition, we examined kidney Epo transcription in Phd2 knockout mice with obstructive nephropathy. RESULTS In AN, molidustat-induced Epo transcripts were not found in areas of fibrosis and did not colocalize with interstitial cells that expressed α-smooth muscle actin, a marker of myofibroblast transdifferentiation. Epo transcription was associated with megalin-expressing, kidney injury molecule 1-negative nephron segments and contingent on residual renal function. Liver-derived EPO did not contribute to serum EPO in molidustat-treated mice. Epo transcription was not associated with myofibroblasts in Phd2 knockout mice with obstructive nephropathy. CONCLUSIONS Our studies suggest that HIF-PHIs do not reactivate Epo transcription in interstitial myofibroblasts and that their efficacy in inducing kidney EPO in CKD is dependent on the degree of myofibroblast formation, the preservation of renal parenchyma and the level of residual renal function.
Collapse
Affiliation(s)
- Hanako Kobayashi
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Olena Davidoff
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | | | | | - Volker H Haase
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Molecular Physiology & Biophysics and Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Dahl SL, Bapst AM, Khodo SN, Scholz CC, Wenger RH. Fount, fate, features, and function of renal erythropoietin-producing cells. Pflugers Arch 2022; 474:783-797. [PMID: 35750861 PMCID: PMC9338912 DOI: 10.1007/s00424-022-02714-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 12/19/2022]
Abstract
Renal erythropoietin (Epo)-producing (REP) cells represent a rare and incompletely understood cell type. REP cells are fibroblast-like cells located in close proximity to blood vessels and tubules of the corticomedullary border region. Epo mRNA in REP cells is produced in a pronounced "on-off" mode, showing transient transcriptional bursts upon exposure to hypoxia. In contrast to "ordinary" fibroblasts, REP cells do not proliferate ex vivo, cease to produce Epo, and lose their identity following immortalization and prolonged in vitro culture, consistent with the loss of Epo production following REP cell proliferation during tissue remodelling in chronic kidney disease. Because Epo protein is usually not detectable in kidney tissue, and Epo mRNA is only transiently induced under hypoxic conditions, transgenic mouse models have been developed to permanently label REP cell precursors, active Epo producers, and inactive descendants. Future single-cell analyses of the renal stromal compartment will identify novel characteristic markers of tagged REP cells, which will provide novel insights into the regulation of Epo expression in this unique cell type.
Collapse
Affiliation(s)
- Sophie L Dahl
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
| | - Andreas M Bapst
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
| | - Stellor Nlandu Khodo
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
| | - Carsten C Scholz
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
- Institute of Physiology, University Medicine Greifswald, D-17475, Greifswald, Germany
| | - Roland H Wenger
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland.
| |
Collapse
|
15
|
Fuchs MAA, Kurtz A. The fate of erythropoietin-producing cells: another piece of the puzzle. Kidney Int 2022; 102:230-233. [PMID: 35870811 DOI: 10.1016/j.kint.2022.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 10/17/2022]
Abstract
In this issue, Kaneko et al. reported the generation of a mouse line that allows for the labeling of cells under control of the erythropoietin (Epo) gene promotor. The authors show that Epo-producing cells become proliferating, profibrotic cells after kidney injury and lose their ability to produce Epo. Furthermore, they show that the fluorescent-labeled cells can recover their Epo synthesis capability subsequently to a recovery period.
Collapse
Affiliation(s)
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
16
|
Kaneko K, Sato Y, Uchino E, Toriu N, Shigeta M, Kiyonari H, Endo S, Fukuma S, Yanagita M. Lineage tracing analysis defines erythropoietin-producing cells as a distinct subpopulation of resident fibroblasts with unique behaviors. Kidney Int 2022; 102:280-292. [DOI: 10.1016/j.kint.2022.04.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 04/16/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022]
|
17
|
Dahl SL, Pfundstein S, Hunkeler R, Dong X, Knöpfel T, Spielmann P, Scholz CC, Nolan KA, Wenger RH. Fate-mapping of erythropoietin-producing cells in mouse models of hypoxaemia and renal tissue remodelling reveals repeated recruitment and persistent functionality. Acta Physiol (Oxf) 2022; 234:e13768. [PMID: 34982511 PMCID: PMC9286872 DOI: 10.1111/apha.13768] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/08/2021] [Accepted: 01/01/2022] [Indexed: 12/17/2022]
Abstract
Aim Fibroblast‐like renal erythropoietin (Epo) producing (REP) cells of the corticomedullary border region “sense” a decrease in blood oxygen content following anaemia or hypoxaemia. Burst‐like transcription of Epo during tissue hypoxia is transient and is lost during fibrotic tissue remodelling, as observed in chronic kidney disease. The reason for this loss of Epo expression is under debate. Therefore, we tested the hypothesis that REP cell migration, loss and/or differentiation may cause Epo inhibition. Methods Using a reporter mouse that allows permanent labelling of active REP cells at any given time point, we analysed the spatiotemporal fate of REP cells following their initial hypoxic recruitment in models of hypoxaemia and renal tissue remodelling. Results In long‐term tracing experiments, tagged REP reporter cells neither died, proliferated, migrated nor transdifferentiated into myofibroblasts. Approximately 60% of tagged cells re‐expressed Epo upon a second hypoxic stimulus. In an unilateral model of tissue remodelling, tagged cells proliferated and ceased to produce Epo before a detectable increase in myofibroblast markers. Treatment with a hypoxia‐inducible factor (HIF) stabilizing agent (FG‐4592/roxadustat) re‐induced Epo expression in the previously active REP cells of the damaged kidney to a similar extent as in the contralateral healthy kidney. Conclusions Rather than cell death or differentiation, these results suggest cell‐intrinsic transient inhibition of Epo transcription: following long‐term dormancy, REP cells can repeatedly be recruited by tissue hypoxia, and during myofibrotic tissue remodelling, dormant REP cells are efficiently rescued by a pharmaceutic HIF stabilizer, demonstrating persistent REP cell functionality even during phases of Epo suppression.
Collapse
Affiliation(s)
- Sophie L. Dahl
- Institute of Physiology University of Zurich Zurich Switzerland
- National Center of Competence in Research “Kidney.CH” Zurich Switzerland
| | - Svende Pfundstein
- Institute of Physiology University of Zurich Zurich Switzerland
- National Center of Competence in Research “Kidney.CH” Zurich Switzerland
| | - Rico Hunkeler
- Institute of Physiology University of Zurich Zurich Switzerland
- National Center of Competence in Research “Kidney.CH” Zurich Switzerland
| | - Xingtong Dong
- Institute of Physiology University of Zurich Zurich Switzerland
| | - Thomas Knöpfel
- Institute of Physiology University of Zurich Zurich Switzerland
| | | | - Carsten C. Scholz
- Institute of Physiology University of Zurich Zurich Switzerland
- National Center of Competence in Research “Kidney.CH” Zurich Switzerland
| | - Karen A. Nolan
- Institute of Physiology University of Zurich Zurich Switzerland
- National Center of Competence in Research “Kidney.CH” Zurich Switzerland
| | - Roland H. Wenger
- Institute of Physiology University of Zurich Zurich Switzerland
- National Center of Competence in Research “Kidney.CH” Zurich Switzerland
| |
Collapse
|
18
|
Bapst AM, Knöpfel T, Nolan KA, Imeri F, Schuh CD, Hall AM, Guo J, Katschinski DM, Wenger RH. Neurogenic and pericytic plasticity of conditionally immortalized cells derived from renal erythropoietin-producing cells. J Cell Physiol 2022; 237:2420-2433. [PMID: 35014036 PMCID: PMC9303970 DOI: 10.1002/jcp.30677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022]
Abstract
In adult mammals, the kidney is the main source of circulating erythropoietin (Epo), the master regulator of erythropoiesis. In vivo data in mice demonstrated multiple subtypes of interstitial renal Epo‐producing (REP) cells. To analyze the differentiation plasticity of fibroblastoid REP cells, we used a transgenic REP cell reporter mouse model to generate conditionally immortalized REP‐derived (REPD) cell lines. Under nonpermissive conditions, REPD cells ceased from proliferation and acquired a stem cell‐like state, with strongly enhanced hypoxia‐inducible factor 2 (HIF‐2α), stem cell antigen 1 (SCA‐1), and CD133 expression, but also enhanced alpha‐smooth muscle actin (αSMA) expression, indicating myofibroblastic signaling. These cells maintained the “on‐off” nature of Epo expression observed in REP cells in vivo, whereas other HIF target genes showed a more permanent regulation. Like REP cells in vivo, REPD cells cultured in vitro generated long tunneling nanotubes (TNTs) that aligned with endothelial vascular structures, were densely packed with mitochondria and became more numerous under hypoxic conditions. Although inhibition of mitochondrial oxygen consumption blunted HIF signaling, removal of the TNTs did not affect or even enhance the expression of HIF target genes. Apart from pericytes, REPD cells readily differentiated into neuroglia but not adipogenic, chondrogenic, or osteogenic lineages, consistent with a neuronal origin of at least a subpopulation of REP cells. In summary, these results suggest an unprecedented combination of differentiation features of this unique cell type.
Collapse
Affiliation(s)
- Andreas M Bapst
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Thomas Knöpfel
- Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Karen A Nolan
- Institute of Physiology, University of Zürich, Zürich, Switzerland.,National Centre of Competence in Research "Kidney.CH", University of Zürich, Zürich, Switzerland
| | - Faik Imeri
- Institute of Physiology, University of Zürich, Zürich, Switzerland.,National Centre of Competence in Research "Kidney.CH", University of Zürich, Zürich, Switzerland
| | - Claus D Schuh
- National Centre of Competence in Research "Kidney.CH", University of Zürich, Zürich, Switzerland.,Institute of Anatomy, University of Zürich, Zürich, Switzerland
| | - Andrew M Hall
- National Centre of Competence in Research "Kidney.CH", University of Zürich, Zürich, Switzerland.,Institute of Anatomy, University of Zürich, Zürich, Switzerland
| | - Jia Guo
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Dörthe M Katschinski
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Roland H Wenger
- Institute of Physiology, University of Zürich, Zürich, Switzerland.,National Centre of Competence in Research "Kidney.CH", University of Zürich, Zürich, Switzerland
| |
Collapse
|
19
|
Shih HM, Pan SY, Wu CJ, Chou YH, Chen CY, Chang FC, Chen YT, Chiang WC, Tsai HC, Chen YM, Lin SL. Transforming growth factor-β1 decreases erythropoietin production through repressing hypoxia-inducible factor 2α in erythropoietin-producing cells. J Biomed Sci 2021; 28:73. [PMID: 34724959 PMCID: PMC8561873 DOI: 10.1186/s12929-021-00770-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/26/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Renal erythropoietin (EPO)-producing (REP) cells produce EPO through hypoxia-inducible factor (HIF) 2α-activated gene transcription. Insufficient EPO production leads to anemia in patients with chronic kidney disease. Although recombinant EPO is effective to improve anemia, no reliable REP cell lines limit further progress of research and development of novel treatment. METHODS We screened Epo mRNA expression in mouse fibroblast cell lines. Small interfering RNA specific for HIF1α or HIF2α was transfected to study Epo expression in C3H10T1/2 cells. The effect of transforming growth factor-β1 (TGF-β1) on HIF-EPO axis was studied in C3H10T1/2 cells and mice. RESULTS Similar to mouse REP pericytes, C3H10T1/2 cells differentiated to α-smooth muscle actin+ myofibroblasts after exposure to TGF-β1. Specific HIF knockdown demonstrated the role of HIF2α in hypoxia-induced Epo expression. Sustained TGF-β1 exposure increased neither DNA methyltransferase nor methylation of Epas1 and Epo genes. However, TGF-β1 repressed HIF2α-encoding Epas1 promptly through activating activin receptor-like kinase-5 (ALK5), thereby decreasing Epo induction by hypoxia and prolyl hydroxylase domain inhibitor roxadustat. In mice with pro-fibrotic injury induced by ureteral obstruction, upregulation of Tgfb1 was accompanied with downregulation of Epas1 and Epo in injured kidneys and myofibroblasts, which were reversed by ALK5 inhibitor SB431542. CONCLUSION C3H10T1/2 cells possessed the property of HIF2α-dependent Epo expression in REP pericytes. TGF-β1 induced not only the transition to myofibroblasts but also a repressive effect on Epas1-Epo axis in C3H10T1/2 cells. The repressive effect of TGF-β1 on Epas1-Epo axis was confirmed in REP pericytes in vivo. Inhibition of TGF-β1-ALK5 signaling might provide a novel treatment to rescue EPO expression in REP pericytes of injured kidney.
Collapse
Affiliation(s)
- Hong-Mou Shih
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road Section 1, Taipei, 100, Taiwan.,Division of Nephrology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Szu-Yu Pan
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Chih-Jen Wu
- Division of Nephrology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan.,Department of Pharmacology, Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsiang Chou
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City, Taiwan
| | - Chun-Yuan Chen
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fan-Chi Chang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Ting Chen
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan.,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital Jin-Shan Branch, New Taipei City, Taiwan
| | - Wen-Chih Chiang
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsing-Chen Tsai
- Division of Chest Medicine, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Ming Chen
- Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shuei-Liong Lin
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road Section 1, Taipei, 100, Taiwan. .,Department of Integrated Diagnostics and Therapeutics, National Taiwan University Hospital, Taipei, Taiwan. .,Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan. .,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
20
|
Teng FY, Jiang ZZ, Guo M, Tan XZ, Chen F, Xi XG, Xu Y. G-quadruplex DNA: a novel target for drug design. Cell Mol Life Sci 2021; 78:6557-6583. [PMID: 34459951 PMCID: PMC11072987 DOI: 10.1007/s00018-021-03921-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/13/2021] [Accepted: 08/12/2021] [Indexed: 02/08/2023]
Abstract
G-quadruplex (G4) DNA is a type of quadruple helix structure formed by a continuous guanine-rich DNA sequence. Emerging evidence in recent years authenticated that G4 DNA structures exist both in cell-free and cellular systems, and function in different diseases, especially in various cancers, aging, neurological diseases, and have been considered novel promising targets for drug design. In this review, we summarize the detection method and the structure of G4, highlighting some non-canonical G4 DNA structures, such as G4 with a bulge, a vacancy, or a hairpin. Subsequently, the functions of G4 DNA in physiological processes are discussed, especially their regulation of DNA replication, transcription of disease-related genes (c-MYC, BCL-2, KRAS, c-KIT et al.), telomere maintenance, and epigenetic regulation. Typical G4 ligands that target promoters and telomeres for drug design are also reviewed, including ellipticine derivatives, quinoxaline analogs, telomestatin analogs, berberine derivatives, and CX-5461, which is currently in advanced phase I/II clinical trials for patients with hematologic cancer and BRCA1/2-deficient tumors. Furthermore, since the long-term stable existence of G4 DNA structures could result in genomic instability, we summarized the G4 unfolding mechanisms emerged recently by multiple G4-specific DNA helicases, such as Pif1, RecQ family helicases, FANCJ, and DHX36. This review aims to present a general overview of the field of G-quadruplex DNA that has progressed in recent years and provides potential strategies for drug design and disease treatment.
Collapse
Affiliation(s)
- Fang-Yuan Teng
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, and Sichuan Clinical Research Center for Nephropathy, and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- State Key Laboratory of Crop Stress Biology for Arid Areas and College of Life Sciences, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zong-Zhe Jiang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, and Sichuan Clinical Research Center for Nephropathy, and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Man Guo
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, and Sichuan Clinical Research Center for Nephropathy, and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiao-Zhen Tan
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, and Sichuan Clinical Research Center for Nephropathy, and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Feng Chen
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xu-Guang Xi
- State Key Laboratory of Crop Stress Biology for Arid Areas and College of Life Sciences, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- LBPA, Ecole Normale Supérieure Paris-Saclay, CNRS, Université Paris Saclay, 61, Avenue du Président Wilson, 94235, Cachan, France.
| | - Yong Xu
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, and Sichuan Clinical Research Center for Nephropathy, and Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|