1
|
Chu C, Huang Y, Cao L, Ji S, Zhu B, Shen Q. Role of macrophages in peritoneal dialysis-associated peritoneal fibrosis. Ren Fail 2025; 47:2474203. [PMID: 40044628 PMCID: PMC11884102 DOI: 10.1080/0886022x.2025.2474203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
Peritoneal dialysis (PD) can be used as renal replacement therapy when chronic kidney disease (CKD) progresses to end-stage renal disease. However, peritoneal fibrosis (PF) is a major cause of PD failure. Studies have demonstrated that PD fluid contains a significantly larger numbers of macrophages compared with the healthy individuals. During PD, macrophages can secrete cytokines to keep peritoneal tissue in sustained low-grade inflammation, and participate in the regulation of fibrosis-related signaling pathways, such as NF-κB, TGF-β/Smad, IL4/STAT6, and PI3K/AKT. A series of basic pathological changes occurs in peritoneal tissues, including epithelial mesenchymal transformation, overgeneration of neovasculature, and abnormal deposition of extracellular matrix. This review focuses on the role of macrophages in promoting PF during PD, summarizes the targets of macrophage-related inhibition of fibrosis, and provides new ideas for clinical research on delaying PF, maintaining the function and integrity of peritoneum, prolonging duration of PD as a renal replacement modality, and achieving longer survival in CKD patients.
Collapse
Affiliation(s)
- Chenling Chu
- Department of Clinical Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Ying Huang
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Public Health and Preventive Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Luxi Cao
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Shuiyu Ji
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Bin Zhu
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Quanquan Shen
- Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
- Department of Nephrology, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, Guizhou, China
| |
Collapse
|
2
|
Yang X, Luo Q, Wu Z, Wang C, Yang Y, Zheng L, Li K, Zhao L, Jurong Y. Tanshinone IIA reduces tubulointerstitial fibrosis by suppressing GSDMD-mediated pyroptosis. PHARMACEUTICAL BIOLOGY 2025; 63:364-373. [PMID: 40331369 PMCID: PMC12064128 DOI: 10.1080/13880209.2025.2498166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 03/02/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
CONTEXT Tanshinone IIA (Tan IIA), a bioactive compound derived from the traditional Chinese herb Salvia miltiorrhiza (Family Lamiaceae, Authority Bunge), is well-known for its protective effects in various kidney diseases. However, its role in obstructive nephropathy has not been thoroughly investigated. OBJECTIVE This study aimed to explore the protective effects of Tan IIA in a mouse model of unilateral ureteral obstruction (UUO) and to elucidate the cellular and molecular mechanisms underlying these effects. MATERIALS AND METHODS Gasdermin D (GSDMD) knockout mice and their wild-type (WT) littermates underwent UUO surgery, with Tan IIA treatment administered 24 h prior. Human proximal tubular cells (HK-2 cells) were treated with TGF-β1 to induce fibrosis (50 ng/mL for 24 h), followed by Tan IIA treatment (5 μM) for an additional 3 h. RESULTS Tan IIA significantly reduced the expression of extracellular matrix (ECM) components, including collagen I, α-smooth muscle actin (α-SMA), vimentin and fibronectin, in UUO mice. Tan IIA attenuated GSDMD-mediated pyroptosis. However, in GSDMD knockout mice subjected to UUO, the protective effects of Tan IIA on ECM gene expression and collagen deposition in the tubular interstitium were reduced. In vitro studies showed that Tan IIA reduced GSDMD activation and fibronectin protein expression in HK-2 cells. DISCUSSION AND CONCLUSIONS Tan IIA may mitigate GSDMD-mediated pyroptosis in renal tubular epithelial cells (RTECs) and reduce kidney fibrosis, highlighting its potential as a therapeutic strategy to prevent the progression of kidney disease after ureteral obstruction.
Collapse
Affiliation(s)
- Xueling Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinglin Luo
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhifen Wu
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunxuan Wang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanjing Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Luquan Zheng
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ke Li
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Lei Zhao
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Jurong
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Yuan G, Liao Z, Liang P, Cai L, Zhou K, Yin T, Chen W, Darwish O, Xu C, Han M, Li Z. Noninvasive grading of renal interstitial fibrosis and prediction of annual renal function loss in chronic kidney disease: the optimal solution of seven MR diffusion models. Ren Fail 2025; 47:2480751. [PMID: 40133226 PMCID: PMC11938308 DOI: 10.1080/0886022x.2025.2480751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
OBJECTIVES To explore the optimal choice of seven diffusion models (DWI, IVIM, DKI, CTRW, FROC, SEM, and sADC) to assess renal interstitial fibrosis (IF) and annual renal function loss in chronic kidney disease (CKD). METHODS One hundred thirty-three CKD patients and 30 controls underwent multi-b diffusion sequence scans. Patients were divided into the training, testing, and temporal external validation sets. Least absolute shrinkage and selection operator regression and logistic regression were used to select the optimal metrics for distinguishing the mild from moderate-to-severe IF. The performances of imaging, clinical, and combined models were compared. A linear mixed-effects model calculated estimated glomerular filtration rate (eGFR) slope, and multiple linear regression assessed the association between metrics and 1-3-year eGFR slopes. RESULTS The training, testing, and temporal external validation sets had 75, 30, and 28 patients, respectively. The combined model incorporating cortical fIVIM, MKDKI and eGFR was superior to the clinical model combining the eGFR and 24-hour urinary protein in all sets (net reclassification index [NRI] > 0, p < 0.05). Decision curve analysis showed the combined model provided greater net clinical benefit across most thresholds. Fifty-two, 35, and 16 patients completed 1-, 2-, and 3-year follow-ups. After adjusting for covariates, cortical fIVIM correlated with the 1-year eGFR slope (β = 30.600, p = 0.001), and cortical αSEM correlated with the 2- and 3-year eGFR slopes (β = 44.859, p = 0.002; β = 95.631, p = 0.019). CONCLUSIONS A combined model of cortical fIVIM, MKDKI and eGFR provides a useful comprehensive tool for grading IF, with cortical fIVIM and αSEM as potential biomarkers for CKD progression.
Collapse
Affiliation(s)
- Guanjie Yuan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhouyan Liao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Liang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingli Cai
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kailun Zhou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Yin
- MR Research Collaboration Team, Siemens Healthineers Ltd., Shanghai, China
| | - Wei Chen
- MR Research Collaboration Team, Siemens Healthineers Ltd., Shanghai, China
| | - Omar Darwish
- MR Application Predevelopment, Siemens Healthcare, Erlangen, Germany
| | - Chuou Xu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Han
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
4
|
Li CSZ, Yu B, Gao Q, Dong HL, Li ZL. The critical role of ion channels in kidney disease: perspective from AKI and CKD. Ren Fail 2025; 47:2488139. [PMID: 40289808 PMCID: PMC12039425 DOI: 10.1080/0886022x.2025.2488139] [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: 11/03/2024] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Ion channels, particularly those in the transient receptor potential (TRP) family, play key roles in cellular stress responses like inflammation and apoptosis, significantly impacting renal disease progression. Some channels such as TRPV1, TRPM2, TRPC6 impact renal pathology by mediating detrimental calcium influx, exacerbating oxidative stress, and promoting inflammatory pathways. Their activities are especially pronounced in conditions like ischemia and nephrotoxicity, common in acute kidney injury, and persist into chronic kidney injury, influencing fibrosis and nephron loss. Additionally, potassium and sodium channels like Kir4.1, KATP, and ENaC play critical roles in maintaining electrolyte balance and cellular energy under stress conditions. Further exploration of ion channel functionality and regulation is necessary to clarify their roles in renal disease. This review summarizes the involvement of ion channels in AKI and CKD and examines their potential clinical value in diagnosing and treating kidney disease.
Collapse
Affiliation(s)
- Chen sui zi Li
- Department of Pharmacy, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Yu
- Department of Pharmacy, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Gao
- Department of Pharmacy, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong liang Dong
- Pediatric Translational Medicine Institute, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi ling Li
- Department of Pharmacy, Shanghai Children’s Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Himanshu, Gunjan, Pandey RP, Mukherjee R, Chang CM. Meta-analysis study of the therapeutic impact of Mesenchymal stem cells derived exosomes for chronic kidney diseases. Biochem Biophys Rep 2025; 43:102072. [PMID: 40524833 PMCID: PMC12169747 DOI: 10.1016/j.bbrep.2025.102072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 05/19/2025] [Accepted: 05/28/2025] [Indexed: 06/19/2025] Open
Abstract
Mesenchymal stem cell-derived exosomes (EXOs) represent a promising avenue for treating chronic kidney diseases (CKD), though their precise impact remains somewhat elusive. To address this gap, we conducted a systematic analysis, scouring databases and clinical trial repositories for relevant studies from 2019 to 2023. Seventeen papers were meticulously selected for their focus on mesenchymal stem cell-derived exosomes (MSC-EXOs) and their potential in CKD treatment. Our comprehensive meta-analysis, incorporating 15 preclinical and 6 clinical studies, underscores the efficacy of MSC-EXOs in improving renal function while attenuating tubular injury, inflammation, apoptosis, collagen deposition, and renal fibrosis. Notably, post-treatment with MSC-EXOs exhibited significant associations with various CKD markers, with pooled proportions indicating a considerable impact on blood urea nitrogen (BUN) and serum creatinine (SCR) levels. Subgroup analyses based on animal models further elucidated heterogeneity within the studies. In conclusion, MSC-EXOs demonstrate promise in enhancing renal function and reducing CKD risk, as evidenced by both preclinical and clinical data. Their efficacy in lowering SCR and BUN levels while enhancing filtration rate suggests MSC-EXOs as a viable and secure alternative to cell-based therapies, thereby providing valuable insights for personalized CKD treatments despite inherent limitations.
Collapse
Affiliation(s)
- Himanshu
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District Taoyuan City, 33302, Taiwan, ROC
| | - Gunjan
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District Taoyuan City, 33302, Taiwan, ROC
| | - Ramendera Pati Pandey
- School of Health Sciences and Technology (SoHST) UPES, Bidholi, Dehradun, 248007, Uttarakhand, India
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, Haryana, India
| | - Riya Mukherjee
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District Taoyuan City, 33302, Taiwan, ROC
| | - Chung-Ming Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District Taoyuan City, 33302, Taiwan, ROC
- Master & Ph.D. Program in Biotechnology Industry, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, Taoyuan City, 33302, Taiwan, ROC
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, Taoyuan City, 33302, Taiwan, ROC
| |
Collapse
|
6
|
Peng Z, Huang X, Pan Y, Li W, Hu H, Chen X, Zhang Z, Hu J, Qi Y, Chen W, Cui X, Liu H, Liang W, Ding G, Chen Z. USP22 promotes angiotensin II-induced podocyte injury by deubiquitinating and stabilizing HMGB1. Cell Signal 2025; 131:111771. [PMID: 40154587 DOI: 10.1016/j.cellsig.2025.111771] [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: 01/03/2025] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Chronic kidney disease (CKD) remains a significant global health burden, with hypertensive nephropathy (HN) as one of its primary causes. Podocyte injury is a key factor in the progression of CKD. However, the molecular mechanisms underlying angiotensin II-induced podocyte injury remain incompletely understood. Ubiquitin-specific protease 22 (USP22) has been reported to facilitate a range of cellular processes, including cell proliferation and apoptosis. However, the role of USP22 in HN pathogenesis is unclear. METHODS The expression of USP22 was assessed in kidney samples from hypertensive nephropathy patients, angiotensin II-induced hypertensive nephropathy mouse models, and cultured podocytes treated with angiotensin II. Podocyte-specific USP22 knockout mice were used to investigate the effects of USP22 deletion on podocyte injury and inflammation. RESULTS USP22 expression was significantly upregulated in kidneys of HN patients, angiotensin II-induced mouse models, and cultured podocytes. Podocyte-specific deletion of USP22 markedly reduced angiotensin II-induced podocyte injury and inflammatory responses. Furthermore, we identified high-mobility group box protein 1 (HMGB1) as a protein that interacts with USP22. USP22 deubiquitinated and stabilized HMGB1 through K48-linked ubiquitination. Downregulation of USP22 expression improved kidney function and pathological changes in HN by promoting HMGB1 degradation. CONCLUSION This study identifies USP22 as a key regulator of angiotensin II-induced podocyte injury and inflammation through its interaction with HMGB1. Our findings revealed that following glomerular injury, damage and shedding of tubular cells also occurred. Targeting the USP22-HMGB1 axis offers a promising therapeutic strategy for treating hypertensive nephropathy and other types of CKD.
Collapse
Affiliation(s)
- Zhuan Peng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxiao Huang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yangbin Pan
- Division of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
| | - Weiwei Li
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinghua Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Yue Qi
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenjie Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofei Cui
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyan Liu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
7
|
Lin L, Shen D, Su Y, Zhang Z, Yu J, Xu C, Pan K, Wang Y, Zhang L, Jin S, Song N, Ding X, Teng J, Xu X. Magnesium Lithospermate B Protects Against Ischemic AKI-to-CKD progression via regulating the KLF5/CDK1/Cyclin B1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156765. [PMID: 40262199 DOI: 10.1016/j.phymed.2025.156765] [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: 09/24/2024] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is the primary cause of acute kidney injury (AKI), which can result in chronic kidney disease (CKD) with renal fibrosis. Magnesium lithospermate B (Mlb), a bioactive compound produced from Salvia miltiorrhiza Bunge, exerts nephroprotective effects against AKI. However, the significance of Mlb in the evolution of IRI-induced AKI in patients with CKD remains unclear. Notably, the specific mechanisms underlying the putative antifibrotic activities of Mlb during this progression remain to be fully elucidated. PURPOSE This study sought to explore the therapeutic benefits of Mlb in AKI-to-CKD progression and uncover the potential mechanisms, with a special interest in its effects on renal fibrosis and cell cycle regulation. STUDY DESIGN AND METHODS Unilateral ischemia/reperfusion (UIR)-induced mouse AKI-to-CKD progression (in vivo) and HK-2 cells with TGF-β-induced fibrosis model (in vitro) were used in the study. The beneficial effects of Mlb on renal fibrosis and cell cycle-related signaling pathways were investigated using histological analysis, molecular assays, network pharmacology, and RNA sequencing. RESULTS Mlb treatment significantly reduced renal dysfunction, inflammation, apoptosis, and the G2/M phase cell cycle stalling in mice 14 days post-UIR-induced AKI, subsequently improving renal fibrosis. Mechanistically, Mlb promotes the activity of the CDK1/Cyclin B1 signaling pathway, thereby alleviating the G2/M phase cell cycle stalling. Network pharmacology and RNA sequencing analyses identified the KLF5/CDK1/Cyclin B1 signaling pathway as a potential target of the antifibrotic effects of Mlb, which was further verified in both in vivo and in vitro experiments. The KLF5 inhibitor ML264 attenuated the protective effects of Mlb by reducing CDK1/Cyclin B1 expression and reinstating the G2/M phase cell cycle stalling, highlighting the critical role of this pathway in Mlb-mediated renal protection. CONCLUSIONS Mlb decreases renal fibrosis by inhibiting the G2/M phase cell cycle stalling via the KLF5/CDK1/Cyclin B1 signaling pathway during AKI-to-CKD progression. Our findings offer new insight into the therapeutic potential of Mlb in preventing CKD progression following AKI and identify a previously unrecognized mechanism involving the KLF5/CDK1/Cyclin B1 pathway.
Collapse
Affiliation(s)
- Liyu Lin
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, PR China; Xiamen Clinical Quality Control Center of Nephrology, Xiamen, 361015, PR China
| | - Daoqi Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Yiqi Su
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, PR China; Xiamen Clinical Quality Control Center of Nephrology, Xiamen, 361015, PR China
| | - Zhen Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Jinbo Yu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Chenqi Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Kunming Pan
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, 20032, PR China
| | - Yaqiong Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Lin Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Shi Jin
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China
| | - Jie Teng
- Department of Nephrology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, 361015, PR China; Xiamen Clinical Quality Control Center of Nephrology, Xiamen, 361015, PR China; Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China.
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China; Kidney and Dialysis Institute of Shanghai, Shanghai, 200032, PR China; Shanghai Medical Center for Kidney Diseases, Shanghai, 200032, PR China; Shanghai Key Laboratory for Kidney Diseases and Blood Purification, Shanghai, 200032, PR China.
| |
Collapse
|
8
|
Huang Q, Huang F, Chen C, Xiao P, Liu J, Gao Y. Machine-learning model based on ultrasomics for non-invasive evaluation of fibrosis in IgA nephropathy. Eur Radiol 2025; 35:3707-3720. [PMID: 39853332 DOI: 10.1007/s00330-025-11368-9] [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: 06/05/2024] [Revised: 12/02/2024] [Accepted: 12/19/2024] [Indexed: 01/26/2025]
Abstract
OBJECTIVES To develop and validate an ultrasomics-based machine-learning (ML) model for non-invasive assessment of interstitial fibrosis and tubular atrophy (IF/TA) in patients with IgA nephropathy (IgAN). MATERIALS AND METHODS In this multi-center retrospective study, 471 patients with primary IgA nephropathy from four institutions were included (training, n = 275; internal testing, n = 69; external testing, n = 127; respectively). The least absolute shrinkage and selection operator logistic regression with tenfold cross-validation was used to identify the most relevant features. The ML models were constructed based on ultrasomics. The Shapley Additive Explanation (SHAP) was used to explore the interpretability of the models. Logistic regression analysis was employed to combine ultrasomics, clinical data, and ultrasound imaging characteristics, creating a comprehensive model. A receiver operating characteristic curve, calibration, decision curve, and clinical impact curve were used to evaluate prediction performance. RESULTS To differentiate between mild and moderate-to-severe IF/TA, three prediction models were developed: the Rad_SVM_Model, Clinic_LR_Model, and Rad_Clinic_Model. The area under curves of these three models were 0.861, 0.884, and 0.913 in the training cohort, and 0.760, 0.860, and 0.894 in the internal validation cohort, as well as 0.794, 0.865, and 0.904 in the external validation cohort. SHAP identified the contribution of radiomics features. Difference analysis showed that there were significant differences between radiomics features and fibrosis. The comprehensive model was superior to that of individual indicators and performed well. CONCLUSIONS We developed and validated a model that combined ultrasomics, clinical data, and clinical ultrasonic characteristics based on ML to assess the extent of fibrosis in IgAN. KEY POINTS Question Currently, there is a lack of a comprehensive ultrasomics-based machine-learning model for non-invasive assessment of the extent of Immunoglobulin A nephropathy (IgAN) fibrosis. Findings We have developed and validated a robust and interpretable machine-learning model based on ultrasomics for assessing the degree of fibrosis in IgAN. Clinical relevance The machine-learning model developed in this study has significant interpretable clinical relevance. The ultrasomics-based comprehensive model had the potential for non-invasive assessment of fibrosis in IgAN, which helped evaluate disease progress.
Collapse
Affiliation(s)
- Qun Huang
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fangyi Huang
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chengcai Chen
- Department of Ultrasound, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Pan Xiao
- Department of Ultrasound, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiali Liu
- Department of Ultrasound, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yong Gao
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
9
|
Tao Y, Zhang W, Liu D, Cao H, Yi X, Deng X, Li P, Shen X, Zhou H. Succinate Facilitates CD4 + T Cell Infiltration and CCL1 Production to Promote Myofibroblast Activation and Renal Fibrosis in UUO Mice. J Inflamm Res 2025; 18:7827-7840. [PMID: 40538778 PMCID: PMC12178263 DOI: 10.2147/jir.s510637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/19/2025] [Indexed: 06/22/2025] Open
Abstract
Aim Obstructive nephropathy is a leading cause of kidney injury and fibrosis, which is always associated with metabolic aberrations and chronic inflammation. Succinate is an important intermediate metabolite involved in inflammatory responses and various diseases. However, the precise pathogenic mechanisms of succinate in obstructive nephropathy remain to be elucidated. Methods Succinate was supplemented in the drinking water to study its impact on the pathogenesis of obstructive nephropathy induced by unilateral ureteral obstruction (UUO) in mice. Kidney fibrosis, injury, inflammatory cytokines, and infiltrated immune cells were analyzed. Transcriptome analysis and in vitro studies were performed to study the cellular and molecular mechanisms by which succinate regulates CD4+ T cells and renal fibrosis. Results Kidney proteomics revealed that the tricarboxylic acid (TCA) cycle and mitochondrial dysfunction were the hallmarks of obstructive nephropathy. Succinate was significantly accumulated in the obstructed kidneys. Succinate supplementation promoted UUO-induced renal fibrosis, injury, and inflammation. Moreover, succinate facilitated renal infiltration of CD4+ T cells by upregulating the T-cell chemokines CXCL9 and CXCL10. Transcriptome analysis suggested that succinate promoted CD4+ T cell activation and induced the production of CCL1, which mediated the transition of fibroblasts to myofibroblasts through the ERK signaling pathway. Recombinant CCL1 treatment promoted UUO-induced renal fibrosis and inflammation. Conclusion Our study uncovers the important role of succinate in mediating T-cell response that orchestrates the pathogenesis of obstructive nephropathy. Targeting succinate accumulation may be a therapeutic strategy for the treatment of obstructive nephropathy.
Collapse
Affiliation(s)
- Yuandong Tao
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Wei Zhang
- Department of Urology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People’s Republic of China
| | - Dehong Liu
- Department of Pediatric Surgery, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, People’s Republic of China
| | - Hualin Cao
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Xiaoyu Yi
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xiangling Deng
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Pin Li
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xiaoli Shen
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Huixia Zhou
- Department of Pediatric Urology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
10
|
Huang JY, Yu HN. The role of the Nrf2 pathway in inhibiting ferroptosis in kidney disease and its future prospects. Pathol Res Pract 2025; 272:156084. [PMID: 40527053 DOI: 10.1016/j.prp.2025.156084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 06/12/2025] [Accepted: 06/12/2025] [Indexed: 06/19/2025]
Abstract
Kidney disease (KD) has gradually become a major social and economic burden on the healthcare system. Recent studies highlight ferroptosis as a critical mechanism in the progression of these conditions. Recognized for its essential role in renal pathogenesis, ferroptosis is attracting increasing research attention and emerging as a key focus of investigation. The Nrf2 signaling pathway, known for its regulatory influence on ferroptosis, plays a central role in this context. Activation of the nuclear factor E2-related factor 2 (Nrf2) pathway and the subsequent attenuation of ferroptosis present substantial opportunities as novel therapeutic targets for managing kidney injury (KI). This article summarizes the latest mechanism of action of the Nrf2 pathway in ferroptosis, explores how the Nrf2 pathway affects ferroptosis in KD therapy, and investigates the potential therapeutic effects of natural products targeting the Nrf2 pathway. These natural products have been shown to inhibit ferroptosis through the Nrf2 pathway, providing new insights and therapeutic strategies for the clinical and individualized management of KD.
Collapse
Affiliation(s)
- Jia-Yuan Huang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Hai-Ning Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
11
|
Peng H, Ren J, Zhao Y, Fang X, Wang X, Liu C, Wan Z. Unraveling the Connection between PCOS and renal Complications: Current insights and Future Directions. Diabetes Res Clin Pract 2025; 224:112235. [PMID: 40334925 DOI: 10.1016/j.diabres.2025.112235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025]
Abstract
Polycystic ovary syndrome (PCOS) represents the most prevalent endocrine disorder among women of reproductive age, affecting approximately 5-18% of females worldwide. Characterized by irregular ovulation, hyperandrogenism, and polycystic ovaries, hyperandrogenism is the defining feature. Recent evidence highlights that, in addition to its notable reproductive and metabolic consequences, PCOS may also contribute to an elevated risk of renal complications. This increased risk is attributed to chronic low-grade inflammation, hormonal dysregulation, and disturbances in lipid metabolism inherent to the condition. However, the pathological mechanisms, clinical manifestations, and progression of secondary renal damage in this cohort remain insufficiently studied. This review consolidates current understanding of the relationship between PCOS and chronic kidney disease (CKD), aiming to clarify potential mechanisms by which PCOS may induce secondary renal dysfunction, encompassing both direct renal impairment and indirect damage mediated through systemic alterations. Furthermore, it advocates for comprehensive management strategies to mitigate renal risks in patients with PCOS, emphasizing the necessity of multidisciplinary approaches and further research to address these critical gaps.
Collapse
Affiliation(s)
- Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Junyi Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Zhao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinyi Fang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiao Wang
- Department of Organ Transplantation, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, China.
| | - Zhengwei Wan
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
12
|
Ajiki T, Ozono T, Sera F, Yonishi H, Matsuda J, Aguirre C, Kakuda K, Yasuhara Y, Isaka Y, Sakata Y, Ikenaka K, Mochizuki H. Therapeutic effect of TTR siRNA on hereditary transthyretin amyloidosis (ATTRv) nephropathy. Amyloid 2025; 32:171-178. [PMID: 39988769 DOI: 10.1080/13506129.2025.2470369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/06/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Hereditary transthyretin amyloidosis (ATTRv) is a fatal disease that affects multiple organs. Up to 30% of patients with ATTRv also experience renal complications, including proteinuria and a decline in eGFR. Recently, new treatments for ATTRv, a tetramer stabiliser and transthyretin small interfering RNA (TTR siRNA) therapeutics, have emerged. However, the effectiveness of these new treatments on renal complications in ATTRv remains unknown. METHODS We retrospectively collected clinical data from ATTRv patients and analysed the relationship between the initial renal complications and age. We also examined whether the new treatments affected the clinical course of renal symptoms, using eGFR changes or longitudinal data on urine protein/albumin creatinine ratio. RESULTS A total of 16 patients' data were collected. Regarding their initial renal complications, we found that patients with proteinuria had an earlier age at onset than those with a decline in eGFR. Notably, longitudinal data showed that TTR siRNA therapeutics reduced proteinuria and increased serum protein, while none of the new treatments could demonstrate a significant improvement in the slope of eGFR decline. CONCLUSIONS We demonstrated that TTR siRNA therapeutics represent potential candidates for ATTRv nephropathy, despite the fact that their use has been limited to neurological symptoms to date.
Collapse
Affiliation(s)
- Takahiro Ajiki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tatsuhiko Ozono
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fusako Sera
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Yonishi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Matsuda
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - César Aguirre
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keita Kakuda
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yumiko Yasuhara
- Department of Diagnostic Pathology, Sakai City Medical Center, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kensuke Ikenaka
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
13
|
Yin L, Li F, Xiao T, Zhang J, Li Y, Luo J, Zhao J, Xiong J. Functional Performance Decline Outperforms Sarcopenia and Its Components in Predicting New-Onset Chronic Kidney Disease: A Nationwide Multicenter Study. Kidney Med 2025; 7:101005. [DOI: 10.1016/j.xkme.2025.101005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2025] Open
|
14
|
Tsai HJ, Wu CF, Li SS, Chen JJ, Hsieh CJ, Chen CC, Wang SL, Chen ML, Wu MT. Sex-specific association of co-exposures to melamine and phthalates in children with their early renal injury. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 374:126206. [PMID: 40210160 DOI: 10.1016/j.envpol.2025.126206] [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: 09/13/2024] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
Studies concerning the effect of co-exposure to melamine and phthalates on kidney function in children are rare. Thus, this study examines the above-mentioned relationship and their sex-different effect. Whether the exposure of the two chemicals from their mothers, when children were in the womb during the third trimester, affected renal injury markers in children afterwards is also examined. This study was from Taiwan Maternal and Infant Cohort Study cohort established in October 2012 to enroll third-trimester pregnant mothers up to May 2015. Their offspring were subsequently recruited between 2016 and 2020 as our study children. One-spot urine specimens were collected from both pregnant mothers (2012-2015) and study children (2016-2020) for the simultaneous measurement of melamine and 11 phthalate metabolites. Daily intakes of melamine and five phthalates, including DEHP (di-2-ethylhexylphthalate), DiBP (Dibutyl phthalate), DnBP (Di-n-butyl phthalate), BBzP (Butyl benzyl phthalate), and DEP (Diethyl phthalate), were estimated using a creatinine excretion-based model in both study children and their mothers. Two early markers of renal injury, microalbumin and N-acetyl-beta-D-glucosaminidas (NAG), were measured in urine samples of study children (2016-2020). A total of 552 eligible children were studied, with a mean age of 4 years. We found that boys in the highest quartile of estimated melamine intake (≥0.68 μg/kg/day) had significantly higher urine ACR levels and in the highest quartile of estimated phthalate intake of DEHP (≥5.36 μg/kg/day), DEP (≥0.89 μg/kg/day), and DiBP (≥1.19 μg/kg/day) had significantly higher urine NAG levels when compared to the combined three lowest quartile ones as comparison groups. No significant associations were found between their mothers' phthalates and melamine intake during the third trimester and urine ACR and NAG in children. We conclude that children (particularly boys) with high co-exposure of melamine and certain phthalate chemicals among children have increased early markers of kidney injury.
Collapse
Affiliation(s)
- Hui-Ju Tsai
- Department of Family Medicine, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Family Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Fang Wu
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan
| | - Sih-Syuan Li
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Jen Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Jung Hsieh
- Department of Public Health, Tzu Chi University, Hualien, Taiwan
| | - Chu-Chih Chen
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Shu-Li Wang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Mei-Lien Chen
- Institute of Environmental and Occupational Health Sciences, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Tsang Wu
- Department of Family Medicine, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
15
|
Liang W, Wu H, Long Q, Lin H, Lv X, Ma W, Wu T, Li A, Zheng Q, Guo J, Chen X, Guo J, Sun D. LKB1 activated by NaB inhibits the IL-4/STAT6 axis and ameliorates renal fibrosis through the suppression of M2 macrophage polarization. Life Sci 2025; 370:123564. [PMID: 40097066 DOI: 10.1016/j.lfs.2025.123564] [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: 12/07/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Renal fibrosis is a critical pathological characteristic of chronic kidney disease, and current antifibrotic therapies has limited efficacy. Sodium butyrate (NaB) has been shown to be highly effective in mitigating bleomycin-induced pulmonary fibrosis; however, its specific impact on renal fibrosis and the underlying mechanisms remain unclear. This study aims to elucidate the role and mechanism of NaB in renal fibrosis by using a mouse model of renal fibrosis induced through Unilateral Ureteral Obstruction (UUO) and folic acid (FA) administration. RESULTS NaB significantly decreased the distribution of collagen fibers in renal tissues and mitigated fibrosis in a dose-dependent manner. Further analysis indicated that NaB inhibited M2 macrophage polarization in the renal tissues of UUO model mice by blocking the phosphorylation of STAT6, hence reducing renal fibrosis. Additionally, in vitro experiments demonstrated that NaB inhibited fibroblast activation induced by M2 macrophages. Mechanistic studies revealed that NaB attenuates fibroblast activation and M2 macrophage polarization by upregulating LKB1 and inhibiting the activation of the STAT6 signaling pathway. CONCLUSION NaB may exert its effects by inhibiting the activation of the IL-4/STAT6 signaling pathway through the upregulation of LKB1, which suppress the polarization of M2 macrophages and consequently reduce renal fibrosis. These findings establish a theoretical foundation for NaB as a novel drug candidate for renal fibrosis and indicate its potential applicability in clinical treatments for this condition.
Collapse
Affiliation(s)
- Weifei Liang
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China; Center for Cancer and Immunology Research, State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, 510180 Guangzhou, Guangdong, China
| | - Haoyu Wu
- School of Public Health, Wenzhou Medical University, Wenzhou 325035, China; South Zhejiang Institute of Radiation Medicine and Nuclear Technology Application, Wenzhou 325809, China; Zhejiang Provincial Key Laboratory of Watershed Sciences and Health, Wenzhou Medical University, Wenzhou 325035, China
| | - Qishan Long
- Department of Urology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Hong Lin
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, 511518 Qingyuan, Guangdong, China
| | - Xiaoyu Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, 138 Yixueyuan Road, Xuhui District, Shanghai 200032, China
| | - Wen Ma
- Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
| | - Tao Wu
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
| | - Ai Li
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou 510000, China
| | - Qingyou Zheng
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
| | - Jinan Guo
- Department of Urology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China.
| | - Xiangqiu Chen
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China.
| | - Jing Guo
- Center of Oncology, Heyou Hospital, Shunde District, Foshan City 528306, Address:No. 1 of Heren Road, Junlan Community, Beijiao Town, Shunde District, Foshan City, Guangdong Province, China.
| | - Donglin Sun
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China.
| |
Collapse
|
16
|
Zuo S, Yuan H, Li X, Chen M, Peng R, Chen S, Zou X, Yang Y, Long H, Liu Z, Wang T, Guo B, Liu L. SMYD2 Promotes Renal Tubular Cell Apoptosis and Chronic Kidney Disease Following Cisplatin Nephrotoxicity. FASEB J 2025; 39:e70651. [PMID: 40391402 PMCID: PMC12090038 DOI: 10.1096/fj.202402703r] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 03/25/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025]
Abstract
The protein lysine methyltransferase 2 (SMYD2) can affect cell proliferation, differentiation, and survival through methylation of its histone and non-histone substrates. SMYD2 has been shown to act as an oncogene to promote disease progression in a variety of cancer diseases, but its role in chronic kidney diseases (CKD) pathogenesis has not been fully elucidated. This study aims to investigate the effect of SMYD2 on cisplatin-induced CKD and its underlying mechanisms. In this study, we found that cisplatin caused severe renal injury in mice, which was accompanied by the up-regulation of SMYD2 expression. AZ505 treatment significantly down-regulated cisplatin-induced renal injury and fibrosis. It also alleviated renal apoptosis and inhibited the phosphorylation level of NF-κB p65. Conditional knockdown of Smyd2 achieved similar effects as AZ505. In renal tubular epithelial cells, inhibition or silencing of SMYD2 down-regulated cisplatin-induced apoptotic response, while overexpression of SMYD2 induced apoptotic response and activated NF-κB in response to the up-regulation of SMYD2 expression. Up-regulation of SMYD2 induced interaction and phosphorylation of SMYD2 and NF-κB p65, and inhibition of NF-κB activation further suppressed cisplatin-induced NF-κB activation and apoptosis. The present study suggests that up-regulation of SMYD2 expression in cisplatin-induced CKD may promote apoptosis of renal tubular epithelial cells and accelerate the process of renal injury through NF-κB activation. SMYD2 may serve as a potential target for effective CKD treatment.
Collapse
Affiliation(s)
- Siyang Zuo
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Huixiong Yuan
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xia Li
- Key Laboratory of Kidney Disease Pathogenesis Research and Transformation ApplicationGuizhou UniversityGuiyangChina
| | - Ming Chen
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Rui Peng
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Siyu Chen
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xue Zou
- Key Laboratory of Kidney Disease Pathogenesis Research and Transformation ApplicationGuizhou UniversityGuiyangChina
| | - Yuan Yang
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Hehua Long
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Zeying Liu
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Teng Wang
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Bing Guo
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic DiseasesGuizhou Medical UniversityGui'an New DistrictChina
| | - Lirong Liu
- Center for Clinical LaboratoriesThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
- Guizhou Institute of Precision MedicineAffiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
17
|
Ko MS, Yun JY, Kim S, Kim MO, Go SH, Jin HJ, Koh EH. Mesenchymal Stem Cells Mediated Suppression of GREM2 Inhibits Renal Epithelial-Mesenchymal Transition and Attenuates the Progression of Diabetic Kidney Disease. Int J Stem Cells 2025; 18:158-172. [PMID: 39757007 PMCID: PMC12122246 DOI: 10.15283/ijsc24113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease worldwide. Despite advancements in various treatments, the prevalence of DKD continues to rise, leading to a significant increase in the demand for dialysis and kidney transplantation. This study aimed to evaluate the effects of a Small cell+Ultra Potent+Scale UP cell (SMUP-Cell), a type of human umbilical cord blood-derived mesenchymal stem cell, on DKD in the db/db mouse model of type 2 diabetes mellitus. After administering SMUP-Cells via tail vein injection in db/db mice, the animals were monitored over a three-month period. The db/db mice exhibited an increased urine albumin-to-creatinine ratio (UACR). However, the administration of SMUP-Cells resulted in a reduction of the UACR. The expression levels of desmin, α-smooth muscle actin, and fibronectin-markers of epithelial-mesenchymal transition (EMT)-as well as kidney injury molecule 1, a sensitive marker of tubular injury, were significantly elevated in db/db mice. Treatment with SMUP-Cells ameliorated all of these changes. Notably, Gremlin isoform 2 (Grem2) exhibited the most significant difference in expression according to the transcriptome analysis. The elevated expression of Grem2 in db/db mice was significantly reduced following SMUP-Cell treatment. In vitro, treatment with high glucose and cholesterol induced Grem2 expression in renal tubular epithelial cells (RTECs), while Grem2 knockdown effectively prevented fibrosis and senescence induced by high glucose and cholesterol in RTECs. These observations suggest that SMUP-Cells inhibit the progression of DKD by inhibiting EMT through the reduction of Grem2 expression in RTECs.
Collapse
Affiliation(s)
- Myoung Seok Ko
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, Korea
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Young Yun
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, Korea
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Serin Kim
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, Korea
| | - Mi-Ok Kim
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, Korea
| | - Sang-hyeok Go
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam, Korea
| | - Eun Hee Koh
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, Korea
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Cuevas-Delgado P, Miguel V, Lamas S, Barbas C, Rupérez FJ. Multiplatform Metabolomic Profiling of the Unilateral Ureteral Obstruction Murine Model of CKD. Int J Mol Sci 2025; 26:4933. [PMID: 40430073 PMCID: PMC12112560 DOI: 10.3390/ijms26104933] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
In chronic kidney disease (CKD) research, animal models such as the unilateral ureteral obstruction (UUO) rodent model are crucial to understanding disease progression, particularly renal fibrosis. Despite its widespread use, the molecular mechanisms driving CKD remain incompletely understood. Given the interplay between metabolism and fibrosis, a comprehensive metabolomic analysis of UUO renal tissue is necessary. This study involved untargeted multiplatform analysis using liquid chromatography (LC), gas chromatography (GC), and capillary electrophoresis (CE) coupled with mass spectrometry (MS) to examine murine kidney tissue from the UUO model. The results highlight metabolic changes associated with tubulointerstitial fibrosis, which affect pathways such as the tricarboxylic acid (TCA) cycle, the urea cycle, and lipid metabolism. In particular, fibrosis impacts the lipidomic profile, with decreases in most lipid classes and increases in specific glycerophospholipids, hexosylceramides, and cholesterol esters. These findings demonstrate the value of a multiplatform approach in elucidating metabolic alterations in CKD, providing information on the underlying molecular mechanisms and paving the way for further research.
Collapse
Affiliation(s)
- Paula Cuevas-Delgado
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid 28660, Spain; (P.C.-D.); (C.B.)
| | - Verónica Miguel
- Program of Physiological and Pathological Processes, Centro de Biología Molecular “Severo Ochoa” (CBMSO, CSIC-UAM), c. Nicolás Cabrera 1, Madrid 28049, Spain; (V.M.); (S.L.)
| | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular “Severo Ochoa” (CBMSO, CSIC-UAM), c. Nicolás Cabrera 1, Madrid 28049, Spain; (V.M.); (S.L.)
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid 28660, Spain; (P.C.-D.); (C.B.)
| | - Francisco J. Rupérez
- Centre for Metabolomics and Bioanalysis (CEMBIO), School of Pharmacy, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid 28660, Spain; (P.C.-D.); (C.B.)
| |
Collapse
|
19
|
Tejedor-Santamaria L, Marquez-Exposito L, Villacampa A, Marchant V, Battaglia-Vieni A, Rayego-Mateos S, Rodrigues-Diez RR, Santos FM, Valentijn FA, Knoppert SN, Broekhuizen R, Ruiz-Torres MP, Goldschmeding R, Ortiz A, Peiró C, Nguyen TQ, Ramos AM, Ruiz-Ortega M. CCN2 Activates Cellular Senescence Leading to Kidney Fibrosis in Folic Acid-Induced Experimental Nephropathy. Int J Mol Sci 2025; 26:4401. [PMID: 40362638 PMCID: PMC12072722 DOI: 10.3390/ijms26094401] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Cellular communication network factor 2 (CCN2, also known as CTGF) is a complex protein that regulates numerous cellular functions. This biomolecule exhibits dual functions, depending on the context, and can act as a matricellular protein or as a growth factor. CCN2 is an established marker of fibrosis and a well-known mediator of kidney damage, involved in the regulation of inflammation, extracellular matrix remodeling, cell death, and activation of tubular epithelial cell (TECs) senescence. In response to kidney damage, cellular senescence mechanisms are activated, linked to regeneration failure and progression to fibrosis. Our preclinical studies using a total conditional CCN2 knockout mouse demonstrate that CCN2 plays a significant role in the development of a senescence phenotype after exposure to a nephrotoxic agent. CCN2 induces cell growth arrest in TECs, both in the early phase and in the chronic phase of folic acid nephropathy (FAN), associated with cell-death/necroinflammation and fibrosis, respectively. Renal CCN2 overexpression was found to be linked to excessive collagen accumulation in tubulointerstitial areas, microvascular rarefaction, and a decline in renal function, which were observed three weeks following the initial injury. All these findings were markedly diminished in conditional CCN2 knockout mice. In the FAN model, injured senescent TECs are associated with microvascular rarefaction, and both were modulated by CCN2. In primary cultured endothelial cells, as previously described in TECs, CCN2 directly induced senescence. The findings collectively demonstrate the complexity of CCN2, highlight the pivotal role of cellular senescence as an important mechanism in renal injury, and underscore the critical function of this biomolecule in kidney damage progression.
Collapse
Affiliation(s)
- Lucia Tejedor-Santamaria
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Laura Marquez-Exposito
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Alicia Villacampa
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (A.V.); (C.P.)
- Vascular Pharmacology and Metabolism (FARMAVASM) Group, IdiPAZ, 28029 Madrid, Spain
| | - Vanessa Marchant
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Antonio Battaglia-Vieni
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
| | - Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Raul R. Rodrigues-Diez
- Department of Cell Biology, School of Medicine, Complutense University, 28040 Madrid, Spain;
| | - Fatima Milhano Santos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| | - Floris A. Valentijn
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Sebastian N. Knoppert
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Roel Broekhuizen
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | | | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Alberto Ortiz
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28049 Madrid, Spain
| | - Concepción Peiró
- Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (A.V.); (C.P.)
- Vascular Pharmacology and Metabolism (FARMAVASM) Group, IdiPAZ, 28029 Madrid, Spain
| | - Tri Q. Nguyen
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.B.); (R.G.); (T.Q.N.)
| | - Adrián M. Ramos
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28049 Madrid, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Department of Medicine, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.T.-S.); (L.M.-E.); (V.M.); (A.B.-V.); (S.R.-M.); (F.M.S.)
- Instituto de Salud Carlos III., 28029 Madrid, Spain; (A.O.); (A.M.R.)
| |
Collapse
|
20
|
Zhou X, Jiang C, Song B, Sun S, Yan Z. Association between dietary index for gut microbiota and chronic kidney disease: A cross-sectional study from U.S. population. Prev Med Rep 2025; 53:103060. [PMID: 40264750 PMCID: PMC12013330 DOI: 10.1016/j.pmedr.2025.103060] [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: 11/27/2024] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
Objective: Emerging evidence suggests that diet modulates gut microbiota, which in turn influences chronic kidney disease (CKD) progression. This study investigates the association between the newly proposed Dietary Index for Gut Microbiota (DI-GM) and the prevalence and prognosis of CKD. Methods: This cross-sectional study analyzed data from the U.S. National Health and Nutrition Examination Survey 2007-2020. DI-GM scores were calculated based on dietary intake of 14 food components, categorized as beneficial or unfavorable. Weighted linear regression model, logistic regression model, and restricted cubic spline analysis were used to assess the associations of DI-GM with CKD. Results: The prevalence of CKD among 28,512 participants was 17.4 %. Higher DI-GM was negatively associated with CKD prevalence (OR = 0.967, 95 %CI: 0.939-0.995, p = 0.026) and with very high-risk prognosis (OR = 0.877, 95 %CI: 0.821-0.937, p < 0.001). Beneficial DI-GM components were significantly associated with lower CKD risk (OR = 0.928, 95 %CI: 0.892-0.966, p < 0.001), while no significant association was observed for unfavorable components. Higher DI-GM and beneficial DI-GM levels were linearly associated with improved CKD prognosis (p for trend <0.001). Coffee and fiber were primary contributors to both the prevalence and prognosis of CKD, while whole grains primarily impacted its prognosis. Conclusions: Higher DI-GM, driven by beneficial dietary components, is associated with reduced CKD prevalence and improved prognosis. These findings suggest that promoting beneficial dietary patterns to enhance gut microbiota may play a pivotal role in CKD management.
Collapse
Affiliation(s)
- Xuanzhen Zhou
- Department of Urology, Ningbo Clinical Research Center for Urological Disease, Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseases, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Chengxiao Jiang
- Department of Urology, Xiangshan Frist People's Hospital Medical and Health Group, Xiangshan, China
| | - Baiyang Song
- Department of Urology, Ningbo Clinical Research Center for Urological Disease, Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseases, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shuben Sun
- Department of Urology, Ningbo Clinical Research Center for Urological Disease, Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseases, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Zejun Yan
- Department of Urology, Ningbo Clinical Research Center for Urological Disease, Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseases, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
21
|
Abbad L, Esteve E, Chatziantoniou C. Advances and challenges in kidney fibrosis therapeutics. Nat Rev Nephrol 2025; 21:314-329. [PMID: 39934355 DOI: 10.1038/s41581-025-00934-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Chronic kidney disease (CKD) is a major global health burden that affects more than 10% of the adult population. Current treatments, including dialysis and transplantation, are costly and not curative. Kidney fibrosis, defined as an abnormal accumulation of extracellular matrix in the kidney parenchyma, is a common outcome in CKD, regardless of disease aetiology, and is a major cause of loss of kidney function and kidney failure. For this reason, research efforts have focused on identifying mediators of kidney fibrosis to inform the development of effective anti-fibrotic treatments. Given the prominent role of the transforming growth factor-β (TGFβ) family in fibrosis, efforts have focused on inhibiting TGFβ signalling. Despite hopes raised by the efficacy of this approach in preclinical models, translation into clinical practice has not met expectations. Antihypertensive and antidiabetic drugs slow the decline in kidney function and could slow fibrosis but, owing to the lack of technologies for in vivo renal imaging, their anti-fibrotic effect cannot be truly assessed at present. The emergence of new drugs targeting pro-fibrotic signalling, or enabling cell repair and cell metabolic reprogramming, combined with better stratification of people with CKD and the arrival of nanotechnologies for kidney-specific drug delivery, open up new perspectives for the treatment of this major public health challenge.
Collapse
Affiliation(s)
- Lilia Abbad
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Emmanuel Esteve
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France.
| |
Collapse
|
22
|
Hoda F, Arshad M, Khan MA, Habib MA, Najmi AK. Diagnostic Potential of Dp-ucMGP as a Biomarker for Early Detection of Diabetic Kidney Disease in Patients With Type 2 Diabetes Mellitus: A Cross-Sectional Study. Nephrology (Carlton) 2025; 30:e70050. [PMID: 40329762 DOI: 10.1111/nep.70050] [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: 02/16/2025] [Revised: 04/10/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
AIM The limited evidence reports the relationship between plasma dephosphorylated uncarboxylated matrix Gla protein (dp-ucMGP) and Diabetic kidney disease (DKD) in patients with Type 2 Diabetes Mellitus (T2DM). Therefore, the present study aimed to evaluate the diagnostic potential of dp-ucMGP in DKD among T2DM patients. METHOD This cross-sectional study was conducted from December 2023 to January 2025, including 75 T2DM patients. Participants were classified into three groups based on urinary albumin-to-creatinine ratio (UACR): Normoalbuminuria, microalbuminuria, and macroalbuminuria with n = 25 in each group. Pearson correlation analysis was performed to assess the relationship between dp-ucMGP and other biochemical parameters. Receiver Operating Characteristic (ROC) curve analysis was used to evaluate the diagnostic potential of dp-ucMGP for early detection of DKD. A p value < 0.05 was considered statistically significant. RESULTS The plasma dp-ucMGP levels were significantly higher in T2DM patients in the macroalbuminuria group, with a mean of 1069.86 ± 417.56 pmol/L, followed by the microalbuminuria (842.72 ± 342.02 pmol/L) and normoalbuminuria (586.38 ± 336.15 pmol/L) groups. Similarly, higher dp-ucMGP levels were observed in patients with DKD severity stage IV (1401.53 ± 401.49 pmol/L). A negative correlation with eGFR (r = -0.807, p < 0.0001) and a positive correlation with age, serum creatinine, UACR, blood urea, uric acid and triglycerides were observed. The area under the curve (AUC) was 0.913 (95% CI: 0.820-0.960; p < 0.0001) for dp-ucMGP. CONCLUSION In conclusion, our findings revealed that plasma dp-ucMGP could be a potential biomarker to predict the early detection of DKD in patients with T2DM.
Collapse
Affiliation(s)
- Farazul Hoda
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mawrah Arshad
- Department of Pharmacology, Integral University, Lucknow, Uttar Pradesh, India
| | - Mohammad Ahmad Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Anwar Habib
- Department of Medicine, Hamdard Institute of Medical Sciences & Research, Jamia Hamdard, New Delhi, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
23
|
Boaz M, Kelly JT, Wesson DE. Introduction to the Lifestyle Medicine and Kidney Health Series. Clin J Am Soc Nephrol 2025; 20:739-741. [PMID: 40063110 PMCID: PMC12097175 DOI: 10.2215/cjn.0000000703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/04/2025] [Indexed: 04/10/2025]
Affiliation(s)
- Mona Boaz
- Department of Nutrition Sciences, Ariel University, Ariel, Israel
| | - Jaimon T. Kelly
- Centre for Health Services Research, Centre for Online Health, University of Queensland, Brisbane, Queensland, Australia
| | - Donald E. Wesson
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
24
|
Qin X, Liu X, Xiao W, Luo Q, Xia L, Zhang C. Interpretable Deep-learning Model Based on Superb Microvascular Imaging for Noninvasive Diagnosis of Interstitial Fibrosis in Chronic Kidney Disease. Acad Radiol 2025; 32:2730-2738. [PMID: 39690075 DOI: 10.1016/j.acra.2024.11.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024]
Abstract
RATIONALE AND OBJECTIVES To develop an interpretable deep learning (XDL) model based on superb microvascular imaging (SMI) for the noninvasive diagnosis of the degree of interstitial fibrosis (IF) in chronic kidney disease (CKD). METHODS We included CKD patients who underwent renal biopsy, two-dimensional ultrasound, and SMI examinations between May 2022 and October 2023. Based on the pathological IF score, they were divided into two groups: minimal-mild IF (≤25%) and moderate-severe IF (>25%). An XDL model based on the SMI while establishing an ultrasound radiomics model and a color doppler ultrasonography (CDUS) model as the control group. The utility of the proposed model was evaluated using the receiver operating characteristic curve (ROC) and decision curve analysis. RESULTS In total, 365 CKD patients were included herein. In the validation group, AUCs of the ROC curves for the DL, ultrasound radiomics, and CDUS models were 0.854, 0.784, and 0.745, respectively. In the test group, AUCs of the ROC curve for the DL ultrasound radiomics, and CDUS models were 0.824, 0.792, and 0.752, respectively. The pie chart and heat map based on Shapley additive explanations (SHAP) provided substantial interpretability for the model. CONCLUSION Compared with the ultrasound radiomics and CDUS models, the DL model based on the SMI had higher accuracy in the noninvasive judgment of the degree of IF in CKD patients. Pie and heat maps based on Shapley can explain which image regions are helpful in diagnosing the degree of IF.
Collapse
Affiliation(s)
- Xiachuan Qin
- Department of Ultrasound, Chengdu Second People's Hospital, Chengdu, Sichuan 610000, China (X.Q.); Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Xiaoling Liu
- Department of Ultrasound, Beijing Anzhen Hospital Nanchong Hospital, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nan Chong, Sichuan 637000, China (X.L., W.X.); Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Weihan Xiao
- Department of Ultrasound, Beijing Anzhen Hospital Nanchong Hospital, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College (University), Nan Chong, Sichuan 637000, China (X.L., W.X.)
| | - Qi Luo
- Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Linlin Xia
- Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.)
| | - Chaoxue Zhang
- Department of Ultrasound, The first affiliated hospital of Anhui Medical University, Hefei, Anhui 230022, China (X.Q., X.L., Q.L., L.X., C.Z.).
| |
Collapse
|
25
|
He LM, Deng X, Ni LH, Cai SQ, Chen J, Liao Z, Zhang M, Shui H, Zhu KK, Wu S, Gao P, Sarotti AM, Hong K, Wu XY, Cai YS. Penicipyrrolidines A-N, pyrrolidine derivatives with inhibitory effects on EMT and fibroblast activation from the mangrove-derived fungus Penicillium sp. DM27. MARINE LIFE SCIENCE & TECHNOLOGY 2025; 7:313-327. [PMID: 40417247 PMCID: PMC12102406 DOI: 10.1007/s42995-025-00282-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 02/10/2025] [Indexed: 05/27/2025]
Abstract
An investigation of the mangrove-derived fungus Penicillium sp. DM27 led to the isolation of 19 new compounds, including three pairs of piperidinone enantiomers ( ±)-1, ( ±)-2, and ( ±)-3, two pairs of pyrrolidinone enantiomers ( ±)-4 and ( ±)-5, and nine pyrrolidine derivatives 6-14. The structures of 1-14 were elucidated through NMR and HRESIMS analysis, coupled with experimental and calculated ECD spectroscopy and the modified Mosher method. Quantitative real time PCR and Western bolt analyses revealed that 11 blocked EMT in TGF-β1-treated HK-2 cells and suppressed fibroblast activation in TGF-β1-stimulated NIH-3T3 cells. Molecular simulations demonstrated that compound 11 could dock ADAM17, showing a high negative binding affinity. Additionally, the overexpression of ADAM17 by lentiviral infection triggered renal tubular EMT, while compound 11 suppressed this process. Overall, our research suggests that pyrrolidine derivatives may be potential therapeutic agents for the treatment of fibrotic kidney disease. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-025-00282-0.
Collapse
Affiliation(s)
- Li-Ming He
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Xuan Deng
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Li-Hua Ni
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Shi-Qi Cai
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Jinhu Chen
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Zejin Liao
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Mengke Zhang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Hua Shui
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Kong-Kai Zhu
- Advanced Medical Research Institute, Shandong University, Jinan, 250012 China
| | - Song Wu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Ping Gao
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Ariel M. Sarotti
- Facultad de Ciencias Bioquímicas y Farmaceuticas, Instituto de Química Rosario (CONICET), Universidad Nacional de Rosario, Suipacha 531, 2000 Rosario, Argentina
| | - Kui Hong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - Xiao-Yan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| | - You-Sheng Cai
- Department of Nephrology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education and School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071 China
| |
Collapse
|
26
|
Hu J, Pang X, Liang X, Shao X, Xia Q, Sun J, Wang Y, Wang G, Li S, Zha L, Guo J, Peng C, Huang P, Ding Y, Jin C, He N, Huang Y, Gui S. Raspberry ameliorates renal fibrosis in rats with chronic kidney disease via the PI3K/Akt pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156589. [PMID: 40056634 DOI: 10.1016/j.phymed.2025.156589] [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: 10/15/2024] [Revised: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Renal fibrosis is a hallmark of chronic kidney disease (CKD). In traditional Chinese medicine, Rubus chingii Hu (raspberry) is believed to have kidney-tonifying properties. However, whether raspberry can effectively treat CKD, along with the specific active compounds and underlying mechanisms, remains unclear. PURPOSE This study aims to investigate the potential of raspberries in treating CKD and elucidate the mechanisms involved. METHODS CKD model was established in rats using adenine. The effects of raspberry treatment on CKD were assessed through macroscopic observations and pathological changes in the kidney. The expression of fibrotic proteins in renal tissues was analyzed to evaluate the impact of raspberry on renal fibrosis. Data mining combined with compositional analysis were employed to identify the active ingredients, targets, and pathways of raspberry that may improve CKD. Subsequently, Western blotting and immunofluorescence analysis were conducted to confirm the involvement of the PI3K/AKT signaling pathway in the renoprotective mechanism of raspberry. RESULTS Raspberry treatment significantly alleviated renal pathological damage, fibrosis and inflammation in model rats, showing effects comparable to irbesartan (Avapro). Chemical composition analysis and network pharmacology predicted AKT1 as the core target, and the PI3K/AKT pathway plays a pivotal role in mediating the therapeutic effects of raspberry extract in CKD. Molecular docking studies further confirmed that active compounds in raspberry have a strong binding affinity with AKT1. Western blotting and immunofluorescence results demonstrated that raspberry inhibited phosphorylation, thereby suppressing the PI3K/AKT pathway, leading to its antifibrotic effect on the kidney. CONCLUSION Raspberry was firstly discovered to potentially treat CKD by alleviating renal fibrosis through inhibition of the PI3K/AKT pathway. Raspberry, as a medicinal and edible traditional herb, could serve as a promising therapeutic agent or health supplement for improving renal fibrosis and slowing CKD progression.
Collapse
Affiliation(s)
- Jingjing Hu
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Xingyuan Pang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Xiao Liang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Xinyuan Shao
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Qijun Xia
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Jianwen Sun
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Yuxiao Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Guichun Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Shuhan Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Liangping Zha
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Jian Guo
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui 230012, PR China
| | - Chengjun Peng
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui 230012, PR China
| | - Peng Huang
- Department of Neurology, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230031, PR China
| | - Yang Ding
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China
| | - Cheng Jin
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui 230012, PR China
| | - Ning He
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui 230012, PR China
| | - Yuzhe Huang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui 230012, PR China.
| | - Shuangying Gui
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230012, PR China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui 230012, PR China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui 230012, PR China; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, PR China; MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, Anhui 230012, PR China; Anhui Engineering Research Center for Quality Improvement and Utilization of Genuine Chinese Medicinal Materials, Hefei, Anhui 230012, PR China.
| |
Collapse
|
27
|
Wu YJ, Yang YR, Yan YL, Yang HY, Du JR. Targeting mitochondrial dysfunction: an innovative strategy for treating renal fibrosis. Mol Cell Biochem 2025:10.1007/s11010-025-05297-w. [PMID: 40299265 DOI: 10.1007/s11010-025-05297-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
The incidence and hospitalization rate of kidney disease, especially end-stage renal disease, have increased significantly, which seriously endangers the health of patients. Mitochondria are the core organelles of cellular energy metabolism, and their dysfunction can lead to kidney energy supply insufficiency and oxidative stress damage, which has become a global public health problem. Studies have shown that the disturbance of mitochondrial quality control mechanisms, including mitochondrial dynamics, autophagy, oxidative stress regulation and biosynthesis, is closely related to the occurrence and development of renal fibrosis (RF). As a multicellular pathological process, RF involves the injury and shedding of podocytes, the transdifferentiation of renal tubular epithelial cells, the activation of fibroblasts, and the infiltration of macrophages, among which the mitochondrial dysfunction plays an important role. This review systematically elaborates the molecular mechanisms of mitochondrial damage during RF progression, aiming to provide theoretical foundations for developing novel therapeutic strategies to delay RF advancement.
Collapse
Affiliation(s)
- Yi-Jin Wu
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yan-Rong Yang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Ling Yan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Han-Yinan Yang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
28
|
Pang N, Zhao N, An C, Li K, Li P, Wang N, Li J, Cheng X, Zheng N, Guo D, Xiong X. Development of a Long-Acting Interleukin-11 Antagonist for the Treatment of Renal Fibrosis. J Med Chem 2025; 68:8429-8438. [PMID: 40198895 DOI: 10.1021/acs.jmedchem.4c03185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Renal fibrosis, a key progression of chronic kidney disease (CKD), remains a major challenge in nephrology, with no FDA-approved drugs specifically targeting this condition. Interleukin-11 (IL-11) has emerged as a potential therapeutic target for renal fibrosis. In this study, we identified the antifibrotic effects of a recombinant human IL-11 analogue, IL-11-6M, in a mouse model of unilateral ureteral obstruction (UUO). We generated additional IL-11-6M variants via an optimized Escherichia coli expression system, with one variant (D46C) exhibiting comparable efficacy. Further modified through cysteine-specific PEGylation, analogue 13 demonstrated similar potency to IL-11-6M with an IC50 value of 61.5 ± 26.2 nM and maintained strong binding affinity to IL-11Rα (KD = 3.0 nM). Notably, analogue 13 exhibited a prolonged half-life and showed significant therapeutic effects in the UUO-induced renal fibrosis model. These findings suggest analogue 13 should be a promising candidate for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Ningning Pang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Na Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Chunmei An
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Keqiang Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peiying Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Naiyuan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jian Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xing Cheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Nan Zheng
- Center for Translational Research, Shenzhen Bay Laboratory, Shenzhen 518000, Guangdong, China
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xiaochun Xiong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
29
|
Liu L, Li Q, Zhang G. Systemic inflammation accelerates the development of focal segmental glomerulosclerosis in a mouse model of adriamycin induced nephrosis. Sci Rep 2025; 15:14304. [PMID: 40274873 PMCID: PMC12022324 DOI: 10.1038/s41598-025-96125-0] [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: 02/05/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Research indicates that minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) may reflect varying severities of the same underlying condition, with inflammation potentially facilitating the progression from MCD to FSGS. The aim of this study was to determine the whether systemic inflammation accelerated the progression from MCD to FSGS in a mouse model of Adriamycin-induced nephrosis. In this model, systemic inflammation induced transient proteinuria without significant serum biochemical alterations or significant renal histological changes in control mice that did not develop nephrotic syndrome. In contrast, both mice with Adriamycin-induced nephrosis mice and mice with Adriamycin-induced nephrosis with systemic inflammation showed histological features of MCD at week 4 and progressive exacerbation of FSGS. However, the glomerular lesions of mice with Adriamycin-induced nephrosis in a state of systemic inflammation were more obvious than those of mice with Adriamycin-induced nephrosis. These findings suggest that systemic inflammation may hasten histological development from MCD to FSGS in this mouse model.
Collapse
Affiliation(s)
- Lian Liu
- Department of pediatrics, University-town Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Qiu Li
- Department of Nephrology; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Gaofu Zhang
- Department of Nephrology; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
30
|
Deng B, Su P, Cheng L, Zhang J, Zhang X, Yu T, Bao G, Yan T, Yin Y, Shen L, Wang D, Hong L, Miao X, Yang W, Wang C, Xie J, Wang R. Iterative Optimization Yields Stapled Peptides with Superior Pharmacokinetics and Potency for Renal Fibrosis Treatment. J Med Chem 2025; 68:8516-8529. [PMID: 40199779 DOI: 10.1021/acs.jmedchem.5c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Renal fibrosis, resulting from myofibroblast-mediated excessive extracellular matrix (ECM) deposition, lacks effective treatments. Novel peptide DR3penA developed by our group showed therapeutic potential for fibrotic diseases; however, its application was hindered by poor stability and bioavailability. To address this unmet need, we implemented stepwise optimization of DR3penA. The conformationally restricted analogs designed via structural predictions enhanced both activity and stability. Through structure-activity relationship analysis and cleavage site mapping, introducing unnatural amino acids improved stability. Fatty acid modifications conferred fibroblast-selective cytotoxicity and improved pharmacokinetics. After several rounds of progressive modification, peptide 27 exhibited remarkable stability, with a 5.68-fold extended half-life compared to DR3penA. Following profibrotic stimuli, peptide 27 effectively inhibited myofibroblast activation, epithelial-mesenchymal transition, and ECM synthesis. It also attenuated renal fibrosis in a unilateral ureteral obstruction model. Our study leverages multiple modifications that integrate cell and animal models to identify peptide 27 as a promising candidate for renal fibrosis therapy.
Collapse
Affiliation(s)
- Bochuan Deng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Ping Su
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Lu Cheng
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jiao Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Xiang Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Tingli Yu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Tiantian Yan
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Yue Yin
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Lei Shen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Dan Wang
- Institute of Basic Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Liang Hong
- Guangdong Provincial Key Laboratory of Chiral Molecular and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Xiaokang Miao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Wenle Yang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Chenyu Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Junqiu Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
| | - Rui Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, Lanzhou 730000, China
- Institute of Materia Medica and Research Unit of Peptide Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
31
|
Zhang P, Liu YH, Xiong WY, Fan YB, Zhu XL, Zhou K, Li H. Association of long-term remnant cholesterol with the incidence of chronic kidney disease in a high-risk population. Hormones (Athens) 2025:10.1007/s42000-025-00651-5. [PMID: 40249462 DOI: 10.1007/s42000-025-00651-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 04/02/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND AND AIMS Chronic kidney disease (CKD) is creating an ever heavier global health burden with population ageing. This study aimed to examine the longitudinal associations of remnant cholesterol (RC) with CKD morbidity in a large high-risk population (type 2 diabetes and hypertension). METHODS A total of 11,881 participants who participated in annual health examinations from 2021 to 2023 were included in our analysis. The Cox proportional hazards model was performed to analyze the associations of baseline RC, cumulative RC, and variability of RC with CKD morbidity. The cross-lagged panel analysis was used to examine the temporal relationship between RC and renal function. RESULTS The results of the multivariable-adjusted models showed that higher baseline, cumulative RC, and variability of RC were related to higher risks of developing CKD, the adjusted HR (95% CI) comparing tertile 3 with tertile 1 were 1.26 (95% CI 1.10-1.45), 1.33 (95% CI 1.16-1.52), 1.36 (95% CI 1.20-1.55), respectively. Stratified analysis found that gender did not change these associations. Compared with individuals in the low cumulative and variability RC group, those in the high cumulative and variability RC group had a 1.62 times higher risk of CKD (95% CI: 1.34-1.96). The cross-lagged panel analysis showed that the increase in RC levels may precede the decrease in eGFR. CONCLUSIONS High baseline level, cumulative exposure to RC, and variability of RC are associated with increased CKD risk. Therefore, monitoring RC-related parameters is crucial to delay the occurrence and development of CKD in high-risk populations.
Collapse
Affiliation(s)
- Ping Zhang
- Nanchang First Hospital, Nanchang, China
| | - Yu-Hong Liu
- The Collaboration Unit for State Key Laboratory of Infectious Disease Prevention and Control, Jiangxi Provincial Health Commission Key Laboratory of Pathogenic Diagnosis and Genomics of Emerging Infectious Diseases, Nanchang Center for Disease Control and Prevention, Nanchang, China
| | - Wen-Yan Xiong
- The Collaboration Unit for State Key Laboratory of Infectious Disease Prevention and Control, Jiangxi Provincial Health Commission Key Laboratory of Pathogenic Diagnosis and Genomics of Emerging Infectious Diseases, Nanchang Center for Disease Control and Prevention, Nanchang, China
| | - Yi-Bing Fan
- The Collaboration Unit for State Key Laboratory of Infectious Disease Prevention and Control, Jiangxi Provincial Health Commission Key Laboratory of Pathogenic Diagnosis and Genomics of Emerging Infectious Diseases, Nanchang Center for Disease Control and Prevention, Nanchang, China
| | - Xiao-Lin Zhu
- The Collaboration Unit for State Key Laboratory of Infectious Disease Prevention and Control, Jiangxi Provincial Health Commission Key Laboratory of Pathogenic Diagnosis and Genomics of Emerging Infectious Diseases, Nanchang Center for Disease Control and Prevention, Nanchang, China
| | - Kun Zhou
- The Collaboration Unit for State Key Laboratory of Infectious Disease Prevention and Control, Jiangxi Provincial Health Commission Key Laboratory of Pathogenic Diagnosis and Genomics of Emerging Infectious Diseases, Nanchang Center for Disease Control and Prevention, Nanchang, China
| | - Hui Li
- The Collaboration Unit for State Key Laboratory of Infectious Disease Prevention and Control, Jiangxi Provincial Health Commission Key Laboratory of Pathogenic Diagnosis and Genomics of Emerging Infectious Diseases, Nanchang Center for Disease Control and Prevention, Nanchang, China.
| |
Collapse
|
32
|
Zhao X, Zhang S, Wu M, Zhang B, Wan G, Zhang M, Li J, Fei Z, Zhu G, Jiang S, Xiao M, Liu W, Zhao Z, Huang B, Ran J. High urea promotes mitochondrial fission and functional impairments in astrocytes inducing anxiety-like behavior in chronic kidney disease mice. Metab Brain Dis 2025; 40:186. [PMID: 40244426 DOI: 10.1007/s11011-025-01612-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
High urea can induce depression and anxiety. Activation of astrocytes is closely associated with psychiatric disorders. However, the pathological mechanism of whether high urea affects astrocyte structure and function to induce anxiety-like behaviors remain unclear. We established a high-urea chronic kidney disease (CKD) mouse model and found that these mice exhibited elevated levels of anxiety through behavioral experiments. Immunofluorescence and transmission electron microscopy studies of astrocytes revealed a decrease in density and branching of mPFC astrocytes. Additionally, we observed a significant reduction in ATP and BDNF levels in the mPFC and primary astrocytes of CKD mice induced by high urea. Analysis of gene expression differences in astrocytes between WT and high-urea mice indicated alterations in mitochondrial dynamics-related signaling pathways in astrocytes. We established a high-urea primary astrocyte model to assess mitochondrial function and levels of fusion and fission proteins. Treatment of primary astrocytes with high urea led to mitochondrial fragmentation and downregulation of Mfn2 expression. These results suggested that high urea downregulates Mfn2 expression in mPFC astrocytes, induced mitochondrial fusion-fission abnormalities, disrupted astrocyte energy metabolism, and promoted high-urea-related anxiety. Mfn2 may represent a potential therapeutic target for high-urea-related anxiety.
Collapse
Affiliation(s)
- Xi Zhao
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Shengyao Zhang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Mengna Wu
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Binyun Zhang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Guoran Wan
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Meng Zhang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Jing Li
- Department of Stem Cell and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Zhuo Fei
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Guoqi Zhu
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Shaoqiu Jiang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Mohan Xiao
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Wanjia Liu
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Zhelun Zhao
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Boyue Huang
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.
| | - Jianhua Ran
- Department of Anatomy, Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
33
|
Lang F, Li Y, Yao R, Jiang M. Osteopontin in Chronic Inflammatory Diseases: Mechanisms, Biomarker Potential, and Therapeutic Strategies. BIOLOGY 2025; 14:428. [PMID: 40282293 PMCID: PMC12024743 DOI: 10.3390/biology14040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025]
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), atherosclerosis, and inflammatory bowel disease (IBD), pose major global health concerns. These disorders are marked by persistent inflammation, immune system dysfunction, tissue injury, and fibrosis, ultimately leading to severe organ dysfunction and diminished quality of life. Osteopontin (OPN), a multifunctional extracellular matrix protein, plays a crucial role in immune regulation, inflammation, and tissue remodeling. It promotes immune cell recruitment, stimulates pro-inflammatory cytokine production, and contributes to fibrosis through interactions with integrins and CD44 receptors. Additionally, OPN activates key inflammatory pathways, including NF-κB, MAPK, and PI3K/Akt, further aggravating tissue damage in chronic inflammatory conditions. Our review highlights the role of OPN in chronic inflammation, its potential as a biomarker, and its therapeutic implications. We explore promising preclinical approaches, such as monoclonal antibodies, small molecule inhibitors, and natural compounds like curcumin, which have demonstrated potential in mitigating OPN-driven inflammation. However, challenges persist in selectively targeting OPN while maintaining its essential physiological roles, including bone remodeling and wound healing. Our review offers insights into therapeutic strategies and future research directions.
Collapse
Affiliation(s)
- Fuyuan Lang
- Queen Mary College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330001, China; (F.L.); (Y.L.); (R.Y.)
| | - Yuanheng Li
- Queen Mary College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330001, China; (F.L.); (Y.L.); (R.Y.)
| | - Ruizhe Yao
- Queen Mary College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330001, China; (F.L.); (Y.L.); (R.Y.)
| | - Meixiu Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang 330031, China
| |
Collapse
|
34
|
Liu H, Yang Z, Li J, Zhang J, Sun C. Expanding the horizons of bicyclol in multiple diseases: Mechanisms, therapeutic implications and challenges. Eur J Pharmacol 2025; 993:177381. [PMID: 39954842 DOI: 10.1016/j.ejphar.2025.177381] [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: 11/01/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Bicyclol, a drug stemmed from the traditional Chinese medicine Schisandra chinensis, has been widely utilized in clinical practice due to its efficacy and safety to manage hepatopathy. Its diverse biological properties-including antiviral, anti-inflammatory, antifibrotic, immunomodulatory, antioxidative, antisteatotic, and antitumor effects-underscore its significant medicinal effects in versatile hepatic disorders, incorporating viral hepatitis, non-alcoholic fatty liver disease, hepatocellular carcinoma, acute hepatic failure, hepatic fibrosis as well as drug-induced liver injury. Furthermore, ongoing researches into the molecular mechanisms, biological activities and mode of actions concerning bicyclol have uncovered its potential therapeutic implications in other multiple diseases/conditions. Studies have indicated promising efficacy pertaining to bicyclol to treat idiopathic pulmonary fibrosis, acute lung injury, cerebral ischemia/reperfusion injury, renal dysfunction, renal cell carcinoma, and cardiovascular diseases. Accordingly, this narrative review article summarizes the current understanding of diverse biological activities and underpinning mechanisms of bicyclol across a range of diseases, as well as its pharmacokinetics, toxicity profile and shed light on future perspectives.
Collapse
Affiliation(s)
- Heng Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, China; Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, East Street 6, Tianjin Airport Economic Area, Tianjin, 300308, China
| | - Ziyi Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, China
| | - Jia Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, China
| | - Jie Zhang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, China.
| | - Chao Sun
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052, China; Department of Gastroenterology, Tianjin Medical University General Hospital Airport Hospital, East Street 6, Tianjin Airport Economic Area, Tianjin, 300308, China.
| |
Collapse
|
35
|
Yang Y, Gong S, Zhou C, Xin W, Qin S, Yao M, Lan Q, Liao W, Zhao J, Huang Y. REST contributes to renal fibrosis through inducing mitochondrial energy metabolism imbalance in tubular epithelial cells. Cell Commun Signal 2025; 23:176. [PMID: 40200371 PMCID: PMC11980176 DOI: 10.1186/s12964-025-02166-3] [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: 11/20/2024] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Renal fibrosis represents the final common pathological manifestation of chronic kidney disease (CKD), yet the underlying mechanism remains elusive, and there is still a lack of effective targeted therapeutic strategy. Although previous research indicated that repressor element 1-silencing transcription factor (REST) contributed to acute kidney injury (AKI) in renal tubular epithelial cells (RTECs), its specific contribution to renal fibrosis and associated mechanisms remains largely unexplored. METHODS Renal biopsies from CKD patients were collected to evaluate the expression of REST. Kidney-specific Rest conditional knockout (Cdh16-Cre/Restflox/flox) mice were generated and employed unilateral ureter obstruction (UUO) models to investigate the role of REST in renal fibrosis. RNA sequencing was performed to elucidate the mechanism. Mitochondrial function was evaluated by transmission electron microscopy (TEM), reactive oxygen species (ROS), oxygen consumption rates (OCR), extracellular acidifcation rate (ECAR) and adenosine triphosphate (ATP). The severity of renal fibrosis was assessed through Western blot, immunofluorescent staining and immumohistochemical staining. Bioinformatic prediction, dual luciferase reporter gene assay, point mutation and chromatin immunoprecipitation (ChIP) assay were utilized to clarify the molecular mechanism. RESULTS REST was significantly up-regulated in the kidney tissues from CKD patients, UUO-induced fibrotic mouse models and TGF-β1-incubated RTECs. Notably, kidney-specific knockout of Rest prominently alleviated renal fibrosis by improving mitochondrial energy metabolism and restoring fatty acid oxidation. Mechanically, REST disturbed mitochondrial energy metabolism through repressing the transcription of oxoglutarate dehydrogenase-like (OGDHL) via directly binding to its promotor region. Further, pharmacological inhibition of REST using the specific REST inhibitor, X5050, significantly ameliorated the progression of renal fibrosis both in vitro and in vivo. CONCLUSIONS Our explorations revealed the upregulation of REST in renal fibrosis disrupts mitochondrial energy metabolism through transcriptionally suppressing OGDHL, which may act as a promising therapeutic target for renal fibrosis.
Collapse
Affiliation(s)
- Yingxian Yang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Shuiqin Gong
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Chun Zhou
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Wang Xin
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Shaozong Qin
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Mengying Yao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Qigang Lan
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Wenhao Liao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China
| | - Jinghong Zhao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China.
| | - Yinghui Huang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
36
|
Addario G, Moroni L, Mota C. Kidney Fibrosis In Vitro and In Vivo Models: Path Toward Physiologically Relevant Humanized Models. Adv Healthc Mater 2025; 14:e2403230. [PMID: 39906010 PMCID: PMC11973949 DOI: 10.1002/adhm.202403230] [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: 08/27/2024] [Revised: 01/14/2025] [Indexed: 02/06/2025]
Abstract
Chronic kidney disease (CKD) affects over 10% of the global population and is a leading cause of mortality. Kidney fibrosis, a key endpoint of CKD, disrupts nephron tubule anatomy and filtration function, and disease pathomechanisms are not fully understood. Kidney fibrosis is currently investigated with in vivo models, that gradually support the identification of possible mechanisms of fibrosis, but with limited translational research, as they do not fully recapitulate human kidney physiology, metabolism, and molecular pathways. In vitro 2D cell culture models are currently used, as a starting point in disease modeling and pharmacology, however, they lack the 3D kidney architecture complexity and functions. The failure of several therapies and drugs in clinical trials highlights the urgent need for advanced 3D in vitro models. This review discusses the urinary system's anatomy, associated diseases, and diagnostic methods, including biomarker analysis and tissue biopsy. It evaluates 2D and in vivo models, highlighting their limitations. The review explores the state-of-the-art 3D-humanized in vitro models, such as 3D cell aggregates, on-chip models, biofabrication techniques, and hybrid models, which aim to mimic kidney morphogenesis and functions. These advanced models hold promise for translating new therapies and drugs for kidney fibrosis into clinics.
Collapse
Affiliation(s)
- Gabriele Addario
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| | - Lorenzo Moroni
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| | - Carlos Mota
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityER Maastricht6229The Netherlands
| |
Collapse
|
37
|
Luo L, Wang J, Zhao J, Yang B, Ma W, Lin J. Dental pulp stem cells derived exosomes inhibit ferroptosis via regulating the Nrf2-keap1/GPX4 signaling pathway to ameliorate chronic kidney disease injury. Tissue Cell 2025; 93:102670. [PMID: 39667244 DOI: 10.1016/j.tice.2024.102670] [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: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Chronic kidney disease (CKD) has long represented a substantial global health challenge. Regrettably, current therapeutic interventions exhibit limited efficacy in halting the progression of CKD. Ferroptosis may play a crucial role in CKD, as indicated by substantial evidence. Dental pulp stem cell-derived exosomes (DPSC-Exos) possess advantages such as abundant sources and low immunogenicity, holding promising prospects in CKD treatment. METHODS This study constructed a mouse CKD model to investigate the therapeutic effects of DPSC-Exos. First, we successfully extracted and identified DPSC-Exos. Then, mice were randomly divided into sham, PBS, CKD, and CKD+Exos groups. Our study determined the expression of ferroptosis-related pathway molecules Nrf2, GPX4, Keap1, and HO-1 in each group. Finally, we detected the expression levels of inflammatory factors, TNF-α, IL-1β, and IL-6, at the injury site. RESULTS Mice treated with DPSC-Exos showed increased expression of the ferroptosis inhibitory factor Nrf2 and its downstream regulatory factors GPX4 and HO-1, while the expression of Keap1 decreased. The expression of TNF-α, IL-1β, and IL-6 also decreased. CONCLUSION DPSC-Exos may help inhibit ferroptosis through the Keap1-Nrf2/GPX4 pathway and reduce the inflammatory response at the injury site, revealing their potential therapeutic effects on CKD.
Collapse
Affiliation(s)
- Lin Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao; Department of spine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing Wang
- Department of spine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jie Zhao
- Department of Magnetic Resonance Imaging, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Bin Yang
- Department of Magnetic Resonance Imaging, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Jiaru Lin
- Department of nephropathy, the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
38
|
Chen Z, Jiang J, Gunda ST, Han X, Wu C, Ying MTC, Chen F. Ultrasonic renal length as an indicator of renal fibrosis severity in non-diabetic patients with chronic kidney disease. Clin Exp Nephrol 2025; 29:460-468. [PMID: 39560816 PMCID: PMC11937224 DOI: 10.1007/s10157-024-02598-0] [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: 09/07/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Debate continues regarding the potential of the ultrasonic renal length to serve as an indicator for evaluating the advancement of renal fibrosis in chronic kidney disease (CKD). This study investigates the independent association between renal length and renal fibrosis in non-diabetic CKD patients and assesses its diagnostic performance. METHODS From April 2019 to December 2021, 144 non-diabetic patients diagnosed with CKD who underwent a renal ultrasound examination and kidney biopsy were prospectively enrolled. Patients were categorized into the mild fibrosis group (n = 70) and the moderate-severe group (n = 74) based on the extent of fibrotic involvement. Ultrasonic renal length was measured from pole-to-pole in the coronal plane. A receiver operating characteristic (ROC) curve, multivariable logistic regression analysis, and a generalized additive model were performed. RESULTS A negative linear correlation was found between renal length and moderate-severe renal fibrosis risk. Each centimeter increase in renal length decreased the odds of moderate-severe fibrosis by 38% (OR: 0.62; 95% CI 0.41-0.93; P = 0.020). After adjusting for confounders, the relationship persisted (OR: 0.58; 95% CI 0.33-1.00; P = 0.048). However, renal length presented limited discrimination ability in distinguishing degrees of renal fibrosis while controlling the key confounding factors, yielding an area under the ROC curve of only 0.58 (95% CI 0.45-0.70). CONCLUSION While an inverse relationship exists between renal length and risk of having moderate-severe renal fibrosis in non-diabetic CKD patients, renal length alone is insufficient for diagnosing fibrosis severity, underscoring the need for additional diagnostic parameters in CKD assessment.
Collapse
Affiliation(s)
- Ziman Chen
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, 999077, Kowloon, Hong Kong.
| | - Jun Jiang
- Department of Radiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Simon Takadiyi Gunda
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, 999077, Kowloon, Hong Kong
| | - Xinyang Han
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, 999077, Kowloon, Hong Kong
| | - Chaoqun Wu
- Department of Ultrasound, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Meihua East Rd, Zhuhai, 519000, China
| | - Michael Tin Cheung Ying
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, 999077, Kowloon, Hong Kong.
| | - Fei Chen
- Department of Ultrasound, The Fifth Affiliated Hospital of Sun Yat-Sen University, 52 Meihua East Rd, Zhuhai, 519000, China.
| |
Collapse
|
39
|
Li L, Xia G, Lei L, Hu Q, Wei X, Cui M, Tang Q, Yang D, Zhao A. Role of TGF-β1/Smad3 signalling pathway in renal tubulointerstitial fibrosis and renal damage in elderly rats with isolated systolic hypertension induced by increased pulse pressure. Acta Cardiol 2025; 80:135-147. [PMID: 39782012 DOI: 10.1080/00015385.2024.2445339] [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: 02/20/2024] [Revised: 09/30/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVE Elevated systolic blood pressure and increased pulse pressure are closely associated with renal damage; however, the exact mechanism remains unclear. Therefore, we investigated the effects of increased pulse pressure on tubulointerstitial fibrosis and renal damage in elderly rats with isolated systolic hypertension (ISH). Additionally, the role of renal tubular epithelial-mesenchymal transition (EMT) and its upstream signalling pathways were elucidated. METHODS Ten-month-old male rats were randomly divided into control and ISH groups, with seven rats in each group administered warfarin and vitamin K1 for 6 weeks. Blood pressure, renal function, mean blood flow in the common iliac artery, and diastolic vessel diameter were assessed, and the rat kidney medulla was collected for histological, genetic, and protein level analysis. RESULTS Increased pulse pressure, abnormal renal function, and increased shear stress were detected in rats with ISH. Histology assessments revealed fibrosis in the interstitium of ISH rats. Epithelial marker E-cadherin protein expression was decreased, while the protein expression of interstitial markers α-SMA and Vimentin was increased, and transforming growth factor (TGF)-β1/Smad3 signalling was upregulated in the kidney tissue of ISH rats. CONCLUSIONS Increased pulse pressure in elderly rats with ISH caused an increase in shear stress. These effects led to the development of EMT and the activation of its upstream TGF-β1/Smad3 signalling pathway, ultimately leading to renal tubular interstitial fibrosis causing renal injury.
Collapse
Affiliation(s)
- Lu Li
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guiling Xia
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Lei Lei
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Qiong Hu
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xueying Wei
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Mengbi Cui
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Qiaoling Tang
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Donghua Yang
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| | - Anju Zhao
- The Cadre Medical Department, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
40
|
Zhou Y, Luan F, Feng X, Yu M, Li L, Guo X, Yin X. TGF-β1 induces ROS to activate ferroptosis via the ERK1/2-WISP1 pathway to promote the progression of renal tubular epithelial cell fibrosis. Cytotechnology 2025; 77:61. [PMID: 39959788 PMCID: PMC11828780 DOI: 10.1007/s10616-025-00719-5] [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] [Received: 12/04/2024] [Accepted: 01/27/2025] [Indexed: 02/18/2025] Open
Abstract
Chronic kidney disease (CKD) often progresses to renal fibrosis, which is characterized by excessive extracellular matrix deposition and is also linked to ferroptosis. The present study investigated how TGF-β1 induces ferroptosis and thereby contributes to renal tubular epithelial cell fibrosis. Bioinformatics was employed to identify the differentially expressed genes relevant to renal fibrosis. An in vitro TGF-β1-induced fibrosis model of HK-2 cells was established, and the cell shape index was calculated. Fer-1, NAC, and PD98059 were utilized for targeted intervention, and their mechanisms were verified by transducing cells with WISP1-targeting shRNA lentivirus. Cell morphology was examined under a microscope, and cells were collected to determine the levels of ferroptosis-related factors (Fe2+, MDA, GSH, and LPO). Western blotting was performed to measure the levels of ERK1/2, WISP1, and ferroptosis indicators (GPX4 and hyperoxidized PRDX4). Flow cytometry was performed to determine the ROS levels and the rate of cell ferroptosis. TGF-β1 induced the transformation of HK-2 cells into fibroblast-like cells, leading to increased ROS levels, activation of the ERK1/2-WISP1 signaling pathway, and upregulation of ferroptosis and fibrosis-related factors. However, these effects could be effectively inhibited through pretreatment with Fer-1, NAC, and PD98059 individually, which further validated the involvement of the ERK1/2-WISP1 signaling pathway. In addition, WISP1 knockdown suppressed the cell transformation into fibroblast-like cells as well as the ferroptosis process, thereby reducing the expression levels of ferroptosis and fibrosis-related factors. The present study substantiated the process through which TGF-β1 elicits the production of ROS and triggers ferroptosis via the ERK1/2-WISP1 signaling pathway to facilitate the development of renal tubular epithelial cell fibrosis. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00719-5.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Nephrology, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan, 750004 Ningxia China
| | - Fengwu Luan
- Department of Nephrology, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan, 750004 Ningxia China
| | - Xiaonan Feng
- Department of Nephrology, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan, 750004 Ningxia China
| | - Min Yu
- Department of Nephrology, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan, 750004 Ningxia China
| | - Lu Li
- Department of Nephrology, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan, 750004 Ningxia China
| | - Xiaoyan Guo
- Department of Nephrology, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan, 750004 Ningxia China
| | - Xiaolong Yin
- Department of Nephrology, General Hospital of Ningxia Medical University, No. 804 South Shengli Street, Xingqing District, Yinchuan, 750004 Ningxia China
| |
Collapse
|
41
|
Huang Z, Liao Y, Zheng Y, Ye S, Zhang Q, Yu X, Liu X, Li N. Zinc Deficiency Causes Glomerulosclerosis and Renal Interstitial Fibrosis Through Oxidative Stress and Increased Lactate Metabolism in Rats. Biol Trace Elem Res 2025; 203:2084-2098. [PMID: 39028478 PMCID: PMC11919932 DOI: 10.1007/s12011-024-04306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024]
Abstract
Chronic kidney disease (CKD) is a highly prevalent condition characterized by renal fibrosis as its ultimate manifestation. Zinc deficiency is closely associated with CKD, evidenced by its link to renal fibrosis. Recently, local lactic acidosis has been demonstrated to promote renal fibrosis. Under zinc-deficient conditions, mitochondrial function is compromised and abnormal lactate metabolism might be induced potentially. However, it remains unclear whether zinc deficiency leads to renal fibrosis through local lactic acidosis. Zinc deficiency rat models were successfully established by feeding zinc-deficient diet. Western blot, qPCR, IHC, and other experiments were employed to investigate the key markers and molecular mechanisms of glomerulosclerosis and renal interstitial fibrosis. Our results indicate that zinc deficiency reduces specific markers of podocytes (podocalyxin, WT1, and nephrin) and activates the Wnt3a/β-catenin pathway, a key pathway in podocyte injury. Concurrently, glomerulosclerosis is indicated by increased urinary microalbumin and serum creatinine levels along with histological alteration observed through PAS and Masson staining in zinc-deficient rats. Furthermore, various degrees of upregulation for several markers of interstitial fibrosis including α-SMA, FN1 and collagen III are also revealed. These findings were further confirmed by Masson staining and IHC. Additionally, alterations in four markers in the EMT process, N-cadherin, E-cadherin, Vimentin, and snail, were consistent with expectations. We then confirmed the activation of the non-canonical TGF-β1 pathway known as the PI3K/AKT/mTOR pathway. An elevation in renal ROS levels accompanied by increased mitochondrial marker cytochrome C expression as well as an elevated NADH/NAD + ratio is also observed within the kidneys. Furthermore, the activity of both MMP/TIMP system and fibrinolytic system was abnormally enhanced under zinc deficiency conditions. Finally, we find zinc supplementation could significantly ameliorate relevant pathological alterations induced by zinc deficiency. These results collectively point that zinc deficiency causes podocyte damage ultimately resulting in glomerulosclerosis via accumulation of ROS and induces interstitial fibrosis via lactic acidosis.
Collapse
Affiliation(s)
- Zixuan Huang
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary University of London, London, E1 4NS, UK
| | - Yajie Liao
- Queen Mary University of London, London, E1 4NS, UK
| | - Yunxi Zheng
- Queen Mary University of London, London, E1 4NS, UK
| | - Shang Ye
- Queen Mary University of London, London, E1 4NS, UK
| | - Qianyu Zhang
- Queen Mary University of London, London, E1 4NS, UK
| | - Xiaohong Yu
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Xiaoxin Liu
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ningxu Li
- Department of Nephrology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
42
|
Xu LH, Tan RZ, Lin JY, Li T, Jia J, Wu LH, Wang R, He YH, Su HW, Li P, Wang L. Chaihuang Yishen Granule ameliorates mitochondrial homeostasis by upregulating PRDX5/TFAM axis to inhibit renal fibrosis in CKD. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156426. [PMID: 39955823 DOI: 10.1016/j.phymed.2025.156426] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/30/2024] [Accepted: 01/24/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Chaihuang Yishen Granules (CHYS) has been clinically proven to be effective for the treatment of chronic kidney disease (CKD), yet its underlying molecular mechanisms remain largely unexplored. OBJECTIVE To explore the innovative mechanisms by which CHYS alleviates CKD, focusing on its role in modulating PRDX5/TFAM-mediated mitochondrial homeostasis in renal cells. METHODS In this study, CKD mouse model was established by unilateral ureteral obstruction (UUO) and adenine (Ade) diet. Treatment interventions were administered by gavage with CHYS at doses of 3.8g/kg (low dose) and 7.6g/kg (high dose). The ameliorative effects of CHYS on CKD were evaluated by changes in renal function, kidney tissue structure, renal fibrosis, and mitochondrial dysfunction markers. Tert‑butyl hydroperoxide (t-BHP)-induced oxidative stress in TCMK1 cells was used to simulate CKD renal fibrosis induced by mitochondrial dysfunction in vitro. RESULTS CHYS significantly improves renal function and mitigates fibrosis while restoring mitochondrial homeostasis. Notably, PRDX5 expression, which is markedly reduced in CKD patients and mouse models, is substantially upregulated following CHYS treatment. Meanwhile, we demonstrate that ultrasound microbubble-mediated in situ overexpression of PRDX5 confers considerable renal protection in the UUO model. In vitro data show that CHYS effectively prevents t-BHP-induced mtDNA leakage in renal tubular cells, preserving mitochondrial function and stability, an effect compromised by PRDX5 knockdown. Moreover, our protein binding assays uncover a previously unreported interaction between PRDX5 and TFAM, with TFAM knockdown reversing the mitochondrial functional and fibrotic improvements achieved through PRDX5 overexpression and CHYS intervention. CONCLUSION These findings introduce a pioneering perspective on CHYS's mechanism of action. CHYS enhance TFAM activation through PRDX5 upregulation, counteract ROS-induced mitochondrial damage, and restoring mitochondrial homeostasis, and alleviates the progression of renal fibrosis in CKD, highlighting the innovative therapeutic potential of CHYS in mitochondrial-related renal pathologies.
Collapse
Affiliation(s)
- Ling-Hui Xu
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Rui-Zhi Tan
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jing-Yi Lin
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tong Li
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jian Jia
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Li-Hua Wu
- College of integrational Chinese and western medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Rui Wang
- College of integrational Chinese and western medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yu-Heng He
- College of integrational Chinese and western medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hong-Wei Su
- Department of Urology Surgery, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ping Li
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Li Wang
- College of integrational Chinese and western medicine, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
43
|
Liu Y, Shi S, Cheng T, Wang H, Wang H, Hu Y. The key role of miR‑378 in kidney diseases (Review). Mol Med Rep 2025; 31:101. [PMID: 39981929 PMCID: PMC11868772 DOI: 10.3892/mmr.2025.13466] [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: 08/09/2024] [Accepted: 01/27/2025] [Indexed: 02/22/2025] Open
Abstract
MicroRNAs (miRNAs/miRs) are endogenous, small non‑coding RNAs conserved across species that post‑transcriptionally regulate gene expression by both suppressing translation and inducing mRNA degradation. miRNAs are found in various tissues, exhibit variable expression and their dysregulation is implicated in numerous disease processes. Furthermore, miRNA expression levels have a key role in the normal development of kidney tissue and are key regulators of kidney function, modulating diverse biological processes across renal cell lineages. miR‑378 participates in pathological processes associated with kidney diseases, including kidney cancer, kidney transplantation and diabetic nephropathy. Despite its considerable effects on these conditions, a comprehensive summary of the roles of miR‑378 is unavailable. In the present review, the existing literature on miR‑378 in kidney diseases is consolidated, and its validated gene targets and biological effects in both malignant and non‑malignant conditions are highlighted, thereby providing a foundation for future research.
Collapse
Affiliation(s)
- Yangyang Liu
- Department of Cardiovascular Diseases, Sanming Integrated Medicine Hospital, Sanming, Fujian 365000, P.R. China
| | - Shuqing Shi
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Tao Cheng
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Haoshuo Wang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Huan Wang
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Yuanhui Hu
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| |
Collapse
|
44
|
Li X, She J, Cai M, Chen X, Qiu R, Luo X, Liu Y, Zhou X, Tang L. New Polyketides from a Marine Sponge-Derived Fungus, Neopestalotiopsis sp., with Anti-Renal Fibrosis Activity. Mar Drugs 2025; 23:148. [PMID: 40278269 PMCID: PMC12028388 DOI: 10.3390/md23040148] [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: 03/03/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
Sixteen polyketides, including six new compounds (1-2, and 5-8), were isolated from the culture of the marine sponge-associated fungus Neopestalotiopsis sp. SCSIO 41422. Their structures were elucidated through NMR, MS spectroscopic analyses, calculated electronic circular dichroism, quantum chemical NMR calculations, and X-ray single-crystal diffraction. To screen and evaluate the inhibitory activity of these polyketides in renal fibrosis, a TGF-β1-stimulated HK-2 cell model was used. All tested compounds (1, 5-8, and 11-12) at 10 µM showed obvious anti-fibrotic activity by inhibiting TGF-β1-induced α-SMA expression and extracellular matrix production (collagen I and fibronectin). Among them, gamahorin A (1) was shown to be the most potent and the most promising inhibitor against renal fibrosis.
Collapse
Affiliation(s)
- Xinlong Li
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (X.L.); (X.L.); (Y.L.)
| | - Jianglian She
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (J.S.); (X.C.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.C.); (R.Q.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meiqun Cai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.C.); (R.Q.)
| | - Xinqi Chen
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (J.S.); (X.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rongxiang Qiu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.C.); (R.Q.)
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaowei Luo
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (X.L.); (X.L.); (Y.L.)
| | - Yonghong Liu
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (X.L.); (X.L.); (Y.L.)
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (J.S.); (X.C.)
| | - Xuefeng Zhou
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (X.L.); (X.L.); (Y.L.)
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; (J.S.); (X.C.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lan Tang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.C.); (R.Q.)
| |
Collapse
|
45
|
Lu Q, Liu J, Xiong Y, Jian J, Wang J, Chen Z, Wan S, Liu X, Wang L. Cyanidin-3-glucoside upregulated NDRG2 through the PI3K/AKT pathway to alleviate EMT and ECM in renal fibrosis. Sci Rep 2025; 15:10695. [PMID: 40155416 PMCID: PMC11953473 DOI: 10.1038/s41598-025-94918-x] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Renal fibrosis is a critical progression of chronic kidney disease, and epithelial-to-mesenchymal transition (EMT) and extracellular matrix(ECM) deposition are crucial pathologic change of renal fibrosis, which still lacks of effective treatment. In this study, it was found that cyanidin-3-O-glucoside (C3G) could inhibit EMT and ECM activated by unilateral ureteral obstruction (UUO) and transforming growth factor-β1 (TGF-β1) stimulation. Moreover, N-Myc downstream-regulated gene 2(NDRG2), which involved in the progression of renal fibrosis, was down-regulated in vivo and in vitro model. However, C3G pretreatment could reverse the reductive expression of NDRG2. Furthermore, we found that the combined treatment of C3G and si-NDRG2 could reverse the decreased EMT and ECM, which induced by C3G treatment only. And the activation of Phosphatidylinositol 3-kinase (PI3K)/ Protein Kinase B (AKT) pathway significantly enhanced EMT and ECM, which was decreased by C3G treatment only in TGF-β1 induced Human Kidney 2 (HK-2) cells. In conclusion, our results demonstrated that C3G alleviated EMT and ECM by elevating NDRG2 expression through the PI3K/AKT pathway, indicating that C3G could be a potential treatment against renal fibrosis.
Collapse
Affiliation(s)
- Qianxue Lu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jin Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yufeng Xiong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jun Jian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jingsong Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Shanshan Wan
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
46
|
Ma K, Fujino M, Yang Y, Ding Z, Hu X, Ito H, Takahashi K, Nakajima M, Isaka Y, Li XK. 5-aminolaevulinic acid with sodium ferrous citrate alleviated kidney injury and fibrosis in a unilateral ureteral obstruction model. Int Immunopharmacol 2025; 150:114321. [PMID: 39970714 DOI: 10.1016/j.intimp.2025.114321] [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: 09/29/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
PURPOSE This study aimed to investigate the potential therapeutic effects of 5-aminolaevulinic acid (5-ALA) combined with sodium ferrous citrate (SFC) on kidney injury and fibrosis in a mouse model of unilateral ureteral obstruction (UUO)-induced chronic kidney disease (CKD). METHODS A murine UUO model was used to mimic human CKD. The mice received daily intragastric administration of 5-ALA/SFC for 7 and 14 consecutive days. Serum creatinine (Cr) and blood urea nitrogen (BUN) levels and histological evaluations were performed to assess the renal function parameters underlying 5-ALA/SFC treatment in the UUO model. Differentially expressed genes (DEGs) were analyzed by RNA sequencing (RNA-Seq), and the results were validated by quantitative real-time PCR (qRT-PCR). The severity of renal fibrosis was evaluated using Sirius red and Masson's trichrome (MT) staining techniques, while the expression of fibrosis-related genes was examined using western blotting and immunohistochemistry. RESULTS Our findings demonstrated that 5-ALA/SFC treatment improved UUO-induced renal dysfunction, attenuated tubular damage, and significantly reduced serum Cr and BUN levels as well as the mRNA expression and secretion of pro-inflammatory and programmed cell death-related cytokines in kidney tissues. Furthermore, 5-ALA/SFC suppressed renal tissue fibrosis and downregulated the mRNA and protein expression of fibrosis-related genes. Notably, treatment with 5-ALA/SFC led to the significant upregulation of protein expression levels of PPAR gamma-coactivator-1α (PGC-1α), indicating its role in inhibiting inflammation and fibrosis through the activation of the PGC-1α signaling pathway. CONCLUSION 5-ALA/SFC exhibits renoprotective effects in UUO-induced CKD by attenuating inflammation, cell death, and suppressing renal fibrosis. These findings suggest a specific renal protective mechanism for 5-ALA/SFC, highlighting its potential as a novel therapeutic agent for human CKD treatment.
Collapse
Affiliation(s)
- Kuai Ma
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Nephrology, Osaka University Graduate School of Medicine, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Research Center for Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan.
| | - Yang Yang
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Zhaolun Ding
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Xin Hu
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | | | | | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Japan.
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
47
|
Tiscornia C, Tapia V, Águila D, Lorca-Ponce E, Aicardi V, Vásquez F. Maqui and Chronic Kidney Disease: A Narrative Review on the Potential Nephroprotective Role of Anthocyanins. Nutrients 2025; 17:1058. [PMID: 40292440 PMCID: PMC11944665 DOI: 10.3390/nu17061058] [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: 01/29/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Chronic kidney disease (CKD) is a progressive pathology, with high global prevalence, associated with inflammation and oxidative stress. Given the limited capacity of conventional treatments to reverse renal damage, complementary alternatives have emerged such as supplementation with anthocyanins from maqui (Aristotelia chilensis), known for their antioxidant and anti-inflammatory properties. This review analyzes the evidence for their impact on CKD progression. Methods: A narrative review of the experimental literature regarding maqui anthocyanins, their bioavailability, and their effects on oxidative stress, inflammation, and CKD to January 2025 was conducted. Articles without peer review or without a focus on Aristotelia chilensis were excluded, guaranteeing an updated compilation on its nephroprotective potential. Results: Anthocyanins have shown benefits in reducing oxidative stress, inflammation, and glycemia regulation. Preclinical studies suggest improvements in renal function as well as less fibrosis. Human trials indicate positive effects on metabolism, although evidence in CKD patients is limited. Bioavailability remains a challenge to optimizing efficacy. Conclusions: Maqui is a promising source of anthocyanins, with nephroprotective potential. However, robust clinical studies are required to determine its safety, optimal dose, and long-term impact in CKD. Its incorporation into evidence-based therapeutic strategies could offer an innovative approach in the management of this disease. More clinical studies are needed to validate the preclinical findings and optimize the therapeutic use of maqui in CKD.
Collapse
Affiliation(s)
- Caterina Tiscornia
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| | - Violeta Tapia
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| | - Daniela Águila
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| | - Enrique Lorca-Ponce
- Escuela de Enfermería, Universidad Finis Terrae, Santiago 7501014, Chile;
- Escuela de Kinesiología, Facultad de Arte y Educación Física, Universidad Metropolitana en Ciencias de la Santiago, Santiago 7760197, Chile
| | - Valeria Aicardi
- Unidad de Diálisis, Clínica Indisa, Santiago 7501014, Chile;
| | - Fabián Vásquez
- Escuela de Nutrición y Dietética, Universidad Finis Terrae, Santiago 7501014, Chile; (C.T.); (V.T.); (D.Á.)
| |
Collapse
|
48
|
Han WW, Miao MY, Lyu JQ, Tao HW, Jia YP, Liu YJ, Wang JM, Chen JS, Qin LQ, Chen GC. Female Reproductive Factors, Exogenous Hormone use, and Incident Chronic Kidney Disease and end-stage Renal Disease. J Clin Endocrinol Metab 2025; 110:e970-e979. [PMID: 38829052 DOI: 10.1210/clinem/dgae374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/06/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
CONTEXT Younger women have a slower progressive loss of kidney function than age-matched men and the sex advantage diminishes after menopause, suggesting a role for female hormones in the development of kidney diseases. OBJECTIVE To examine the relationships of numerous reproductive factors and exogenous hormone use with long-term risk of chronic kidney disease (CKD) and end-stage renal disease (ESRD) in women. METHODS A total of 260 108 women without prevalent CKD and ESRD were included. The relationships of various reproductive factors and exogenous hormone use with incident CKD and ESRD were assessed, with multivariable adjustment for potential confounders. RESULTS During a median of ∼12.5 years of follow-up, 8766 CKD and 554 ESRD cases were identified. Younger age at first live birth, hysterectomy or bilateral oophorectomy before age 50 years, menopausal before age 45 years, and menopausal hormone therapy initiated before age 50 years was associated with a higher risk of CKD. The relationships of these factors with ESRD were generally consistent with those for CKD. Each 5-year increment in menopausal age was associated with an 11% lower risk of CKD (hazard ratio [HR] = 0.89; 95% CI, 0.87-0.91) and a 13% lower risk of ESRD (HR = 0.87; 95% CI, 0.79-0.95). Each 5-year delay in starting menopausal hormone therapy was associated with a 13% lower risk of CKD (HR = 0.87; 95% CI, 0.84-0.90) and a 15% lower risk of ESRD (HR = 0.85; 95% CI, 0.73-0.99). CONCLUSIONS Several reproductive characteristics reflecting shorter cumulative exposure to endogenous estrogen or premature exposure to exogenous hormones are associated with a greater risk of CKD and ESRD in women, supporting a potential role of female hormones in renal pathophysiology.
Collapse
Affiliation(s)
- Wen-Wen Han
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Meng-Yuan Miao
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jie-Qiong Lyu
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Hao-Wei Tao
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yi-Ping Jia
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yu-Jie Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jia-Min Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jing-Si Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Guo-Chong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, China
| |
Collapse
|
49
|
Noda P, Francini ALR, Teles F, Júnior SJ, Fonseca FLA, Borges FT, Sobrinho AC, Taniwaki N, Noronha IL, Fanelli C. Extracellular Vesicles (EVs) Derived from Mesenchymal Stem Cells (MSCs) as Adjuvants in the Treatment of Chronic Kidney Disease (CKD). Cells 2025; 14:434. [PMID: 40136683 PMCID: PMC11941753 DOI: 10.3390/cells14060434] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/18/2025] [Accepted: 01/24/2025] [Indexed: 03/27/2025] Open
Abstract
Chronic kidney disease (CKD) is considered an important health issue worldwide. The renin-angiotensin-aldosterone system (RAAS) blockade through the administration of angiotensin II receptor blockers, such as Losartan (LOS), has been considered the best strategy for CKD treatment for decades. However, this approach promotes only partial detention of CKD progression and cannot reverse renal damage. The aim of the present study was to investigate whether the therapeutic administration of extracellular vesicles (EVs) derived from adipose stem cells (ASCs), associated to LOS treatment, would promote additional renoprotection in rats underwent the 5/6 renal ablation CKD model. ASC-derived EV were administered locally, in the renal subcapsular area, 15 days after CKD induction, when LOS therapy also began. Animals were followed for additional 15 days and our results demonstrated that subcapsular injection of ASC-derived EV associated with LOS significantly reduced glomerulosclerosis, renal interstitial infiltration by myofibroblasts, and macrophages in the 5/6 CKD model. Additionally, LOS + EV abrogated systemic hypertension, proteinuria, and albuminuria, and stimulated local gene overexpression of the endogenous anti-inflammatory Il-4. Although more studies are still required to establish the best EV dose and administration route, these findings point to therapy with ASC-derived EV as a potential adjuvant in CKD treatment.
Collapse
Affiliation(s)
- Paloma Noda
- Laboratory of Cellular, Genetic and Molecular Nephrology, Renal Division, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, 4° Andar, 4304, São Paulo 01246-903, SP, Brazil (I.L.N.)
| | - Ana L. R. Francini
- Laboratory of Cellular, Genetic and Molecular Nephrology, Renal Division, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, 4° Andar, 4304, São Paulo 01246-903, SP, Brazil (I.L.N.)
| | - Flavio Teles
- Renal Division, Faculty of Medicine, Federal University of Alagoas, Maceio 57200-000, AL, Brazil
| | - Samuel J. Júnior
- Laboratory of Cellular, Genetic and Molecular Nephrology, Renal Division, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, 4° Andar, 4304, São Paulo 01246-903, SP, Brazil (I.L.N.)
| | - Fernando L. A. Fonseca
- Department of Clinical Laboratory, University Center of ABC Medical School, Santo Andre 09060-650, SP, Brazil
| | - Fernanda T. Borges
- Departament of Medicine, Nephrology Division, Paulista School of Medicine, Federal University of São Paulo, São Paulo 04023-062, SP, Brazil;
| | - Adão C. Sobrinho
- Laboratory of Cellular Biology, Department of Pathology, School of Medicine, University of São Paulo, São Paulo 01246-903, SP, Brazil
| | - Noemi Taniwaki
- Laboratory of Electron Microscopy, Instituto Adolfo Lutz, São Paulo 01246-000, SP, Brazil
| | - Irene L. Noronha
- Laboratory of Cellular, Genetic and Molecular Nephrology, Renal Division, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, 4° Andar, 4304, São Paulo 01246-903, SP, Brazil (I.L.N.)
| | - Camilla Fanelli
- Laboratory of Cellular, Genetic and Molecular Nephrology, Renal Division, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, 4° Andar, 4304, São Paulo 01246-903, SP, Brazil (I.L.N.)
| |
Collapse
|
50
|
Gouveia PQ, Fanelli C, Ornellas FM, Garnica MR, Francini ALR, Murata GM, Matheus LHG, Morales MM, Noronha IL. Adipose Tissue Stem Cells (ASCs) and ASC-Derived Extracellular Vesicles Prevent the Development of Experimental Peritoneal Fibrosis. Cells 2025; 14:436. [PMID: 40136685 PMCID: PMC11941392 DOI: 10.3390/cells14060436] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 03/27/2025] Open
Abstract
Cell therapy utilizing mesenchymal stromal cells (MSCs) through paracrine mechanisms holds promise for regenerative purposes. Peritoneal fibrosis (PF) is a significant complication of peritoneal dialysis. Various strategies have been proposed to protect the peritoneal membrane (PM). This study explores the effectiveness of adipose-tissue-derived stem cells (ASCs) and extracellular vesicles (EVs) at mitigating PF using a rat model of PF induced by chlorhexidine gluconate. ASC and EV treatments effectively prevented an increase in the thickness of the PM and diminished the number of myofibroblasts, fibronectin expression, collagen III expression, and PF-related factors such as TGF-β and FSP-1. Smad3 gene expression decreased in the treatment groups, whereas Smad7 gene expression increased in treated animals. In addition, ASC and EV injections showed potent anti-inflammatory effects. Glucose transport through the PM remained unaffected in relation to the PF group; both treatments promoted an increase in ultrafiltration (UF) capacity. The PF+EVs treated group showed the highest increase in UF capacity. Another critical aspect of ASC and EV treatments was their impact on neoangiogenesis in the PM which is vital for UF capacity. Although the treated groups displayed a significant decrease in VEGF expression in the PM, peritoneal function remained effective. In conclusion, within the experimental PF model, both ASC and EV treatments demonstrated anti-inflammatory effects and comparably hindered the progression of PF. The EV treatment exhibited superior preservation of peritoneal function, along with enhanced UF capacity. These findings suggest the potential of ASCs and EVs as novel therapeutic approaches to prevent the development of PF associated with peritoneal dialysis.
Collapse
Affiliation(s)
- Priscila Q. Gouveia
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Camilla Fanelli
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Felipe M. Ornellas
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Margoth R. Garnica
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Ana L. R. Francini
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Gilson M. Murata
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| | - Luiz H. G. Matheus
- Laboratory of Carbohydrate and Radioimmunoassay, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil;
| | - Marcelo M. Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-853, Brazil;
| | - Irene L. Noronha
- Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, Medical School, University of São Paulo, São Paulo 01246-903, Brazil; (P.Q.G.); (C.F.); (F.M.O.); (M.R.G.); (G.M.M.)
| |
Collapse
|