1
|
Ji YW, Wen XY, Tang HP, Su WT, Xia ZY, Lei SQ. Necroptosis: a significant and promising target for intervention of cardiovascular disease. Biochem Pharmacol 2025; 237:116951. [PMID: 40268251 DOI: 10.1016/j.bcp.2025.116951] [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: 12/20/2024] [Revised: 03/18/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Due to changes in dietary structures, population aging, and the exacerbation of metabolic risk factors, the incidence of cardiovascular disease continues to rise annually, posing a significant health burden worldwide. Cell death plays a crucial role in the onset and progression of cardiovascular diseases. As a regulated endpoint encountered by cells under adverse stress conditions, the execution of necroptosis is regulated by classicalpathways, the calmodulin-dependent protein kinases (CaMK) pathway, and mitochondria-dependent pathways, and implicated in various cardiovascular diseases, including atherosclerosis, myocardial infarction, myocardial ischemia-reperfusion injury (IRI), heart failure, diabetic cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, chemotherapy drug-induced cardiomyopathy, and abdominal aortic aneurysm (AAA). To further investigate potential therapeutic targets for cardiovascular diseases, we also analyzed the main molecules and their inhibitors involved in necroptosis in an effort to uncover insights for treatment.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Tang K, Ye T, He Y, Ba X, Xia D, Peng E, Chen Z, Ye Z, Yang X. Ferroptosis, necroptosis, and pyroptosis in calcium oxalate crystal-induced kidney injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167791. [PMID: 40086520 DOI: 10.1016/j.bbadis.2025.167791] [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: 11/30/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Kidney stones represent a highly prevalent urological disorder worldwide, with high incidence and recurrence rates. Calcium oxalate (CaOx) crystal-induced kidney injury serves as the foundational mechanism for the formation and progression of CaOx stones. Regulated cell death (RCD) such as ferroptosis, necroptosis, and pyroptosis are essential in the pathophysiological process of kidney injury. Ferroptosis, a newly discovered RCD, is characterized by its reliance on iron-mediated lipid peroxidation. Necroptosis, a widely studied programmed necrosis, initiates with a necrotic phenotype that resembles apoptosis in appearance. Pyroptosis, a type of RCD that involves the gasdermin protein, is accompanied by inflammation and immune response. In recent years, increasing amounts of evidence has demonstrated that ferroptosis, necroptosis, and pyroptosis are significant pathophysiological processes involved in CaOx crystal-induced kidney injury. Herein, we summed up the roles of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury. Furthermore, we delved into the curative potential of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury.
Collapse
Affiliation(s)
- Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
3
|
Toba H, Jin D, Takai S. Suppressing SPARC gene with siRNA exerts therapeutic effects and inhibits MMP-2/9 and ADAMTS1 overexpression in a murine model of ischemia/reperfusion-induced acute kidney injury. J Pharmacol Sci 2025; 158:103-112. [PMID: 40288820 DOI: 10.1016/j.jphs.2025.03.010] [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: 01/14/2025] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC), a collagen-binding matricellular protein, is reported to facilitate inflammation and fibrosis in various tissues including the kidneys. Ischemia/reperfusion (I/R) is a major process of acute kidney injury. To investigate whether SPARC inhibition might attenuate renal I/R injury, we injected small interfering RNA (siRNA) targeting SPARC into male BALB/c mice one day before 45 min of renal ischemia followed by 72 h of reperfusion. Serum creatinine concentration, blood urea nitrogen, histological tubular damage, tubulointerstitial fibrosis, and expression of collagen I and transforming growth factor-β were increased after I/R. Expression of 4-hydroxy-2-nonenal, an oxidative stress marker, and the inflammatory cytokines monocyte chemoattractant protein-1 and tumor necrosis factor-α, were also upregulated in I/R kidneys. Overexpression of SPARC mRNA was observed after I/R, and immunohistochemistry revealed that SPARC was localized mainly in damaged tubuloepithelial cells. Additionally, a disintegrin and metalloproteinase with thrombospondin type 1 motif (ADAMTS1) expression colocalized with SPARC. Injection of siRNA targeting SPARC attenuated renal dysfunction, histological abnormalities, collagen deposition, oxidative stress, and renal inflammation. In addition, SPARC gene knockdown suppressed the I/R-induced increases in ADAMTS1 and matrix metalloproteinase-2/9 expression. In conclusion, I/R-induced SPARC could be a novel therapeutic target against acute kidney injury.
Collapse
Affiliation(s)
- Hiroe Toba
- Laboratory of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, 1 Misasagi Shichono-cho, Yamashina-ku, Kyoto, 607-8412, Japan; Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan.
| | - Denan Jin
- Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan
| | - Shinji Takai
- Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan
| |
Collapse
|
4
|
Li S, Duan F, Qiu Z, Nan Z, Cao X, Yang C, Li W, Shen B. Polymer-Conjugated SOD-Pt⁰ Micelles Enhance ROS Cascade Scavenging to Alleviate Ischemia-Reperfusion Injury During Kidney Transplantation. Adv Healthc Mater 2025:e2500696. [PMID: 40394947 DOI: 10.1002/adhm.202500696] [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: 02/07/2025] [Revised: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Ischemia-reperfusion injury (IRI) during kidney transplantation is linked to oxidative stress induced by excessive reactive oxygen species (ROS), which causes the injury of transplanted kidney, leading to further intensified organ shortages. Protein-based antioxidants have been developed for ROS scavenging via cascade biocatalyst. The in situ growth of metal nanozymes on proteins effectively decreases the steric hindrance between active sites, improving the efficiency of cascade biocatalysts. However, the poor stability of protein during the process of preparation and intracellular delivery leads to low therapeutic effects. In this study, three different functional polymers are conjugated to SOD for the formation of micelles. Surprisingly, it is found that the conjugated ultra-acid sensitive polymer efficiently preserves the enzymatic activity of SOD, due to great endo/lysosomal escape capacity. Subsequently, SOD micelles (SOE) are used as a template to prepare SOE-Pt0 (SOEP) through in situ growth of Pt0 with vicinal enzymatic active sites. The preparation process minimally impacts on the activity of SOD, owing to improved stability. The system exhibits effective cascade ROS scavenging, significantly reducing kidney damage and inflammation caused by IRI. The research offers a novel approach for addressing IRI challenges in organ transplantation and provides a promising strategy to mitigate organ shortages.
Collapse
Affiliation(s)
- Shengzhou Li
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Fei Duan
- Department of Nanomedicine, Translational Medicine Research Center, & Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Naval Medical University, Shanghai, 200433, China
| | - Zhiwen Qiu
- Department of Nanomedicine, Translational Medicine Research Center, & Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Naval Medical University, Shanghai, 200433, China
| | - Zhuofan Nan
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xiangqian Cao
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Chenkai Yang
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Wei Li
- Department of Nanomedicine, Translational Medicine Research Center, & Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Naval Medical University, Shanghai, 200433, China
| | - Bing Shen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
- Department of Urology, Shanghai Tenth People's Hospital of Tongji University, Shanghai, 200072, China
| |
Collapse
|
5
|
Qian Z, Zhang X, Huang J, Hou Y, Hu C, Cao Y, Wu N, Zhu T, Wu G. Glucose deprivation-restoration induces labile iron overload and ferroptosis in renal tubules through V-ATPase-mTOR axis-mediated ferritinophagy and iron release by TPC2. Free Radic Biol Med 2025; 236:204-219. [PMID: 40379157 DOI: 10.1016/j.freeradbiomed.2025.05.390] [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: 03/18/2025] [Revised: 04/28/2025] [Accepted: 05/10/2025] [Indexed: 05/19/2025]
Abstract
Renal ischemia-reperfusion injury (IRI), a common complication following kidney transplantation and partial nephrectomy, is the leading cause of renal dysfunction with limited treatment. Excessive cellular iron accumulation drives lipid peroxidation and activates pathways associated with ferroptosis, which has been implicated in renal IRI. However, the regulatory mechanisms of cellular iron metabolism and its relationship with ferroptosis during ischemia-reperfusion (IR) remain unclear. In this study, in vitro OGSD-R (oxygen, glucose, and serum deprivation-restoration) models and in vivo IR models were employed to investigate alterations in iron metabolism, ferroptosis, and the underlying molecular mechanisms using immunofluorescence, immunoblotting and biochemical testing. We identified glucose deprivation-restoration (GD-R) as a key trigger of cellular iron overload under renal IR condition. Mechanistically, GD-R-induced iron overload is driven by the dysfunction of vacuolar ATPase (V-ATPase)-mammalian target of rapamycin (mTOR) pathway. Inactivation of mTOR results in lysosomal iron releases via two-pore channel 2 (TPC2) and ferritin degradation through ferritinophagy. This process elevates intracellular iron levels, thereby promoting ferroptosis in renal IRI. Targeting cellular iron metabolism effectively alleviates renal IRI. These findings highlight the critical role of glucose metabolism and V-ATPase-mTOR pathway in the regulation of iron homeostasis and ferroptosis during renal IRI, and establish a mechanistic link among glucose metabolism, iron overload and ferroptosis, providing potential therapeutic targets for renal IRI.
Collapse
Affiliation(s)
- Zhiyu Qian
- Department of Kidney Transplantation, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai, 200030, China; Shanghai Key Laboratory of Organ Transplantation, 170 Fenglin Road, Shanghai, 200030, China; Department of Urology, Huadong Hospital Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, 1278 Baode Road, Shanghai, 200443, China
| | - Jiahua Huang
- Department of Neurology, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai, 200030, China
| | - Yumin Hou
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China
| | - Chunlan Hu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China
| | - Yirui Cao
- Department of Kidney Transplantation, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai, 200030, China; Shanghai Key Laboratory of Organ Transplantation, 170 Fenglin Road, Shanghai, 200030, China
| | - Nannan Wu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China
| | - Tongyu Zhu
- Department of Kidney Transplantation, Zhongshan Hospital Fudan University, 170 Fenglin Road, Shanghai, 200030, China; Shanghai Key Laboratory of Organ Transplantation, 170 Fenglin Road, Shanghai, 200030, China.
| | - Guoyi Wu
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Shanghai, 201508, China.
| |
Collapse
|
6
|
Liang BE, Long LS, Wu XY, Huang MY, Lai Y, Yuan X, Wang MH, Li M, Zheng QQ, Zhang HL, Chen MC, Liu ZD, Geng X, Lyu QQ, Wang WD, Liu QH, Liu WZ, Li CL. Alginate oligosaccharide prevents renal ischemia-reperfusion injury in rats via MRC1-mediated pathway. Acta Pharmacol Sin 2025:10.1038/s41401-025-01545-3. [PMID: 40263568 DOI: 10.1038/s41401-025-01545-3] [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: 12/24/2024] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 04/24/2025]
Abstract
Acute kidney injury (AKI) is a clinical syndrome that is defined as a sudden decline in renal function and characterized by inflammation and tubular injury. Alginate oligosaccharide (AOSC), a natural product obtained from alginate by acidolysis and hydrolysis, shows activities of antioxidant, immunomodulation, and anti-inflammation. In this study, we investigated the potential of AOSC in the treatment of AKI. Renal ischemia-reperfusion (I/R) was induced in male rats by clipping both the renal artery and vein for 45 min followed by reperfusion for 24 h. The rats were treated with AOSC (100 mg/kg, i.g.) before surgery. At the end of the experiments, both kidneys were collected for protein, mRNA measurement, or histological analysis. We showed that AOSC pretreatment significantly improved glomerular and tubular function in the kidney of I/R rats. AOSC markedly inhibited I/R-induced activation of TLR4/MyD88/NF-κB/IL-1β inflammatory signaling and prevented apoptosis in the kidney. In HK2 cells subjected to hypoxia/reoxygenation (H/R) stimulation, AOSC (250-1000 μg/ml) dose-dependently prevented pro-inflammatory responses and cell apoptosis. Transcriptomic analysis revealed that I/R increased the expression levels of mannose receptor type C1 (MRC1) in the kidney, which was markedly inhibited by AOSC. Molecular docking showed that AOSC interacted with E725, N727, E733, T743, S745, and N747 of MRC1 through hydrogen bonds. MRC1 gene knockout significantly improved renal function and attenuated I/R-induced kidney inflammation and apoptosis in mice. In line with this, AOSC failed to prevent I/R-induced kidney injury in MRC1 gene knockout mice. UPLC analysis showed that the protection of AOSC in HK2 cells subjected to H/R was likely attributed to MRC1-mediated intracellular endocytosis. In conclusion, AOSC prevents I/R-induced AKI, which is at least partially mediated by MRC1.
Collapse
Affiliation(s)
- Bai-En Liang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Luo-Sha Long
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin-Yan Wu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mei-Ying Huang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying Lai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xi Yuan
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ming-Hui Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi-Qi Zheng
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hai-Ling Zhang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Man-Chun Chen
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhen-de Liu
- Haitang (Jiangsu) Biotechnology Co Ltd, Nantong, 226100, China
| | - Xin Geng
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Qian-Qian Lyu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Wei-Dong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Hua Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China.
- Department of Nephrology, Jieyang People's Hospital, Jieyang, 522000, China.
| | - Wei-Zhi Liu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Chun-Ling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Wu D, Ma W, Wang L, Long C, Chen S, Liu J, Qian Y, Zhao J, Zhou C, Jia R. Physically engineered extracellular vesicles targeted delivering miR-21-5p to promote renoprotection after renal ischemia-reperfusion injury. Mater Today Bio 2025; 31:101528. [PMID: 39980630 PMCID: PMC11840549 DOI: 10.1016/j.mtbio.2025.101528] [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: 09/23/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/22/2025] Open
Abstract
Acute kidney injury (AKI) resulting from ischemia-reperfusion injury (IRI) is a common challenge in various clinical practices, yet effective therapies remain elusive. Endothelial injury plays a crucial role in the pathogenesis of renal IRI. Endothelial progenitor cells (EPCs) derived extracellular vesicles (EVs) hold promise as cell-free therapies for treating renal IRI; however, their efficacy is limited by low delivery efficiency. In this study, we developed neutrophils (NEs) membrane-modified EVs (N-EVs) by exploiting the natural properties of NEs to target damaged endothelium. N-EVs inherited the characteristic membrane proteins of NEs along with the biological functions of EPCs-EVs. Results from in vitro and in vivo experiments demonstrated that N-EVs significantly enhanced the targeting efficiency of EVs towards IRI kidneys via P-selectin glycoprotein ligand-1 (PSGL-1). Moreover, N-EVs effectively promoted the proliferation, migration, and tube-formation abilities of injured endothelial cells (ECs) and contributed to overall renal function improvement in IRI kidneys through targeted delivery of miR-21-5p. Additionally, N-EVs could restore damaged endothelial integrity, reduce cytokine release, and inhibit leukocyte infiltration, hence alleviating renal inflammation. In conclusion, our accessible engineering approach represents a promising strategy for treating renal IRI. Furthermore, this membrane hybrid modification can be tailored and optimized for broader applications in treating other diseases.
Collapse
Affiliation(s)
- Di Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wenjie Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Liucheng Wang
- Department of Urology, Lianshui People's Hospital, Kangda College Affiliated to Nanjing Medical University, Jiang Su, 223400, China
| | - Chengcheng Long
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Silin Chen
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yiguan Qian
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Jun Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| |
Collapse
|
8
|
Chen Q, Guo J, Han S, Wang T, Xia K, Yu B, Lu Y, Qiu T, Zhou J. Cordycepin alleviates renal ischemia-reperfusion injury by suppressing the p38/JNK signaling pathway. Int Immunopharmacol 2025; 150:114264. [PMID: 39954658 DOI: 10.1016/j.intimp.2025.114264] [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: 05/18/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Renal ischemia-reperfusion injury (IRI) makes a significant contribution to delayed graft function (DGF) and reduced allograft survival time post-transplantation, thereby complicating the prognosis of renal transplant recipients. Cordycepin, an active compound purified from the traditional Chinese medicine Cordyceps sinensis, has exhibited remarkable anti-inflammatory and organ-protective effects against various diseases, including neurological, hepatic, and metabolic disorders. Therefore, the present study used a murine model of renal ischemia/reperfusion (I/R) and HK2 cell line hypoxia/reoxygenation (H/R) to determine whether cordycepin influences renal IRI. The findings indicated that cordycepin significantly mitigated renal IRI by inhibiting the p38/JNK signaling pathway in the renal tubular epithelial cells, thereby suppressing inflammation, cell apoptosis, and ferroptosis. These findings offer a novel avenue for improving the prognosis of renal transplant recipients and allograft survival.
Collapse
Affiliation(s)
- Qi Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China; National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| | - Shangting Han
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Kang Xia
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Bo Yu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Yifan Lu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China.
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
9
|
Zhuang S, Li F, Wang L, Lai Z, Li D, Wu H, Wu J, Qu J, Zhang X, Zhang M, Chen R, Yuan X. Neutrophil extracellular trap-derived double-stranded RNA aggravates PANoptosis in renal ischemia reperfusion injury. Cell Commun Signal 2025; 23:140. [PMID: 40098148 PMCID: PMC11912734 DOI: 10.1186/s12964-025-02145-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/08/2025] [Indexed: 03/19/2025] Open
Abstract
A dysregulated inflammatory response and inflammation-associated cell death are central features of renal ischemia-reperfusion injury (IRI). PANoptosis, is a recently recognized form of inflammatory programmed cell death characterized by key features of pyroptosis, apoptosis and necroptosis; however, the specific involvement of PANoptosis in renal IRI remains unknown. By using neutrophil extracellular trap (NETs)-depleted Pad4-/- mice, we found that NETs are essential for exacerbating tissue injury in renal IRI. Single-cell RNA sequencing (scRNA-seq) revealed that IRI promoted PANoptosis signalling in proximal tubular epithelial cells (PTs), whereas PAD4 knockout inhibited PANoptosis signalling. PTs expressed mainly RIPK1-PANoptosomes, which executed NET-induced PANoptosis in PTs in renal IRI model mice. Mechanistically, NET-derived double-stranded RNA (dsRNA) promoted PANoptosis in PTs, and PT-expressed TLR3 was responsible for the sensing the extracellular dsRNA. Treating mice with chemical inhibitors of the dsRNA/TLR3 complex suppressed PANoptosis and alleviated tissue injury in renal IRI. Together, the results of this study reveal a mechanism by which the NET-dsRNA-TLR3 axis aggravates PT cell PANoptosis in renal IRI.
Collapse
Affiliation(s)
- Shaoyong Zhuang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Fangzhou Li
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
- Department of Urology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Liya Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Zilong Lai
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Dawei Li
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Haoyu Wu
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Jiajin Wu
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Junwen Qu
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Xianyun Zhang
- Department of Urology, The Affiliated Huaian Hospital of Xuzhou Medical University, Jiangsu, 223200, China.
| | - Ming Zhang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China.
| | - Ruoyang Chen
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China.
| | - Xiaodong Yuan
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
10
|
Calva Lopez A, Robles Garcia JE, Yanez Ruiz CA, Tapia Tapia MD, Talavera Cobo V, Muñoz Bastidas CA, Miñana Lopez B, Sanchez Zalabardo D. Does Oxygen Work? Evidence for Oxygenation During Kidney Graft Preservation: A Review. J Clin Med 2025; 14:1927. [PMID: 40142735 PMCID: PMC11942664 DOI: 10.3390/jcm14061927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/23/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Kidney transplantation (KT) is the gold-standard treatment of end-stage kidney disease (ESKD). Traditional preservation methods, such as static cold storage (SCS), have been replaced by modern and more effective preservation methods, especially hypothermic machine perfusion (HMP). Regardless of improved preservation, ischemia-reperfusion injury (IRI) is inevitable, limiting graft functionality through delayed graft function (DGF) and graft survival. Supplementing the ischemic kidney graft with oxygen during hypothermic preservation has been used in different methods as an attempt to counteract IRI and its effects on graft function and survival. Various oxygenation methods have been studied, from adaptations of classic and well-known preservation strategies, like the addition of oxygen carriers to SCS, or more innovative preservation methods, like hyperbaric oxygenation or retrograde oxygen persufflation. In this review, we will attempt to provide a summary of the available evidence on oxygen carriers, hyperbaric oxygenation, the two-layer method, retrograde oxygen persufflation, and hypothermic oxygenated machine perfusion (HOPE) and discuss the effect these strategies have on kidney graft functionality.
Collapse
Affiliation(s)
| | - Jose Enrique Robles Garcia
- Department of Urology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.C.L.); (C.A.Y.R.); (M.D.T.T.); (V.T.C.); (C.A.M.B.); (B.M.L.); (D.S.Z.)
| | | | | | | | | | | | | |
Collapse
|
11
|
Zhang Y, Hu J, Zhang Y, Ci X. Amentoflavone protects against cisplatin-induced acute kidney injury by modulating Nrf2-mediated oxidative stress and ferroptosis and partially by activating Nrf2-dependent PANoptosis. Front Pharmacol 2025; 16:1508047. [PMID: 40110131 PMCID: PMC11919867 DOI: 10.3389/fphar.2025.1508047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Background Cisplatin is a widely used drug for the treatment of solid organ cancer, but its renal toxicity cannot be ignored. Amentoflavone (AME), a natural flavonoid compound, has remarkable pharmacological effects, including anti-inflammatory and antioxidative effects. The effect and mechanism of AME on cisplatin-induced acute kidney injury (CI-AKI) remain unclear. Methods We investigated the effect of AME on CI-AKI using the HK-2 cell line and C57BL/6 mice. Renal function, tissue damage, and molecular markers were assessed to explore the effects of AME on oxidative stress and cell death pathways. Results In vitro, AME significantly suppressed the cytotoxic effects of cisplatin on HK-2 cells. Furthermore, AME significantly inhibited cisplatin-induced ferroptosis and PANoptosis (apoptosis, pyroptosis and necroptosis). In mice with acute kidney injury induced by a single intraperitoneal injection of cisplatin, the daily administration of AME during AKI effectively improved renal function and alleviated renal tubular injury, characterized by the normalization of blood urea nitrogen (BUN) and serum creatinine (SCr) levels; it also inhibited cisplatin-induced renal ferroptosis and PANoptosis. AME is a natural antioxidant that activates the Nrf2 antioxidant pathway both in vivo and in vitro. In Nrf2 knockout mice and knockdown cells, the protective effect of AME against cisplatin-induced nephrotoxicity disappeared. However, after Nrf2 knockout, the effect of AME on ferroptosis completely disappeared, and that on PANoptosis partially disappeared. Conclusion Amentoflavone has a protective effect on cisplatin-induced acute kidney injury via a mechanism related to the Nrf2-dependent antioxidant pathway and the regulation of ferroptosis and PANoptosis.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Jianqiang Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yanmin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory of Women's Reproductive Health, Changchun, Jilin, China
| |
Collapse
|
12
|
Xiao L, Huang C, Xiao S, Xie L, Zhang X, Xiao F, Cai H, Yang S, Wu S, Qu S, Liu J. Therapeutic effect of umbilical cord mesenchymal stem cells on renal ischemia-reperfusion injury. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2025; 75:103-118. [PMID: 40208785 DOI: 10.2478/acph-2025-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/03/2025] [Indexed: 04/12/2025]
Abstract
Acute kidney injury (AKI) is a growing global health issue with no effective treatments. This study evaluates the therapeutic effects of umbilical cord mesenchymal stem cells (UC-MSCs) on AKI caused by ischemia-reperfusion injury (IRI) in mice. Thirty mice were divided into a sham group, an IRI group, and an MSC-treated group. Renal function was assessed, and histological analysis, immunofluorescence, and real-time PCR were used to evaluate renal damage, inflammatory cell presence, and cytokine expression (TNF-α, IL-6, IL-10). Results showed that MSC treatment reduced renal damage, decreased pro-inflammatory cytokines (TNF-α, IL-6), increased anti-inflammatory IL-10, and promoted kidney repair by homing to injury sites. Thus, umbilical cord MSCs may mitigate AKI by reducing inflammation and enhancing renal repair.
Collapse
Affiliation(s)
- Liang Xiao
- 1Department of Surgery and Oncology, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China
| | - Chengyu Huang
- 2Shenzhen Zhongjia Biomedical Technology Co., Ltd. Shenzhen, Guangdong 518107, China
| | - Shanghua Xiao
- 2Shenzhen Zhongjia Biomedical Technology Co., Ltd. Shenzhen, Guangdong 518107, China
| | - Lingfeng Xie
- 2Shenzhen Zhongjia Biomedical Technology Co., Ltd. Shenzhen, Guangdong 518107, China
| | - Xueyan Zhang
- 2Shenzhen Zhongjia Biomedical Technology Co., Ltd. Shenzhen, Guangdong 518107, China
| | - Fucheng Xiao
- 3The Center of Campus, Shenzhen Senior High School Group, Shenzhen, Guangdong 518040, China
| | - Huajia Cai
- 4Psychiatric Medicine Sophomore, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shuibo Yang
- 5School of Agriculture and Biotechnology Shenzhen Campus of Sun Yat-sen University Shenzhen, Guangdong 518107, China
| | - Shengqing Wu
- 2Shenzhen Zhongjia Biomedical Technology Co., Ltd. Shenzhen, Guangdong 518107, China
| | - Shoukang Qu
- 2Shenzhen Zhongjia Biomedical Technology Co., Ltd. Shenzhen, Guangdong 518107, China
| | - Jia Liu
- 2Shenzhen Zhongjia Biomedical Technology Co., Ltd. Shenzhen, Guangdong 518107, China
- 5School of Agriculture and Biotechnology Shenzhen Campus of Sun Yat-sen University Shenzhen, Guangdong 518107, China
| |
Collapse
|
13
|
Yang J, Zhou R, Zhou M, Li X. Atorvastatin inhibits ischemia‒reperfusion-associated renal tubular cell ferroptosis by blocking the PGE2/EP4 signaling pathway. In Vitro Cell Dev Biol Anim 2025; 61:275-287. [PMID: 39920560 DOI: 10.1007/s11626-025-01020-7] [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: 10/30/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025]
Abstract
Renal ischemia‒reperfusion (I/R) injury is the main cause of acute kidney injury, and its pathological features are manifested primarily by renal tubular epithelial cell injury. The underlying mechanism involves ferroptosis of renal tubular epithelial cells. Atorvastatin (ATO) regulates ferroptosis, and this study explored its role in I/R-induced ferroptosis of renal tubular epithelial cells. We constructed a renal I/R rat model with bilateral renal pedicles using noninvasive arterial clips and placed HK-2 cells in hypoxia/reoxygenation (H/R) incubators to construct the cell model. The damage to rat kidney tissues and HK-2 cells was assessed using enzyme-linked immunosorbent assay (ELISA), hematoxylin and eosin (H&E) staining, and flow cytometry, and the presence of associated proteins was identified through western blotting. Administering ATO markedly lessened the acute kidney damage caused by I/R, decreased the levels of blood urea nitrogen (BUN) and creatinine (CRE), and prevented apoptosis in renal tubular epithelial cells. Treatment with ATO additionally suppressed the production of inflammatory cytokines (TNF-α, IL-1β, and IL-6) and markers linked to ferroptosis (Fe2+, ROS, MDA, ACSL4, and COX2), thereby reducing acute kidney damage associated with I/R. The expression of PGE2 in renal I/R injury is related to the degree of renal injury, and it mainly regulates ferroptosis by binding to EP4. ATO effectively inhibited the expression of PGE2 and EP4. Overall, this study revealed that ATO inhibited ferroptosis of renal tubular epithelial cells by blocking the PGE2/EP4 signaling pathway, thereby alleviating I/R-induced kidney injury.
Collapse
Affiliation(s)
- Jing Yang
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China
| | - Rongrong Zhou
- Expanded Program On Immunization Department, Yunnan Center for Disease Control and Prevention, Kunming, 650022, China
| | - Mengjiao Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xinghuan Li
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China.
| |
Collapse
|
14
|
Shi L, Zha H, Zhao J, An H, Huang H, Xia Y, Yan Z, Song Z, Zhu J. Caloric restriction exacerbates renal post-ischemic injury and fibrosis by modulating mTORC1 signaling and autophagy. Redox Biol 2025; 80:103500. [PMID: 39837191 PMCID: PMC11787690 DOI: 10.1016/j.redox.2025.103500] [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: 12/26/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVE This study investigates the effects of caloric restriction (CR) on renal injury and fibrosis following ischemia-reperfusion injury (IRI), with a focus on the roles of the mechanistic/mammalian target of rapamycin complex 1 (mTORC1) signaling and autophagy. METHODS A mouse model of unilateral IRI with or without CR was used. Renal function was assessed through serum creatinine and blood urea nitrogen levels, while histological analysis and molecular assays evaluated tubular injury, fibrosis, mTORC1 signaling, and autophagy activation. Inducible renal tubule-specific Atg7 knockout mice and autophagy inhibitor 3-MA were used to elucidate autophagy's role in renal outcomes. RESULTS CR exacerbated renal dysfunction, tubular injury, and fibrosis in IRI mice, associated with suppressed mTORC1 signaling and enhanced autophagy. Rapamycin, an mTORC1 inhibitor, mimicked the effects of CR, further supporting the involvement of mTORC1-autophagy pathway. Tubule-specific Atg7 knockout and autophagy inhibitor 3-MA mitigated these effects, indicating a central role for autophagy in CR-induced renal damage. Glucose supplementation, but not branched-chain amino acids (BCAAs), alleviated CR-induced renal fibrosis and dysfunction by restoring mTORC1 activation. Finally, we identified leucyl-tRNA synthetase 1 (LARS1) as a key mediator of nutrient sensing and mTORC1 activation, demonstrating its glucose dependency under CR conditions. CONCLUSION Our study provides novel insights into the interplay between nutrient metabolism, mTORC1 signaling, and autophagy in IRI-induced renal damages, offering potential therapeutic targets for mitigating CR-associated complications after renal IRI.
Collapse
Affiliation(s)
- Lang Shi
- Department of Nephrology, The First Hospital of Lanzhou University, Lanzhou, 730000, China; The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Hongchu Zha
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Juan Zhao
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Haiqian An
- Department of Nephrology, The First Hospital of Lanzhou University, Lanzhou, 730000, China; The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Hua Huang
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Yao Xia
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Ziyu Yan
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, 443000, China
| | - Zhixia Song
- Department of Nephrology, The People's Hospital of Longhua, Shenzhen, 518109, China
| | - Jiefu Zhu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
15
|
Fernández-Acosta R, Vintea I, Koeken I, Hassannia B, Vanden Berghe T. Harnessing ferroptosis for precision oncology: challenges and prospects. BMC Biol 2025; 23:57. [PMID: 39988655 PMCID: PMC11849278 DOI: 10.1186/s12915-025-02154-6] [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: 11/28/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
The discovery of diverse molecular mechanisms of regulated cell death has opened new avenues for cancer therapy. Ferroptosis, a unique form of cell death driven by iron-catalyzed peroxidation of membrane phospholipids, holds particular promise for targeting resistant cancer types. This review critically examines current literature on ferroptosis, focusing on its defining features and therapeutic potential. We discuss how molecular profiling of tumors and liquid biopsies can generate extensive multi-omics datasets, which can be leveraged through machine learning-based analytical approaches for patient stratification. Addressing these challenges is essential for advancing the clinical integration of ferroptosis-driven treatments in cancer care.
Collapse
Affiliation(s)
- Roberto Fernández-Acosta
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Iuliana Vintea
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biobix, Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Ine Koeken
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Behrouz Hassannia
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tom Vanden Berghe
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
16
|
Agudelo-Pérez S, Troncoso G, Botero-Rosas D, Muñoz C, Rodríguez A, Gómez AV, León J. Renal Regional Oxygen Saturation and Acute Kidney Injury in Neonates with Perinatal Asphyxia. Am J Perinatol 2025; 42:379-386. [PMID: 39029915 DOI: 10.1055/a-2369-6811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
OBJECTIVE Neonates with moderate-to-severe perinatal asphyxia often develop acute kidney injury (AKI). Additionally, therapeutic hypothermia (TH) can affect renal blood flow. This study aimed to evaluate the association between renal regional oxygen saturation (rSrO2) during TH and AKI in neonates with moderate and severe perinatal asphyxia. STUDY DESIGN This retrospective longitudinal study included neonates with moderate-to-severe asphyxia who required TH. The primary outcome was the occurrence of AKI, classified as a rate of decrease in creatinine levels of <33% at 72 hours of TH. rSrO2 was continuously monitored by near-infrared spectroscopy during the hypothermia and rewarming phases. Data analysis involved dividing the average rSrO2 levels into 12-hour periods. We analyzed the association between AKI and rSrO2 levels using univariate and multivariate logistic regression models. Furthermore, we assessed the predictive capacity of rSrO2 for AKI by analyzing the area under the receiver operating characteristic curve. RESULTS Ninety-one patients were included in the study. On average, patients with AKI exhibit lower rSrO2 levels during TH. Specifically, rSrO2 levels within the first 12 hours and between 25 and 72 hours of TH demonstrated the highest predictive capability for AKI. Multivariate logistic regression analysis revealed that rSrO2 levels within the initial 12 hours (adjusted odds ratio [aOR] = 1.11, 95% confidence interval [CI]: 1.01-1.21) and between 61 and 72 hours (aOR = 0.85, 95% CI: 0.78-0.92) were significantly associated with AKI. CONCLUSION An increase in rSrO2 during the first 12 hours of TH and lower rSrO2 levels between 61 and 72 hours of treatment were associated with the development of AKI in asphyxiated neonates undergoing TH. KEY POINTS · Neonates with asphyxia often develop AKI.. · Renal saturations are affected by hypothermia and asphyxia. · Patients with AKI initially show higher rSrO2, then lower rSrO2.. · Monitoring rSrO2 identifies early AKI..
Collapse
Affiliation(s)
- Sergio Agudelo-Pérez
- Department of Pediatrics, School of Medicine, Universidad de La Sabana, Chia, Colombia
- Neonatal Unit, Fundación Cardio Infantil-Instituto de Cardiología, Bogotá, Colombia
| | - Gloria Troncoso
- Neonatal Unit, Fundación Cardio Infantil-Instituto de Cardiología, Bogotá, Colombia
| | - Daniel Botero-Rosas
- Department of Bioscience, School of Medicine, Universidad de La Sabana, Chía, Cundinamarca, Colombia
| | - Christian Muñoz
- Department of Pediatrics, School of Medicine, Universidad de La Sabana, Chía, Cundinamarca, Colombia
| | - Andrés Rodríguez
- Department of Pediatrics, School of Medicine, Universidad de La Sabana, Chía, Cundinamarca, Colombia
| | - Andrea Valentina Gómez
- Department of Pediatrics, School of Medicine, Universidad de La Sabana, Chía, Cundinamarca, Colombia
| | - Jennifer León
- Department of Pediatrics, School of Medicine, Universidad de La Sabana, Chía, Cundinamarca, Colombia
| |
Collapse
|
17
|
Liang S, Wu Y, Zhang R, Xu L, Xie F. TNFSF9 Silence Impedes Cerebral Ischemia-Reperfusion Injury via Modulating SLC3A2 Expression in Brain Microvascular Endothelial Cells. J Mol Neurosci 2025; 75:12. [PMID: 39856410 DOI: 10.1007/s12031-025-02310-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI), which stays unresolved in the clinic, occurs after recanalization of blood vessels serving brain tissues in acute ischemic stroke patients and can result in massive brain cell death, and cell ferroptosis contributes greatly to this process. Our research firstly found that TNFSF9 expression harbored diagnostic value on CIRI patients and intended to further investigate its regulatory mechanism in CIRI, which might facilitate its diagnostic and therapeutic application in the clinic. The level of TNSF9 mRNA was augmented in the plasma of CIR patients, and its silence impeded ferroptosis, apoptosis, and release of inflammatory mediators of BMECs with OGD/R treatment. Besides, SP1 positively regulated TNFSF9 expression as one of its transcription factors, and TNFSF9 overexpression reversed SP1 silence-mediated inhibition on ferroptosis, apoptosis, and release of inflammatory mediators in OGD/R-treated BMECs. In addition, silencing SLC3A2 could neutralize the benefit effects of TNFSF9 downregulation on BMECs under OGD/R context in vitro, and silencing TNFSF9 neutralized necrotic volumes in rat brain induced by CIRI via modulating SLC3A2 expression in vivo. TNFSF9 regulated by SP1 aggravated CIRI via boosting ferroptosis, apoptosis, and release of inflammatory mediators of BMECs under OGD/R situation by suppressing SLC3A2 expression in vitro and in vivo.
Collapse
Affiliation(s)
- Shunli Liang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
- The Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou City, 310053, Zhejiang, China
| | - You Wu
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
| | - Rongbo Zhang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
| | - Linsheng Xu
- Department of Neurology, the Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310005, Zhejiang, China
| | - Fangping Xie
- Department of Special Examination, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, No. 305 Tianmushan Road, Hangzhou City, 310013, Zhejiang, China.
| |
Collapse
|
18
|
Pefanis A, Bongoni AK, McRae JL, Salvaris EJ, Fisicaro N, Murphy JM, Ierino FL, Cowan PJ. Inhibition of RIPK1 or RIPK3 kinase activity post ischemia-reperfusion reduces the development of chronic kidney injury. Biochem J 2025; 482:73-86. [PMID: 39705008 DOI: 10.1042/bcj20240569] [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: 09/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/21/2024]
Abstract
Ischemia-reperfusion injury (IRI) occurs when the blood supply to an organ is temporarily reduced and then restored. Kidney IRI is a form of acute kidney injury (AKI), which often progresses to kidney fibrosis. Necroptosis is a regulated necrosis pathway that has been implicated in kidney IRI. Necroptotic cell death involves the recruitment of the RIPK1 and RIPK3 kinases and the activation of the terminal effector, the mixed lineage kinase domain-like (MLKL) pseudokinase. Phosphorylated MLKL causes cell death by plasma membrane rupture, driving 'necroinflammation'. Owing to their apical role in the pathway, RIPK1 and RIPK3 have been implicated in the development of kidney fibrosis. Here, we used a mouse model of unilateral kidney IRI to assess whether the inhibition of RIPK1 or RIPK3 kinase activity reduces AKI and the progression to kidney fibrosis. Mice treated with the RIPK1 inhibitor Nec-1s, either before or after IR, showed reduced kidney injury at 24 hr compared with controls, whereas no protection was offered by the RIPK3 inhibitor GSK´872. In contrast, treatment with either inhibitor from days 3 to 9 post-IR reduced the degree of kidney fibrosis at day 28. These findings further support the role of necroptosis in IRI and provide important validation for the contribution of both RIPK1 and RIPK3 catalytic activities in the progression of kidney fibrosis. Targeting the necroptosis pathway could be a promising therapeutic strategy to mitigate kidney disease following IR.
Collapse
Affiliation(s)
- Aspasia Pefanis
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Nephrology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Anjan K Bongoni
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Jennifer L McRae
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Evelyn J Salvaris
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Nella Fisicaro
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Francesco L Ierino
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Nephrology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Peter J Cowan
- Immunology Research Centre, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
Okami N, Wakui H, Azushima K, Miyazawa T, Kubo E, Tsukamoto S, Sotozawa M, Taguchi S, Urate S, Ishiga K, Kinguchi S, Kanaoka T, Tamura K. Leucine-rich alpha-2-glycoprotein 1 deficiency suppresses ischemia-reperfusion injury-induced renal fibrosis. Sci Rep 2025; 15:1259. [PMID: 39779883 PMCID: PMC11711393 DOI: 10.1038/s41598-024-84798-y] [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: 08/01/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Ischemia reperfusion injury (IRI) is a major cause of acute kidney injury (AKI) and ultimately leads to renal fibrosis, primarily via the transforming growth factor-β (TGF-β) pathway. Leucine-rich alpha-2-glycoprotein 1 (LRG1), a novel modulator of the TGF-β pathway, has been implicated in the modulation of renal fibrosis by affecting the TGF-β/Smad3 signaling axis. However, the role of LRG1 in the transition from AKI to chronic kidney disease (CKD) remains unclear. This study aimed to investigate the functional role of LRG1 during the remodeling phase post-IRI. Unilateral IRI was induced in C57BL/6J wild-type (WT) mice and systemic LRG1 knockout (KO) mice. In C57BL/6J WT mice, renal LRG1 mRNA expression was significantly elevated on the ischemia/reperfusion side compared to the sham side over a 28-day period. In contrast, LRG1 KO mice demonstrated significantly reduced renal fibrosis compared to WT mice on postoperative day 28. Additionally, renal mRNA expression of TGF-β and associated pro-fibrotic genes was diminished in LRG1 KO mice compared to WT mice. Consequently, LRG1 KO mice exhibited attenuated IRI-induced chronic fibrosis. These findings indicate that LRG1 is involved in the pathogenesis of the transition from AKI to CKD and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Naohito Okami
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Tomohito Miyazawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Eisuke Kubo
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Shunichiro Tsukamoto
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Mari Sotozawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Shinya Taguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Shingo Urate
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kohei Ishiga
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
- Yokohama City University Medical Center, Yokohama, Japan
| | - Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
- Yokohama City University Medical Center, Yokohama, Japan
| |
Collapse
|
20
|
Ozgen ZE, Erdinc M, Kaya MS, Aktar F, Ozekinci SO, Erdinc L, Uyar E. Involvement of necroptosıs and apoptosıs ın protectıve effects of cyclosporın a on ischemıa-reperfusıon injury in rat kıdney. J Mol Histol 2024; 56:30. [PMID: 39630315 DOI: 10.1007/s10735-024-10281-7] [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: 09/22/2024] [Accepted: 11/06/2024] [Indexed: 02/07/2025]
Abstract
We aimed to investigate the protective effects of low dose cyclosporin A (CsA) on ischemia-reperfusion (IR) injury in rat the kidney and on the apoptotic and necroptotic mechanisms involved. 1. Control group (received a single intraperitoneal (i.p.) dose of 1 ml sterile saline 15 min before the surgical procedure), 2. IR group (was subjected to 30 min of bilateral kidney ischemia followed by 90 min of reperfusion; and received a single i.p. dose of 1 ml sterile saline 15 min before the IR procedure, 3. IR + CsA group (received a single i.p. dose of 3 mg/kg CsA 15 min before the IR procedure. Renal functions (renal perfusion pressures, and serum urea-creatinine levels), kidney histological scores, MDA levels, and TNF-α, caspase-3, RIP1, RIP3, MLKL, CaMKII and CypD protein expressions were also measured. Renal perfusion pressures (PP), serum urea and creatinine levels, renal tissue MDA levels, and the protein expression levels of TNF-α, caspase-3, RIP1, RIP3, MLKL, CAMKII and CypD were significantly increased in the IR group compared to the control group (p < 0.05), Additionally, there were significant decreases in all the parameters in the IR + CsA group compared to those in the IR group (p < 0.05). Furthermore, histopathological analyses revealed significantly higher kidney injury scores in the IR group compared to the control group, and low dose CsA treatment improved the injury. A single low dose of CsA injection 15 min before IR, demonstrated a protective effect on bilateral renal IR injury and a reduction in apoptotic and necroptopic markers which is resulted in improvement of renal functions.
Collapse
Affiliation(s)
- Zeynep Erdogmus Ozgen
- Department of Pharmacology, School of Pharmacy, Dicle University, Diyarbakir, Turkey.
| | - Meral Erdinc
- Department of Pharmacology, School of Medicine, Dicle University, Diyarbakir, Turkey
| | - Meryem Seyda Kaya
- Department of Pharmacology, School of Pharmacy, Dicle University, Diyarbakir, Turkey
| | - Fesih Aktar
- Department of Pediatrics, School of Medicine, Dicle University, Diyarbakir, Turkey
| | | | - Levent Erdinc
- Department of Biochemistry, School of Medicine, Dicle University, Diyarbakir, Turkey
| | - Emre Uyar
- Department of Pharmacology, School of Medicine, Uskudar University, Istanbul, Turkey
| |
Collapse
|
21
|
Lee K, Jang HR, Rabb H. Lymphocytes and innate immune cells in acute kidney injury and repair. Nat Rev Nephrol 2024; 20:789-805. [PMID: 39095505 DOI: 10.1038/s41581-024-00875-5] [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: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Acute kidney injury (AKI) is a common and serious disease entity that affects native kidneys and allografts but for which no specific treatments exist. Complex intrarenal inflammatory processes driven by lymphocytes and innate immune cells have key roles in the development and progression of AKI. Many studies have focused on prevention of early injury in AKI. However, most patients with AKI present after injury is already established. Increasing research is therefore focusing on mechanisms of renal repair following AKI and prevention of progression from AKI to chronic kidney disease. CD4+ and CD8+ T cells, B cells and neutrophils are probably involved in the development and progression of AKI, whereas regulatory T cells, double-negative T cells and type 2 innate lymphoid cells have protective roles. Several immune cells, such as macrophages and natural killer T cells, can have both deleterious and protective effects, depending on their subtype and/or the stage of AKI. The immune system not only participates in injury and repair processes during AKI but also has a role in mediating AKI-induced distant organ dysfunction. Targeted manipulation of immune cells is a promising therapeutic strategy to improve AKI outcomes.
Collapse
Affiliation(s)
- Kyungho Lee
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Samsung Medical Center, Cell and Gene Therapy Institute, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Wu Y, Shi H, Xu Y, Shu G, Xiao Y, Hong G, Xu S. Targeted Restoration of GPX3 Attenuates Renal Ischemia/Reperfusion Injury by Balancing Selenoprotein Expression and Inhibiting ROS-mediated Mitochondrial Apoptosis. Transplantation 2024; 108:2351-2365. [PMID: 38771110 DOI: 10.1097/tp.0000000000005068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
BACKGROUND Renal ischemia/reperfusion (IR) injury is the leading cause of acute kidney injury in both autologous and transplanted kidneys. Low-level glutathione peroxidase 3 (GPX3) is associated with renal IR injury. The exact mechanism of targeted GPX3 restoration in renal IR injury has yet to be determined. METHODS The distribution of GPX3 in different tissues and organs of the body was investigated. The level of GPX3 in renal IR injury was assessed. To confirm the action of GPX3 and its mechanisms, IR models were used to introduce adeno-associated virus 9 containing GPX3, as well as hypoxia/reoxygenation-exposed normal rat kidney cells that consistently overexpressed GPX3. Reverse molecular docking was used to confirm whether GPX3 was a target of ebselen. RESULTS GPX3 is abundant in the kidneys and decreases in expression during renal IR injury. GPX3 overexpression reduced renal IR injury and protected tubular epithelial cells from apoptosis. Proteomics analysis revealed a strong link between GPX3 and mitochondrial signaling, cellular redox state, and different expression patterns of selenoproteins. GPX3 inhibited reactive oxygen species-induced mitochondrial apoptosis and balanced the disordered expression of selenoproteins. GPX3 was identified as a stable selenoprotein that interacts with ebselen. Ebselen enhanced the level of GPX3 and reduced IR-induced mitochondrial damage and renal dysfunction. CONCLUSIONS Targeted restoration of GPX3 attenuates renal IR injury by balancing selenoprotein expression and inhibiting reactive oxygen species-mediated mitochondrial apoptosis, indicating that GPX3 could be a potential therapeutic target for renal IR injury.
Collapse
Affiliation(s)
- Yikun Wu
- Guizhou University Medical College, Guiyang, China
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hua Shi
- Department of Urology, Tongren City People's Hospital, Tongren, China
| | - Yuangao Xu
- Clinic for Kidney and Hypertension Diseases, Hannover Medical School, Hannover, Germany
| | - Guofeng Shu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yu Xiao
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guangyi Hong
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shuxiong Xu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
23
|
Asli F, Poshtdar S, Dehpour AR, Mohammad Jafari R. Exploring the therapeutic potential of Modafinil in mitigating renal ischemia-reperfusion injury in rats. Fundam Clin Pharmacol 2024; 38:1168-1177. [PMID: 39138143 DOI: 10.1111/fcp.13034] [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: 01/27/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Renal ischemia reperfusion injury (IRI) is a post-ischemic event, which can lead to subsequent acute kidney injury (AKI), transplant failure, renal dysfunction and fibrosis via heightened oxidative stress and production of inflammatory cytokines and chemokines. OBJECTIVE This study aims to assess the effect of Modafinil, a wake-promoting agent with previously proven anti-inflammatory and anti-oxidative properties, on ameliorating renal IRI. METHODS A total of 30 male Wistar rats were divided into five groups: Sham-operated group, ischemia reperfusion (I/R) control group and Modafinil pre-treated groups (at different doses of 50, 100 and 150 mg/kg). IRI was induced by means of bilaterally clamping the renal arteries for 45 min, followed by 24 h of reperfusion. RESULTS Tissue pathological assessments demonstrated a reduction of glomerular, vascular and interstitial injury at doses of 50 and 100 mg/kg of Modafinil. The biochemical studies showed a significant decrease in tissue pro-inflammatory factors, including tumor necrosis factor alpha (TNF-α), Interleukin-18 (IL-18) and lactate dehydrogenase (LDH). Moreover, an elevation was observed in levels of super oxide dismutase (SOD) and catalase, indicating the reduction of oxidative stress. Furthermore, the levels of creatinine (Cr), urea and neutrophil gelatinase-associated lipocalin (NGAL) were declined, indicating the improvement in renal function at effective doses of Modafinil (50 and 100 mg/kg) compared to the I/R control group without Modafinil pre-treatment. CONCLUSION Our findings suggest that Modafinil holds promise as an effective therapeutic agent to address the clinical challenges associated with kidney IRI reducing the need for hospitalization and potentially alleviating related morbidities.
Collapse
Affiliation(s)
- Fatemeh Asli
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Poshtdar
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Ji J, Ma Y, Liu X, Zhou Q, Zheng X, Chen Y, Li Z, Yang L. Identification of Renal Ischemia-Reperfusion Injury Subtypes and Predictive Model for Graft Loss after Kidney Transplantation Based on Programmed Cell Death-Related Genes. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:450-467. [PMID: 39664334 PMCID: PMC11631021 DOI: 10.1159/000540158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/26/2024] [Indexed: 12/13/2024]
Abstract
Introduction Ischemia-reperfusion injury (IRI) is detrimental to kidney transplants and may contribute to poor long-term outcomes of transplantation. Programmed cell death (PCD), a regulated cell death form triggered by IRI, is often indicative of an unfavorable prognosis following transplantation. However, given the intricate pathophysiology of IRI and the considerable variability in clinical conditions during kidney transplantation, the specific patterns of cell death within renal tissues remain ambiguous. Consequently, accurately predicting the outcomes for transplanted kidneys continues to be a formidable challenge. Methods Eight Gene Expression Omnibus datasets of biopsied transplanted kidney samples post-IRI and 1,548 PCD-related genes derived from 18 PCD patterns were collected in our study. Consensus clustering was performed to identify distinct IRI subtypes based on PCD features (IRI PCD subtypes). Differential enrichment analysis of cell death, metabolic signatures, and immune infiltration across these subtypes was evaluated. Three machine learning algorithms were used to identify PCD patterns related to prognosis. Genes associated with graft loss were screened for each PCD type. A predictive model for graft loss was constructed using 101 combinations of 10 machine learning algorithms. Results Four IRI subtypes were identified: PCD-A, PCD-B, PCD-C, and PCD-D. PCD-A, characterized by high enrichment of multiple cell death patterns, significant metabolic paralysis, and immune infiltration, showed the poorest prognosis among the four subtypes. While PCD-D involved the least kind of cell death patterns with the features of extensive activation of metabolic pathways and the lowest immune infiltration, correlating with the best prognosis in the four subtypes. Using various machine learning algorithms, 10 cell death patterns and 42 PCD-related genes were identified as positively correlated with graft loss. The predictive model demonstrated high sensitivity and specificity, with area under the curve values for 0.5-, 1-, 2-, 3-, and 4-year graft survival at 0.888, 0.91, 0.926, 0.923, and 0.923, respectively. Conclusion Our study explored the comprehensive features of PCD patterns in transplanted kidney samples post-IRI. The prediction model shows great promise in forecasting graft loss and could aid in risk stratification in patients following kidney transplantation.
Collapse
Affiliation(s)
- Jing Ji
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuan Ma
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xintong Liu
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingqing Zhou
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xizi Zheng
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Chen
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zehua Li
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Yang
- Renal Division, Peking University Institute of Nephrology, Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Peking University First Hospital, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Shamshirgaran A, Mohammadi A, Zahmatkesh P, Mesbah G, Guitynavard F, Saffarian Z, Khajavi A, Oliveira Reis L, Aghamir SMK. The Use of Autologous Omentum Transposition as a Therapeutic Intervention to Reduce the Complication of Ischemia/Reperfusion Injuries in a Rat Model. Can J Kidney Health Dis 2024; 11:20543581241300773. [PMID: 39610662 PMCID: PMC11603481 DOI: 10.1177/20543581241300773] [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: 08/01/2024] [Accepted: 10/07/2024] [Indexed: 11/30/2024] Open
Abstract
Background Ischemia/reperfusion injury (IRI) causes cellular dysfunction and death in organs like the kidney, heart, and brain. It involves energy depletion during ischemia and oxidative stress, inflammation, and apoptosis during reperfusion. Kidney IRI often leads to acute kidney injury (AKI) in various clinical scenarios. The omentum, an adipose tissue with healing properties, has been used to treat injuries in different organs. Objective This study aimed to assess the omentum's healing effects on reducing IRI's adverse effects after renal ischemia in Wistar rats. Method A total number of 36 male Wistar rats were used in a study on IRI-induced AKI. Rats were divided into 6 groups of normal kidneys wrapped with omentum "Sham-1" and "Sham-2," ischemic kidney wrapped with omentum as "OMT-1" and "OMT-2," and ischemic kidney without omentum as "Control-1" and "Control-2." Ischemia was induced by clamping the left renal artery for 45 minutes. The omentum was transposed onto the injured kidney in "OMT" group. After sacrifice at weeks 4 and 8, kidney histology and blood samples were analyzed for kidney function markers. Results On the first day after surgery, there was an immediate increase in creatinine and blood urea nitrogen (BUN) levels, which then decreased by day 28. Both OMT groups showed significantly lower levels of creatinine and BUN compared to Control groups on day 1, but after 28 days differences were not statistically significant. Histological analysis using H&E and Masson's trichrome staining revealed significantly higher levels of inflammatory cell infiltration and hyperemia in the OMT groups. However, fibrosis and glomerular shrinkage were higher in the Control groups. Conclusion Using an omental flap significantly prevented fibrosis within the renal parenchyma, slow down the AKI progression, and potentially serving as a promising therapeutic strategy for kidney dysfunction.
Collapse
Affiliation(s)
| | | | - Parisa Zahmatkesh
- Urology Research Center, Tehran University of Medical Sciences, Iran
| | - Gholamreza Mesbah
- Urology Research Center, Tehran University of Medical Sciences, Iran
| | | | - Zahra Saffarian
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Alireza Khajavi
- Urology Research Center, Tehran University of Medical Sciences, Iran
- Student Research Committee, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leonardo Oliveira Reis
- UroScience, State University of Campinas, Unicamp, São Paulo, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, São Paulo, Brazil
| | | |
Collapse
|
26
|
Chang TT, Li SY, Tsai MT, Chiang CH, Chen C, Chen JW. CXCL5 inhibition ameliorates acute kidney injury and prevents the progression from acute kidney injury to chronic kidney disease. Clin Sci (Lond) 2024; 138:1451-1466. [PMID: 39503534 DOI: 10.1042/cs20241713] [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: 08/22/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 11/08/2024]
Abstract
Acute kidney injury (AKI) increases the risk of chronic kidney disease (CKD). CXC motif chemokine ligand 5 (CXCL5) is up-regulated in kidney diseases. We aimed to investigate the direct effect of CXCL5 on the pathology of AKI. Serum and renal expression of CXCL5 were increased in animals with renal ischemia-reperfusion injury or unilateral ureteral obstruction. CXCL5-knockout mice exhibited reduced systemic oxidative stress and preserved renal function in the acute and chronic phases of AKI, as evidenced by reductions in serum BUN and creatinine levels, the urinary albumin-to-creatinine ratio, and the kidney-to-body weight ratio. CXCL5-knockout mice improved AKI-induced tubular injury and fibrosis, reduced renal macrophage infiltration, and reduced expression of NADPH oxidase and inflammatory and fibrotic proteins. CXCL5 activated p47 to up-regulate ROS generation and induce cellular damages through CXCR2. CXCL5 knockdown exerted antioxidative, anti-inflammatory, anti-fibrotic, and anti-apoptotic effects on hypoxia-reoxygenation-stimulated renal proximal tubular epithelial cells. Clinical data indicated elevated circulating and renal CXCL5 in CKD patients, and renal CXCL5 was correlated with increased renal fibrosis and decreased estimated glomerular filtration rate. Altogether, CXCL5 levels increased in experimental AKI and clinical CKD, and in vivo and in vitro CXCL5 inhibition may reduce acute tubular injury and prevent the subsequent progression from AKI to CKD.
Collapse
Affiliation(s)
- Ting-Ting Chang
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Szu-Yuan Li
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Tsun Tsai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chih-Hung Chiang
- Division of Urology, Department of Surgery and Department of Research and Development, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, Taiwan
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching Chen
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Department and Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan
- Division of Cardiology, Taipei Medical University Hospital, Taipei, Taiwan
- Faucalty of Medicine, Colleague of Medicine, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
27
|
Xu C, Deng Y, Gong X, Wang H, Man J, Wang H, Cheng K, Gui H, Fu S, Wei S, Zheng X, Che T, Ding L, Yang L. Exploring Cuproptosis-Related Genes and Diagnostic Models in Renal Ischemia-Reperfusion Injury Using Bioinformatics, Machine Learning, and Experimental Validation. J Inflamm Res 2024; 17:8997-9020. [PMID: 39583859 PMCID: PMC11583769 DOI: 10.2147/jir.s490357] [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: 09/04/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024] Open
Abstract
Background Renal ischemia-reperfusion injury (RIRI) is a significant cause of acute kidney injury, complicating clinical interventions such as kidney transplants and partial nephrectomy. Recent research has indicated the role of cuproptosis, a copper-dependent cell death pathway, in various pathologies, but its specific involvement in RIRI remains insufficiently understood. This study aims to investigate the role of cuproptosis-related genes in RIRI and establish robust diagnostic models. Methods We analyzed transcriptomic data from 203 RIRI and 188 control samples using bioinformatics tools to identify cuproptosis-related differentially expressed genes (CRDEGs). The relationship between CRDEGs and immune cells was explored using immune infiltration analysis and correlation analysis. Gene Set Enrichment Analysis (GSEA) was conducted to identify pathways associated with CRDEGs. Machine learning models, including Least Absolute Shrinkage and Selection Operator(LASSO) logistic regression, Support Vector Machine Recursive Feature Elimination (SVM-RFE), Clustering analysis, and weighted gene co-expression network analysis (WGCNA), were used to construct diagnostic gene models. The models were validated using independent datasets. Experimental validation was conducted in vivo using a mouse bilateral RIRI model and in vitro using an HK-2 cell hypoxia-reoxygenation (HR) model with copper chelation intervention. HE, PAS, and TUNEL staining, along with plasma creatinine and blood urea nitrogen (BUN) measurements, were used to evaluate the protective effect of the copper chelator D-Penicillamine (D-PCA) on RIRI in mice. JC-1 and TUNEL staining were employed to assess apoptosis in HK-2 cells under hypoxia-reoxygenation conditions. Immunofluorescence and Western blot (WB) techniques were used to verify the expression levels of the SDHB and NDUFB6 genes. Results A total of 18 CRDEGs were identified, many of which were significantly correlated with immune cell infiltration. GSEA revealed that these genes were involved in pathways related to oxidative phosphorylation and immune response regulation. Four key cuproptosis marker genes (LIPA, LIPT1, SDHB, and NDUFB6) were incorporated into a Cuproptosis Marker Gene Model(CMGM), achieving an area under the curve (AUC) of 0.741-0.834 in validation datasets. In addition, a five-hub-gene SVM model (MOAP1, PPP2CA, SYL2, ZZZ3, and SFRS2) was developed, demonstrating promising diagnostic performance. Clustering analysis revealed two RIRI subtypes (C1 and C2) with distinct molecular profiles and pathway activities, particularly in oxidative phosphorylation and immune responses. Experimental results showed that copper chelation alleviated renal damage and cuproptosis in both in vivo and in vitro models. Conclusion Our study reveals that cuproptosis-related genes are significantly involved in RIRI, particularly influencing mitochondrial dysfunction and immune responses. The diagnostic models developed showed promising predictive performance across independent datasets. Copper chelation demonstrated potential therapeutic effects, suggesting that cuproptosis regulation may be a viable therapeutic strategy for RIRI. This work provides a foundation for further exploration of copper metabolism in renal injury contexts.
Collapse
Affiliation(s)
- Changhong Xu
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Yun Deng
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Xinyi Gong
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Huabin Wang
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Jiangwei Man
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Hailong Wang
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Kun Cheng
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Huiming Gui
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Shengjun Fu
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Shenghu Wei
- School of Physical Science and Technology, Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Xiaoling Zheng
- Innovation Center of Functional Genomics and Molecular Diagnostics Technology of Gansu Province, Lanzhou, People’s Republic of China
| | - Tuanjie Che
- Innovation Center of Functional Genomics and Molecular Diagnostics Technology of Gansu Province, Lanzhou, People’s Republic of China
| | - Liyun Ding
- School of Physical Science and Technology, Lanzhou University, Lanzhou, 730000, People’s Republic of China
| | - Li Yang
- Department of Urology, Institute of Urology, Gansu Province Clinical Research Center for Urinary System Disease, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| |
Collapse
|
28
|
Wang JH, Mao HB, Hu JB, Cheng S, Su H. Engineering of phosphatidylserine-targeting ROS-responsive polymeric prodrug for the repair of ischemia-reperfusion-induced acute kidney injury. J Control Release 2024; 376:1100-1114. [PMID: 39486459 DOI: 10.1016/j.jconrel.2024.10.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/13/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Ischemia-reperfusion-induced acute kidney injury (IR-AKI) commonly occurs in situations such as hemorrhagic shock, kidney transplantation, and cardiovascular surgery. As one of the significant causes of AKI, IR-AKI is characterized by its high incidence and mortality rates. Currently, effective inflammation control is the key for the treatment of IR-AKI. In this study, we developed an ROS-responsive polymeric prodrugs (Zn-D/DTH) which could target the externalized PS of apoptotic cells, and then responsively released HDM (anti-inflammatory peptides) in the presence of intracellular ROS. Zn-D/DTH effectively ameliorated renal function and mitigated pathological alterations such as the loss of the brush border, tubular dilation, and accumulation of cellular debris within the tubular lumens. Furthermore, Zn-D/DTH greatly reduced the generation of pro-inflammatory factors like IL-6, COX-2, and iNOS in renal tissues, suggesting its protective role largely stems from suppression of the inflammatory response. Additional mechanism exploration revealed that Zn-D/DTH markedly decreased the expression levels of TLR4 and MyD88, as well as the phosphorylation of NF-κB in the damaged kidneys. This, in turn, reduced the number of apoptotic tubular cells and the activity of Caspase 9 and Caspase 3 caused by ischemia-reperfusion. Additionally, Zn-D/DTH treatment showed improvement in the long-term renal damage and fibrosis induced by ischemia-reperfusion. The experimental outcomes indicated that Zn-D/DTH attenuated renal ischemia-reperfusion injury and delayed the transition from acute kidney injury to chronic kidney disease by downregulating the TLR4/MyD88/NF-κB signaling pathway and reducing the expression of apoptotic caspases, thereby inhibiting inflammation and reducing cell apoptosis.
Collapse
Affiliation(s)
- Jin-Hui Wang
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Hai-Bo Mao
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Jing-Bo Hu
- Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Shunhua Cheng
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| | - Hao Su
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
29
|
Liu M, Chen J, Sun M, Zhang L, Yu Y, Mi W, Ma Y, Wang G. Protection of Ndrg2 deficiency on renal ischemia-reperfusion injury via activating PINK1/Parkin-mediated mitophagy. Chin Med J (Engl) 2024; 137:2603-2614. [PMID: 38407220 PMCID: PMC11556958 DOI: 10.1097/cm9.0000000000002957] [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: 11/04/2023] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Renal ischemia-reperfusion (R-I/R) injury is the most prevalent cause of acute kidney injury, with high mortality and poor prognosis. However, the underlying pathological mechanisms are not yet fully understood. Therefore, this study aimed to investigate the role of N-myc downstream-regulated gene 2 ( Ndrg2 ) in R-I/R injury. METHODS We examined the expression of Ndrg2 in the kidney under normal physiological conditions and after R-I/R injury by immunofluorescence staining, real-time polymerase chain reaction, and western blotting. We then detected R-I/R injury in Ndrg2-deficient ( Ndrg2-/- ) mice and wild type ( Ndrg2+/+ ) littermates in vivo , and detected oxygen and glucose deprivation and reperfusion (OGD-R) injury in HK-2 cells. We further conducted transcriptomic sequencing to investigate the role of Ndrg2 in R-I/R injury and detected levels of oxidative stress and mitochondrial damage by dihydroethidium staining, biochemical assays, and western blot. Finally, we measured the levels of mitophagy in Ndrg2+/+ and Ndrg2-/- mice after R-I/R injury or HK-2 cells in OGD-R injury. RESULTS Ndrg2 was primarily expressed in renal proximal tubules and its expression was significantly decreased 24 h after R-I/R injury. Ndrg2-/- mice exhibited significantly attenuated R-I/R injury compared to Ndrg2+/+ mice. Transcriptomics profiling showed that Ndrg2 deficiency induced perturbations of multiple signaling pathways, downregulated inflammatory responses and oxidative stress, and increased autophagy following R-I/R injury. Further studies revealed that Ndrg2 deficiency reduced oxidative stress and mitochondrial damage. Notably, Ndrg2 deficiency significantly activated phosphatase and tensin homologue on chromosome ten-induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy. The downregulation of NDRG2 expression significantly increased cell viability after OGD-R injury, increased the expression of heme oxygenase-1, decreased the expression of nicotinamide adenine dinucleotide phosphate oxidase 4, and increased the expression of the PINK1/Parkin pathway. CONCLUSION Ndrg2 deficiency might become a therapy target for R-I/R injury by decreasing oxidative stress, maintaining mitochondrial homeostasis, and activating PINK1/Parkin-mediated mitophagy.
Collapse
Affiliation(s)
- Min Liu
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jianwen Chen
- Department of Nephrology, The First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Miao Sun
- Department of Anesthesiology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Lixia Zhang
- Department of Burn and Plastic Surgery, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Yao Yu
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Weidong Mi
- Department of Anesthesiology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yulong Ma
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
30
|
Giuliani KTK, Adams BC, Healy HG, Kassianos AJ. Regulated cell death in chronic kidney disease: current evidence and future clinical perspectives. Front Cell Dev Biol 2024; 12:1497460. [PMID: 39544363 PMCID: PMC11560912 DOI: 10.3389/fcell.2024.1497460] [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: 09/17/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Chronic kidney disease (CKD) is the progressive loss of kidney function/structure over a period of at least 3 months. It is characterised histologically by the triad of cell loss, inflammation and fibrosis. This literature review focuses on the forms of cell death that trigger downstream inflammation and fibrosis, collectively called regulated cell death (RCD) pathways. Discrete forms of RCD have emerged as central mediators of CKD pathology. In particular, pathways of regulated necrosis - including mitochondrial permeability transition pore (mPTP)-mediated necrosis, necroptosis, ferroptosis and pyroptosis - have been shown to mediate kidney pathology directly or through the release of danger signals that trigger a pro-inflammatory response, further amplifying tissue injury in a cellular process called necroinflammation. Despite accumulating evidence in pre-clinical models, no clinical studies have yet targeted these RCD modes in human CKD. The review summarizes recent advances in our understanding of RCD pathways in CKD, looks at inter-relations between the pathways (with the emphasis on propagation of death signals) and the evidence for therapeutic targeting of molecules in the RCD pathways to prevent or treat CKD.
Collapse
Affiliation(s)
- Kurt T. K. Giuliani
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Benjamin C. Adams
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Helen G. Healy
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Kassianos
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
31
|
Wang X, Xu S, Yan Y, Liu Z, Guo Y, Zhang T, Liu Y, Jiao W. Multi-omics analysis of renal vein serum with Ischemia-Reperfusion injury. Gene 2024; 926:148650. [PMID: 38851364 DOI: 10.1016/j.gene.2024.148650] [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: 11/25/2023] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Acute kidney injury (AKI) is frequently caused by renal ischemia-reperfusion injury (IRI). Identifying potential renal IRI disease biomarkers would be useful for evaluating AKI severity. OBJECTIVE We used proteomics and metabolomics to investigate the differences in renal venous blood between ischemic and healthy kidneys in an animal model by identifying differentially expressed proteins (DEPs) and differentially expressed protein metabolites (DEMs). METHODS Nine pairs of renal venous blood samples were collected before and at 20, 40, and 60 min post ischemia. The ischemia time of Group A, B and C was 20,40 and 60 min. The proteome and metabolome of renal venous blood were evaluated to establish the differences between renal venous blood before and after ischemia. RESULTS We identified 79 common DEPs in all samples of Group A, 80 in Group B, and 131 in Group C. Further common DEPs among all three groups were Tyrosineprotein kinase, GPR15LG, KAZALD1, ADH1B. We also identified 81, 64, and 83 common DEMs in each group respectively, in which 30 DEMs were further common to all groups. Bioinformatic analysis of the DEPs and DEMs was conducted. CONCLUSION This study demonstrated that different pathological processes occur during short- and long-term renal IRI. Tyrosine protein kinase, GPR15LG, Kazal-type serine peptidase inhibitor domain 1, and all-trans-retinol dehydrogenase are potential biomarkers of renal IRI.
Collapse
Affiliation(s)
- Xinning Wang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Shang Xu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yongchao Yan
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Zhilong Liu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yize Guo
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Tianwei Zhang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Yunbo Liu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Wei Jiao
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
32
|
Zhang H, Deng Z, Wang Y, Zheng X, Zhou L, Yan S, Wang Y, Dai Y, Kanwar YS, Chen F, Deng F. CHIP drives proteasomal degradation of NUR77 to alleviate oxidative stress and intrinsic apoptosis in cisplatin-induced nephropathy. Commun Biol 2024; 7:1403. [PMID: 39462094 PMCID: PMC11513124 DOI: 10.1038/s42003-024-07118-0] [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: 03/04/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Carboxy-terminus of Hsc70-interacting protein (CHIP), an E3 ligase, modulates the stability of its targeted proteins to alleviate various pathological perturbations in various organ systems. Cisplatin is a widely used chemotherapeutic agent, but it is also known for its alarming renal toxicity. The role of CHIP in the pathogenesis of cisplatin-induced acute kidney injury (AKI) has not been adequately investigated. Herein, we demonstrated that CHIP was abundantly expressed in the renal proximal tubular epithelia, and its expression was downregulated in cisplatin-induced AKI. Further investigation revealed that CHIP overexpression or activation alleviated, while its gene disruption promoted, oxidative stress and apoptosis in renal proximal tubular epithelia induced by cisplatin. In terms of mechanism, CHIP interacted with and ubiquitinated NUR77 to promote its degradation, which consequently shielded BCL2 to maintain mitochondrial permeability of renal proximal tubular cells in the presence of cisplatin. Also, we demonstrated that CHIP interacted with NUR77 via its central coiled-coil (CC) domain, a non-canonical interactive pattern. In conclusion, these findings indicated that CHIP ubiquitinated and degraded its substrate NUR77 to attenuate intrinsic apoptosis in cisplatin-treated renal proximal tubular epithelia, thus providing a novel insight for the pathogenesis of cisplatin-induced AKI.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Zebin Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Yilong Wang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Zheng
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Lizhi Zhou
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Shu Yan
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Yashpal S Kanwar
- Departments of Pathology & Medicine, Northwestern University, Chicago, IL, USA
| | - Fangzhi Chen
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
| | - Fei Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
33
|
Zhao Y, Wang Q, Zhu J, Cai J, Feng X, Song Q, Jiang H, Ren W, He Y, Wang P, Feng D, Yu J, Liu Y, Wu Q, Jitkaew S, Cai Z. Identification of KW-2449 as a dual inhibitor of ferroptosis and necroptosis reveals that autophagy is a targetable pathway for necroptosis inhibitors to prevent ferroptosis. Cell Death Dis 2024; 15:764. [PMID: 39433736 PMCID: PMC11493980 DOI: 10.1038/s41419-024-07157-9] [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: 03/19/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024]
Abstract
Necroptosis and ferroptosis are two distinct forms of necrotic-like cell death in terms of their morphological features and regulatory mechanisms. These two types of cell death can coexist in disease and contribute to pathological processes. Inhibition of both necroptosis and ferroptosis has been shown to enhance therapeutic effects in treating complex necrosis-related diseases. However, targeting both necroptosis and ferroptosis by a single compound can be challenging, as these two forms of cell death involve distinct molecular pathways. In this study, we discovered that KW-2449, a previously described necroptosis inhibitor, also prevented ferroptosis both in vitro and in vivo. Mechanistically, KW-2449 inhibited ferroptosis by targeting the autophagy pathway. We further identified that KW-2449 functioned as a ULK1 (Unc-51-like kinase 1) inhibitor to block ULK1 kinase activity in autophagy. Remarkably, we found that Necrostatin-1, a classic necroptosis inhibitor that has been shown to prevent ferroptosis, also targets the autophagy pathway to suppress ferroptosis. This study provides the first understanding of how necroptosis inhibitors can prevent ferroptosis and suggests that autophagy is a targetable pathway for necroptosis inhibitors to prevent ferroptosis. Therefore, the identification and design of pharmaceutical molecules that target the autophagy pathway from necroptosis inhibitors is a promising strategy to develop dual inhibitors of necroptosis and ferroptosis in clinical application.
Collapse
Affiliation(s)
- Yaxing Zhao
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingsong Wang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Jing Zhu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Jin Cai
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Xiaona Feng
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qianqian Song
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Hui Jiang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Wenqing Ren
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan He
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Du Feng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jianqiang Yu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yue Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qihui Wu
- Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Siriporn Jitkaew
- Center of Excellence for Cancer and Inflammation, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Zhenyu Cai
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, Yinchuan, China.
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
34
|
Wang N, Huang Z, Guan F, Wang J, Chen Y, Wang H, Jin L, Wang Y. HIF-1α induced FGF18 alleviates renal ischemia/reperfusion injury via YAP. FASEB J 2024; 38:e70092. [PMID: 39373977 DOI: 10.1096/fj.202401238r] [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: 06/04/2024] [Revised: 09/03/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
Acute kidney injury (AKI) is a devastating clinical condition characterized by an abrupt loss of renal function. The pathophysiology of AKI involves diverse processes and elements, of which survival and regeneration have been established to be significant hallmarks. And early studies have confirmed the fundamental role of FGFs in the regulation of AKI pathology, although the association between FGF18 and AKI still remains elusive. Our study demonstrates a substantial up-regulation of FGF18 in the renal tubules of mice subjected to ischemia. Notably, targeted overexpression of FGF18 effectively mitigates the impairment of kidney function induced by AKI. Mechanistically, FGF18 facilitates cell proliferation and anti-apoptosis in RTECs by enhancing the expression of YAP and facilitating its translocation to the nucleus. Aside from that, we also discovered that the substantial expression of FGF18 under ischemic conditions is HIF-1α dependent. This study aims to uncover the inherent mechanism behind the beneficial effects of FGF18 in attenuating AKI. By doing so, it aims to offer novel insights into the development of therapeutic strategies for AKI.
Collapse
Affiliation(s)
- Nan Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
- Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Zhiyuan Huang
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Fangqian Guan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Jiaqi Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yinyun Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Hong Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Yang Wang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Ru Q, Li Y, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in human diseases: mechanisms and therapeutic prospects. Signal Transduct Target Ther 2024; 9:271. [PMID: 39396974 PMCID: PMC11486532 DOI: 10.1038/s41392-024-01969-z] [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: 03/27/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 10/15/2024] Open
Abstract
Iron, an essential mineral in the body, is involved in numerous physiological processes, making the maintenance of iron homeostasis crucial for overall health. Both iron overload and deficiency can cause various disorders and human diseases. Ferroptosis, a form of cell death dependent on iron, is characterized by the extensive peroxidation of lipids. Unlike other kinds of classical unprogrammed cell death, ferroptosis is primarily linked to disruptions in iron metabolism, lipid peroxidation, and antioxidant system imbalance. Ferroptosis is regulated through transcription, translation, and post-translational modifications, which affect cellular sensitivity to ferroptosis. Over the past decade or so, numerous diseases have been linked to ferroptosis as part of their etiology, including cancers, metabolic disorders, autoimmune diseases, central nervous system diseases, cardiovascular diseases, and musculoskeletal diseases. Ferroptosis-related proteins have become attractive targets for many major human diseases that are currently incurable, and some ferroptosis regulators have shown therapeutic effects in clinical trials although further validation of their clinical potential is needed. Therefore, in-depth analysis of ferroptosis and its potential molecular mechanisms in human diseases may offer additional strategies for clinical prevention and treatment. In this review, we discuss the physiological significance of iron homeostasis in the body, the potential contribution of ferroptosis to the etiology and development of human diseases, along with the evidence supporting targeting ferroptosis as a therapeutic approach. Importantly, we evaluate recent potential therapeutic targets and promising interventions, providing guidance for future targeted treatment therapies against human diseases.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
36
|
Qin W, Huang J, Zhang M, Xu M, He J, Liu Q. Nanotechnology-Based Drug Delivery Systems for Treating Acute Kidney Injury. ACS Biomater Sci Eng 2024; 10:6078-6096. [PMID: 39226188 PMCID: PMC11480945 DOI: 10.1021/acsbiomaterials.4c01385] [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: 07/25/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Acute kidney injury (AKI) is a disease that is characterized by a rapid decline in renal function and has a relatively high incidence in hospitalized patients. Sepsis, renal hypoperfusion, and nephrotoxic drug exposure are the main causes of AKI. The major therapy measures currently include supportive treatment, symptomatic treatment, and kidney transplantation. These methods are supportive treatments, and their results are not satisfactory. Fortunately, many new treatments that markedly improve the AKI therapy efficiency are emerging. These include antioxidant therapy, ferroptosis therapy, anti-inflammatory therapy, autophagy therapy, and antiapoptotic therapy. In addition, the development of nanotechnology has further promoted therapeutic effects on AKI. In this review, we highlight recent advances in the development of nanocarriers for AKI drug delivery. Emphasis has been placed on the latest developments in nanocarrier modification and design. We also summarize the applications of different nanocarriers in AKI treatment. Finally, the advantages and challenges of nanocarrier applications in AKI are summarized, and several nanomedicines that have been approved for clinical trials to treat diverse kidney diseases are listed.
Collapse
Affiliation(s)
- Wanbing Qin
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Jiaqi Huang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Manting Zhang
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Mingwei Xu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Junbing He
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
| | - Qinghua Liu
- Jieyang Medical
Research Center, Jieyang People’s
Hospital, Jieyang, 522000 Guangdong, China
- Department
of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080 Guangdong, China
- NHC Key
Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong
Provincial Key Laboratory of Nephrology, Guangzhou, 510080 Guangdong, China
| |
Collapse
|
37
|
Zhu J, Xiang X, Shi L, Song Z, Dong Z. Identification of Differentially Expressed Genes in Cold Storage-associated Kidney Transplantation. Transplantation 2024; 108:2057-2071. [PMID: 38632678 PMCID: PMC11424274 DOI: 10.1097/tp.0000000000005016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
BACKGROUND Although it is acknowledged that ischemia-reperfusion injury is the primary pathology of cold storage-associated kidney transplantation, its underlying mechanism is not well elucidated. METHODS To extend the understanding of molecular events and mine hub genes posttransplantation, we performed bulk RNA sequencing at different time points (24 h, day 7, and day 14) on a murine kidney transplantation model with prolonged cold storage (10 h). RESULTS In the present study, we showed that genes related to the regulation of apoptotic process, DNA damage response, cell cycle/proliferation, and inflammatory response were steadily elevated at 24 h and day 7. The upregulated gene profiling delicately transformed to extracellular matrix organization and fibrosis at day 14. It is prominent that metabolism-associated genes persistently took the first place among downregulated genes. The gene ontology terms of particular note to enrich are fatty acid oxidation and mitochondria energy metabolism. Correspondingly, the key enzymes of the above processes were the products of hub genes as recognized. Moreover, we highlighted the proximal tubular cell-specific increased genes at 24 h by combining the data with public RNA-Seq performed on proximal tubules. We also focused on ferroptosis-related genes and fatty acid oxidation genes to show profound gene dysregulation in kidney transplantation. CONCLUSIONS The comprehensive characterization of transcriptomic analysis may help provide diagnostic biomarkers and therapeutic targets in kidney transplantation.
Collapse
Affiliation(s)
- Jiefu Zhu
- Department of Urology and Department of Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohong Xiang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Lang Shi
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhixia Song
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People’s Hospital of Yichang, Yichang, Hubei 443000, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States
| |
Collapse
|
38
|
Wang L, Shao Z, Wang N, Liu W, Zhang L, Wang Y, Tan J, Jiao X, Liu L, Yan L, Chen S, Cao H, Shao F. Receptor-interacting protein kinase 1 confers autophagic promotion of gasdermin E-mediated pyroptosis in aristolochic acid-induced acute kidney injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116944. [PMID: 39208575 DOI: 10.1016/j.ecoenv.2024.116944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Aristolochic acid (AA) exposure is a severe public health concern worldwide. AAs damage the kidney with an inevitable acute phase that is similar to acute kidney injury (AKI). Gasdermin E (GSDME) is abundant in the kidney; thus; it-mediated pyroptosis might be essential in connecting cell death and inflammation and promoting AAs-AKI. However, the role and exact mechanism of pyroptosis in AAs-AKI have not been investigated. In this study, aristolochic acid I (AAI) was used to establish AKI models. The expression and translocation results showed GSDME-mediated pyroptosis in AAI-AKI. Knocking down GSDME attenuated AAI-induced cell death and transcription of proinflammatory cytokines. Mechanistic research inhibiting caspase (casp) 3, casp 8, and casp 9 with specific chemical antagonists demonstrated that GSDME was activated by cleaved casp 3. Furthermore, the kinase activity of upstream receptor-interacting protein kinase 1 (RIPK1) was significantly elevated, and inhibiting RIPK1 with specific inhibitors markedly improved AAI-induced cell damage. In addition, the level of autophagy was obviously increased. Pretreatment with a specific autophagic inhibitor (3-methyladenine) or knockdown of autophagic genes (Atg5 or Atg7) evidently reduced the activity of RIPK1 and downstream apoptosis and pyroptosis, thus attenuating AA-induced cell injury, which suggested that RIPK1 was a novel link conferring autophagic promotion of pyroptosis. These findings reveal GSDME-mediated pyroptosis for the first time in AAI-induced AKI, propose its novel role in the transcription of cytokines, and demonstrate that autophagy promotes pyroptosis via the RIPK1-dependent apoptotic pathway. This study promotes the understanding of the toxic effects and exact mechanisms of AAs. This will contribute to evaluating the environmental risk of AA exposure and might provide potential therapeutic targets for AA-AKI.
Collapse
Affiliation(s)
- Limeng Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Zehua Shao
- Children's Heart Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450053, China
| | - Ning Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Wenna Liu
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China; Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 451464, China
| | - Lina Zhang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Yanliang Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Jing Tan
- Department of Internal Medicine, Henan Medical College, Longhu Town, Zhengzhou, Henan 451191, China
| | - Xiaojing Jiao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Lu Liu
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Lei Yan
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Song Chen
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450053, China.
| | - Huixia Cao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China.
| | - Fengmin Shao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China.
| |
Collapse
|
39
|
Qin J, Li Z, Feng Y, Guo Y, Zhao Z, Sun S, Zheng J, Zhang M, Zhang J, Zhang Y, Wei J, Ding C, Xue W. Reactive Oxygen Species-Scavenging Mesoporous Poly(tannic acid) Nanospheres Alleviate Acute Kidney Injury by Inhibiting Ferroptosis. ACS Biomater Sci Eng 2024; 10:5856-5868. [PMID: 39164198 PMCID: PMC11389690 DOI: 10.1021/acsbiomaterials.4c00844] [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] [Indexed: 08/22/2024]
Abstract
Acute kidney injury (AKI), predominantly associated with the excess production of endogenous ROS, is a serious renal dysfunction syndrome. Ferroptosis characterized by iron-dependent regulated cell death has significant involvement in AKI pathogenesis. As symptomatic treatment of AKI remains clinically limited, a new class of effective therapies has emerged, which is referred to as nanozyme. In our research, a natural mesoporous poly(tannic acid) nanosphere (referred to as PTA) was developed that can successfully mimic the activity of superoxide dismutase (SOD) by Mussel-inspired interface deposition strategy, for effective ROS scavenging and thus inhibition of ferroptosis to attenuate AKI. As anticipated, PTA mitigated oxidative stress and inhibited ferroptosis, as opposed to other modes of cell death such as pyroptosis or necrosis. Furthermore, PTA exhibited favorable biocompatibility and safeguarded the kidney against ferroptosis by enhancing the expression of SLC7a11/glutathione peroxidase 4(GPX4) and Nrf2/HO-1, while reducing the levels of ACSL4 protein in the ischemia and reperfusion injury (IRI)-induced AKI model. Moreover, PTA effectively suppressed aberrant expression of inflammatory factors. Overall, this study introduced antioxidative nanozymes in the form of mesoporous polyphenol nanospheres, showcasing exceptional therapeutic efficacy in addressing ROS-related diseases. This novel approach holds promise for clinical AKI treatment and broadens the scope of biomedical applications for nanozymes.
Collapse
Affiliation(s)
- Jingyue Qin
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zepeng Li
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Youyou Feng
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Yingcong Guo
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhenting Zhao
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shirui Sun
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jin Zheng
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jing Zhang
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yilei Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University Xi'an ,Shaanxi710061,China
| | - Jing Wei
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Chenguang Ding
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wujun Xue
- Department of Kidney Transplantation, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Institute of Organ Transplantation, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
40
|
Xing Z, Pan L, Yu A, Zhang J, Dong C, Chen J, Xing W, He X, Zhang Z. Value of ultra-high b-value diffusion-weighted imaging for the evaluation of renal ischemia-reperfusion injury. Magn Reson Imaging 2024; 111:1-8. [PMID: 38574980 DOI: 10.1016/j.mri.2024.03.040] [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: 03/03/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
To explore the feasibility of ultra-high b-value diffusion-weighted imaging (ubDWI) in assessment of renal IRI. Thirty-five rabbits were randomized into a control group (n = 7) and a renal IRI group (n = 28). The rabbits in the renal IRI group underwent left renal artery clamping for 60 min. Rabbits underwent axial ubDWI before and at 1, 12, 24, and 48 h after IRI. Apparent diffusion coefficient (ADCst) were calculated from ubDWI with two b-values (b = 0, 1000 s/mm2). Triexponential fits were applied to calculate the pure diffusion coefficients (D), perfusion-related diffusion coefficient (D⁎), and ultra-high ADC (ADCuh). The interobserver reproducibility were evaluated. The repeated measurement analysis of variance and Spearman correlation analysis was used for statistical analysis. The ADCst, D, and ADCuh values showed good reproducibility. The ADCst, D, and D⁎ values of renal Cortex (CO) and outer medulla (OM) significantly decreased after IRI (all P < 0.05). The ADCuh values significantly increased from pre-IRI to 1 h after IRI (P < 0.05) and significantly declined at 24 h and 48 h after IRI (all P < 0.05). ADCuh was strongly positively correlated with AQP-1 in the renal CO and OM (ρ = 0.643, P < 0.001; ρ = 0.662, P < 0.001, respectively). ubDWI can be used to non-invasively evaluate early renal IRI, ADCuh may be adopted to reflect AQP-1 expression.
Collapse
Affiliation(s)
- Zhaoyu Xing
- Department of Urology, Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Liang Pan
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Anding Yu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jinggang Zhang
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Congsong Dong
- Department of Radiology, Affiliated Hospital 6 of Nantong University (Yancheng Third People's Hospital), Yancheng, China
| | - Jie Chen
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wei Xing
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaozhou He
- Department of Urology, Third Affiliated Hospital of Soochow University, Changzhou 213003, China.
| | - Zhiping Zhang
- Department of Radiology, Affiliated Hospital 6 of Nantong University (Yancheng Third People's Hospital), Yancheng, China.
| |
Collapse
|
41
|
Han S, Guo J, Kong C, Li J, Lin F, Zhu J, Wang T, Chen Q, Liu Y, Hu H, Qiu T, Cheng F, Zhou J. ANKRD1 aggravates renal ischaemia‒reperfusion injury via promoting TRIM25-mediated ubiquitination of ACSL3. Clin Transl Med 2024; 14:e70024. [PMID: 39285846 PMCID: PMC11406046 DOI: 10.1002/ctm2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Renal ischaemia‒reperfusion injury (IRI) is the primary cause of acute kidney injury (AKI). To date, effective therapies for delaying renal IRI and postponing patient survival remain absent. Ankyrin repeat domain 1 (ANKRD1) has been implicated in some pathophysiologic processes, but its role in renal IRI has not been explored. METHODS The mouse model of IRI-AKI and in vitro model were utilised to investigate the role of ANKRD1. Immunoprecipitation-mass spectrometry was performed to identify potential ANKRD1-interacting proteins. Protein‒protein interactions and protein ubiquitination were examined using immunoprecipitation and proximity ligation assay and immunoblotting, respectively. Cell viability, damage and lipid peroxidation were evaluated using biochemical and cellular techniques. RESULTS First, we unveiled that ANKRD1 were significantly elevated in renal IRI models. Global knockdown of ANKRD1 in all cell types of mouse kidney by recombinant adeno-associated virus (rAAV9)-mitigated ischaemia/reperfusion-induced renal damage and failure. Silencing ANKRD1 enhanced cell viability and alleviated cell damage in human renal proximal tubule cells exposed to hypoxia reoxygenation or hydrogen peroxide, while ANKRD1 overexpression had the opposite effect. Second, we discovered that ANKRD1's detrimental function during renal IRI involves promoting lipid peroxidation and ferroptosis by directly binding to and decreasing levels of acyl-coenzyme A synthetase long-chain family member 3 (ACSL3), a key protein in lipid metabolism. Furthermore, attenuating ACSL3 in vivo through pharmaceutical approach and in vitro via RNA interference mitigated the anti-ferroptotic effect of ANKRD1 knockdown. Finally, we showed ANKRD1 facilitated post-translational degradation of ACSL3 by modulating E3 ligase tripartite motif containing 25 (TRIM25) to catalyse K63-linked ubiquitination of ACSL3, thereby amplifying lipid peroxidation and ferroptosis, exacerbating renal injury. CONCLUSIONS Our study revealed a previously unknown function of ANKRD1 in renal IRI. By driving ACSL3 ubiquitination and degradation, ANKRD1 aggravates ferroptosis and ultimately exacerbates IRI-AKI, underlining ANKRD1's potential as a therapeutic target for kidney IRI. KEY POINTS/HIGHLIGHTS Ankyrin repeat domain 1 (ANKRD1) is rapidly activated in renal ischaemia‒reperfusion injury (IRI) models in vivo and in vitro. ANKRD1 knockdown mitigates kidney damage and preserves renal function. Ferroptosis contributes to the deteriorating function of ANKRD1 in renal IRI. ANKRD1 promotes acyl-coenzyme A synthetase long-chain family member 3 (ACSL3) degradation via the ubiquitin‒proteasome pathway. The E3 ligase tripartite motif containing 25 (TRIM25) is responsible for ANKRD1-mediated ubiquitination of ACSL3.
Collapse
Affiliation(s)
- Shangting Han
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jiayu Guo
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Chenyang Kong
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jun Li
- Key Laboratory of Medical ElectrophysiologyMinistry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
| | - Fangyou Lin
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jiefu Zhu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Tianyu Wang
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Qi Chen
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yiting Liu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Haochong Hu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Tao Qiu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fan Cheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jiangqiao Zhou
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
42
|
Mishima E. Targeting ferroptosis for treating kidney disease. Clin Exp Nephrol 2024; 28:866-873. [PMID: 38644406 PMCID: PMC11341772 DOI: 10.1007/s10157-024-02491-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/19/2024] [Indexed: 04/23/2024]
Abstract
Ferroptosis is a type of regulated cell death hallmarked by iron-mediated excessive lipid oxidation. Over the past decade since the coining of the term ferroptosis, advances in research have led to the identification of intracellular processes that regulate ferroptosis such as GSH-GPX4 pathway and FSP1-coenzyme Q10/vitamin K pathway. From a disease perspective, the involvement of ferroptosis in pathological conditions including kidney disease has attracted attention. In terms of renal pathophysiology, ferroptosis has been widely investigated for its involvement in ischemia-reperfusion injury, nephrotoxin-induced kidney damage and other renal diseases. Therefore, therapeutic interventions targeting ferroptosis are expected to become a new therapeutic approach for these diseases. However, when considering cell death as a therapeutic target, careful consideration must be given to (i) in which type of cells, (ii) which type of cell death mode, and (iii) in which stage or temporal window of the disease. In the next decade, elucidation of the true involvement of ferroptosis in kidney disease setting in human, and development of clinically applicable and effective therapeutic drugs that target ferroptosis are warranted.
Collapse
Affiliation(s)
- Eikan Mishima
- Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan.
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
43
|
Wang Y, Han J, Zhan S, Guo C, Yin S, Zhan L, Zhou Q, Liu R, Yan H, Wang X, Yan D. Fucoidan alleviates doxorubicin-induced cardiotoxicity by inhibiting ferroptosis via Nrf2/GPX4 pathway. Int J Biol Macromol 2024; 276:133792. [PMID: 38992539 DOI: 10.1016/j.ijbiomac.2024.133792] [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: 02/09/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Doxorubicin (Dox), a chemotherapeutic agent frequently used to treat cancer, elicits cardiotoxicity, a condition referred to as Dox-induced cardiotoxicity (DIC), and ferroptosis plays a contributory role in its pathophysiology. Fucoidan, a polysaccharide with various biological activities and safety profile, has potential therapeutic and pharmaceutical applications. This study aimed to investigate the protective effects and underlying mechanisms of fucoidan in DIC. Echocardiography, biomarkers of cardiomyocyte injury, serum creatine kinase, creatine kinase isoenzyme and lactate dehydrogenase, as well as histological staining results, revealed that fucoidan significantly reduced myocardial damage and improved cardiac function in DIC mice. Transmission electron microscopy; levels of lipid reactive oxygen species, glutathione, and malondialdehyde; ferroptosis-related markers; and regulatory factors such as glutathione peroxidase 4 (GPX4), transferrin receptor protein-1, ferritin heavy chain-1, heme oxygenase-1 in the heart tissue were measured to explore the effect of fucoidan on Dox-induced ferroptosis. These results suggested that fucoidan could inhibit cardiomyocyte ferroptosis caused by Dox. In vitro experiments revealed that silencing nuclear factor-erythroid 2-related factor 2 (Nrf2) in cardiomyocytes reduced the inhibitory effect of fucoidan on ferroptosis. Hence, fucoidan has the potential to ameliorate DIC by inhibiting ferroptosis via the Nrf2/GPX4 pathway.
Collapse
Affiliation(s)
- Yizhi Wang
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Jiawen Han
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Shifang Zhan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Chenyu Guo
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Shuangneng Yin
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Lin Zhan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Qianyi Zhou
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Ruiying Liu
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China
| | - Hua Yan
- Department of Cardiology, Wuhan Asia Heart Hospital, Wuhan University of Science and Technology, Wuhan 430022, Hubei, China
| | - Xiaoyan Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, Hainan, China.
| | - Dan Yan
- Institute of Cardiovascular Diseases, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China; Department of Cardiology, Wuhan Asia Heart Hospital, Wuhan University of Science and Technology, Wuhan 430022, Hubei, China; Institute of Pharmaceutical Innovation, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, Hubei, China.
| |
Collapse
|
44
|
Hui Y, Xia K, Zhong J, Zhang Y, Qiu Q, Chen Z, Wang L, Liu X. SENP1 reduces oxidative stress and apoptosis in renal ischaemia-reperfusion injury by deSUMOylation of HIF-1α. J Cell Mol Med 2024; 28:e70043. [PMID: 39205481 PMCID: PMC11358391 DOI: 10.1111/jcmm.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/23/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Renal ischaemia-reperfusion injury (RIRI) is a primary cause of acute kidney damage, occurring frequently in situations like renal transplantation, yet the underlying mechanisms were not fully understood. Sentrin-specific protease 1 (SENP1) is an important member of the SENP family, which is widely involved in various diseases. However, the role of SENP1 in RIRI has been unclear. In our study, we discovered that SENP1 was involved in RIRI and reduced renal cell apoptosis and oxidative stress at elevated levels. Further mechanistic studies showed that hypoxia-inducible factor-1α (HIF-1α) was identified as a substrate of SENP1. Furthermore, SENP1 deSUMOylated HIF-1α, which reduced the degradation of HIF-1α, and exerted a renoprotective function. In addition, the protective function was lost after application of the HIF-1α specific inhibitor KC7F2. Briefly, our results fully demonstrated that SENP1 reduced the degradation of HIF-1α and attenuated oxidative stress and apoptosis in RIRI by regulating the deSUMOylation of HIF-1α, suggesting that SENP1 may serve as a potential therapeutic target for the treatment of RIRI.
Collapse
Affiliation(s)
- Yumin Hui
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Kang Xia
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jiacheng Zhong
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ye Zhang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Qiangmin Qiu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhiyuan Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Lei Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xiuheng Liu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
45
|
Wang HL, Peng Z, Li YQ, Wang YX, Li JC, Tan RZ, Su HW, Shen HP, Zhao CY, Liu J, Wang L. Calycosin inhibited MIF-mediated inflammatory chemotaxis of macrophages to ameliorate ischemia reperfusion-induced acute kidney injury. Inflamm Res 2024; 73:1267-1282. [PMID: 38844677 DOI: 10.1007/s00011-024-01899-0] [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: 03/03/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Inflammatory macrophage infiltration plays a critical role in acute kidney disease induced by ischemia-reperfusion (IRI-AKI). Calycosin is a natural flavone with multiple bioactivities. This study aimed to investigate the therapeutic role of calycosin in IRI-AKI and its underlying mechanism. METHODS The renoprotective and anti-inflammatory effects of calycosin were analyzed in C57BL/6 mice with IRI-AKI and lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. RNA-seq was used for mechanism investigation. The molecular target of calycosin was screened by in silico methods and validated by surface plasmon resonance (SPR). Macrophage chemotaxis was analyzed using Transwell and agarose gel spot assays. RESULTS Calycosin treatment significantly reduced serum creatinine and urea nitrogen and attenuated tubular destruction in IRI-AKI mice. Additionally, calycosin markedly suppressed NF-κB signaling activation and the expression of inflammatory mediators IL-1β and TNF-α in IRI-AKI kidneys and LPS-stimulated RAW 264.7 cells. Interestingly, RNA-seq revealed calycosin remarkably downregulated chemotaxis-related pathways in RAW 264.7 cells. Among the differentially expressed genes, Ccl2/MCP-1, a critical chemokine mediating macrophage inflammatory chemotaxis, was downregulated in both LPS-stimulated RAW 264.7 cells and IRI-AKI kidneys. Consistently, calycosin treatment attenuated macrophage infiltration in the IRI-AKI kidneys. Importantly, in silico target prediction, molecular docking, and SPR assay demonstrated that calycosin directly binds to macrophage migration inhibitory factor (MIF). Functionally, calycosin abrogated MIF-stimulated NF-κB signaling activation and Ccl2 expression and MIF-mediated chemotaxis in RAW 264.7 cells. CONCLUSIONS In summary, calycosin attenuates IRI-AKI by inhibiting MIF-mediated macrophage inflammatory chemotaxis, suggesting it could be a promising therapeutic agent for the treatment of IRI-AKI.
Collapse
Affiliation(s)
- Hong-Lian Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No. 182, Chunhui Road, District of Longmatan, Luzhou, Sichuan Province, 646000, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 611137, China
| | - Ze Peng
- College of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Yu-Qing Li
- College of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Yi-Xuan Wang
- College of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Jian-Chun Li
- Research Center for Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No. 182, Chunhui Road, District of Longmatan, Luzhou, Sichuan Province, 646000, China
| | - Rui-Zhi Tan
- Research Center for Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No. 182, Chunhui Road, District of Longmatan, Luzhou, Sichuan Province, 646000, China
| | - Hong-Wei Su
- The Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Hong-Ping Shen
- The Clinical Trial Research Center, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Chang-Ying Zhao
- The Department of Endocrinology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Jian Liu
- The Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No. 182, Chunhui Road, District of Longmatan, Luzhou, Sichuan Province, 646000, China.
| | - Li Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, No. 182, Chunhui Road, District of Longmatan, Luzhou, Sichuan Province, 646000, China.
| |
Collapse
|
46
|
Li Y, Cui H, Xu WX, Fu HY, Li JZ, Fan RF. Selenium represses microRNA-202-5p/MICU1 aixs to attenuate mercuric chloride-induced kidney ferroptosis. Poult Sci 2024; 103:103891. [PMID: 38878746 PMCID: PMC11227010 DOI: 10.1016/j.psj.2024.103891] [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: 03/19/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 07/09/2024] Open
Abstract
Mercuric chloride (HgCl2) is a nephrotoxic contaminant that is widely present in the environment. Selenium (Se) can effectively antagonize the biological toxicity caused by heavy metals. Here, in vivo and in vitro models of Se antagonism to HgCl2-induced nephrotoxicity in chickens were established, with the aim of exploring the specific mechanism. Morphological observation and kidney function analysis showed that Se alleviated HgCl2-induced kidney tissue injury and cytotoxicity. The results showed that ferroptosis was the primary mechanism for the toxicity of HgCl2, as indicated by iron overload and lipid peroxidation. On the one hand, Se significantly prevented HgCl2-induced iron overload. On the other hand, Se alleviated the intracellular reactive oxygen species (ROS) levels caused by HgCl2. Subsequently, we focused on the sources of ROS during HgCl2-induced ferroptosis. Mechanically, Se reduced ROS overproduction induced by HgCl2 through mitochondrial calcium uniporter (MCU)/mitochondrial calcium uptake 1 (MICU1)-mediated mitochondrial calcium ion (Ca2+) overload. Furthermore, a dual luciferase reporter assay demonstrated that MICU1 was the direct target of miR-202-5p. Overall, Se represses miR-202-5p/MICU1 axis to attenuate HgCl2-induced kidney ferroptosis.
Collapse
Affiliation(s)
- Yue Li
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Han Cui
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Wan-Xue Xu
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Hong-Yu Fu
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Jiu-Zhi Li
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China
| | - Rui-Feng Fan
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, 271018, China.
| |
Collapse
|
47
|
Zhang F, Ma G, Chaung W, Jacob A, Brenner M, Wang P. TAG-FREE GLYCOSYLATED RHMFG-E8 AS A THERAPY FOR ACUTE KIDNEY INJURY. Shock 2024; 62:286-293. [PMID: 38691106 DOI: 10.1097/shk.0000000000002382] [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: 05/03/2024]
Abstract
ABSTRACT Background: Acute kidney injury (AKI) can result from renal ischemia and reperfusion (I/R) and often occurs during surgical procedures in cardiac, liver, kidney transplantation, and trauma-hemorrhage. Milk fat globule epidermal growth factor-factor VIII (MFG-E8) functions as a bridging molecule to promote the removal of dying cells by professional phagocytes. Because MFG-E8 promotes clearance of apoptotic cells, we have explored its therapeutic potential in various organ injury conditions. To develop human MFG-E8 as a potential therapy, we have generated a human cell-expressed, and thus glycosylated, tag-free recombinant human (rh) MFG-E8 and tested its safety and biological activity in vitro . We hypothesize that the tag-free glycosylated rhMFG-E8 is protective in I/R-induced AKI and it can be developed as an effective therapy for AKI. Methods: To assess the pharmacokinetic properties of the tag-free rhMFG-E8, Sprague-Dawley rats were either untreated or treated with a bolus dose of the tag-free rhMFG-E8, blood collected at various time points and the recovery of human MFG-E8 in the blood were measured by ELISA. Adult male C57BL6 mice underwent bilateral renal ischemia for 30 min, and immediately upon reperfusion, mice were treated intraperitoneally with either normal saline (vehicle) or 20 μg/kg human cell expressed, glycosylated tag-free rhMFG-E8. At either 24 h or 48 h after I/R, blood and kidneys were harvested for further analysis. In separate cohorts of mice after I/R and treatment, mice were observed for 10 days, and survival recorded. Results: AKI rats treated with the tag-free rhMFG-E8 had similar half-life as those in the treated control rats. At 48 h after I/R-induced AKI, renal function markers, blood urea nitrogen, and creatinine were increased and treatment with the tag-free rhMFG-E8 significantly decreased these markers. At both 24 h and 48 h after AKI, inflammatory cytokines, TNF-α, IL-6, and IL-1β were increased and treatment decreased these levels. The kidney mRNA expressions of these cytokines were also increased at 24 h after AKI and treatment significantly decreased those mRNA expressions. Histologically, at 48 h after AKI, tubular damage, and the number of TUNEL staining cells were increased and treatment markedly decreased these measurements. Administration of tag-free rhMFG-E8 at the time of reperfusion improved survival in a 10-day survival study. Conclusion: Our new human cell-expressed tag-free rhMFG-E8 is protective in I/R-induced AKI and it may have the potential to be further developed as a safe and effective therapy for AKI.
Collapse
|
48
|
Xiong W, Tang J, Yu H, Luo Y, Yu M, Li Y. Emodin inhibits M1 macrophage activation that related to acute and chronic kidney injury through EGFR/MAPK pathway. Funct Integr Genomics 2024; 24:131. [PMID: 39078513 DOI: 10.1007/s10142-024-01407-x] [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: 04/23/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Macrophages are the main inflammatory cells involved in kidney injury and play a significant role in the development of acute kidney injury (AKI) and progression of chronic kidney disease (CKD). Emodin is believed to stabilize macrophage homeostasis under pathological conditions. The objective of this study aimed to explore the underlying mechanisms and effects of Emodin on M1 macrophages. METHODS Network pharmacology methods were used to predict target proteins associated with renal injury and identify the pathways affected by emodin. RAW264.7 macrophages were induced into M1 polarization using LPS and then treated with emodin at 20, 40, and 80 µM. The effects of emodin on cell viability, cytokines (IL-1β, IL-6, TNF-α), M1 macrophage markers (F4/80 + CD86+), and the EGFR/MAPK pathway were evaluated. Additionally, we transfected RAW264.7 cells with an EGFR shRNA interference lentivirus to assess its effects on RAW264.7 cells function and MAPK pathway. After RAW264.7 cells were passaged to expanded culture and transfected with EGFR-interfering plasmid, macrophages were induced to polarize towards M1 with LPS and then treated with 80 µM emodin. CKD modeling was performed to test how emodin is regulated during CKD. RESULTS There are 15 common targets between emodin and kidney injury, of which the EGFR/MAPK pathway is the pathway through which emodin affects macrophage function. Emodin significantly reduced the levels of IL-6, IL-1β and TNF-α (p < 0.05) and the ratio of M1 macrophage surface markers F4/80 + CD86+ (p < 0.01) in the supernatant of RAW264.7 cells in a dose-dependent manner. Furthermore, the inhibitory effect of emodin on RAW264.7 cells was achieved by interfering with the EGFR/MAPK pathway. Moreover, emodin also affected the mRNA and protein expression of EGFR and Ras, leading to a decrease in the rate of M1 macrophages, thus inhibiting the pro-inflammatory effect of M1 macrophages. The addition of emodin reduced the rate of M1 macrophages in CKD and inhibited the further polarization of M1 macrophages, thus maintaining the pro-inflammatory and anti-inflammatory homeostasis in CKD, and these effects were achieved by emodin through the control of the EGRF/ERK pathway. CONCLUSION Emodin attenuates M1 macrophage polarization and pro-inflammatory responses via the EGFR/MAPK signalling pathway. And the addition of emodin maintains pro- and anti-inflammatory homeostasis, which is important for maintaining organ function and tissue repair.
Collapse
Affiliation(s)
- Weijian Xiong
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Jing Tang
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Hangxing Yu
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Yan Luo
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Minghuan Yu
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China
| | - Ying Li
- Department of Nephrology, Chongqing Traditional Chinese Medicine Hospital, No.6 Panxi Road, Jiangbei District, Chongqing, 400021, China.
| |
Collapse
|
49
|
Wang J, Wang J, Lu C, Wang Y, Bi H, Zheng J, Ding X. ISL1-overexpressing BMSCs attenuate renal ischemia-reperfusion injury by suppressing apoptosis and oxidative stress through the paracrine action. Cell Mol Life Sci 2024; 81:312. [PMID: 39066917 PMCID: PMC11335236 DOI: 10.1007/s00018-024-05354-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/29/2024] [Accepted: 07/07/2024] [Indexed: 07/30/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major event in renal transplantation, leading to adverse outcomes. Bone marrow mesenchymal stem cells (BMSCs) are novel promising therapeutics for repairing kidney injuries. The therapeutic efficacy of BMSCs with ISL1 overexpression in renal IRI and its underlying mechanism need to be investigated. The unilateral renal IRI rat model was established to mimic clinical acute kidney injury. Rats were injected with PBS, BMSCs-Scrambled or BMSCs-ISL1 via the tail vein at the timepoint of reperfusion, and then sacrificed after 24 h of reperfusion. The administration of BMSCs-ISL1 significantly improved renal function, inhibited tubular cells apoptosis, inflammation, oxidative stress in rats. In vitro, HKC cells subjected to H2O2 stimulation were pretreated with the conditioned medium (CM) of BMSCs-Scrambled or BMSCs-ISL1. The pretreatment of ISL1-CM attenuated apoptosis and oxidative stress induced by H2O2 in HKC cells. Our proteomic data suggested that haptoglobin (Hp) was one of the secretory proteins in ISL1-CM. Subsequent experiments confirmed that Hp was the important paracrine factor from BMSCs-ISL1 that exerted anti-apoptotic and antioxidant functions. Mechanistically, Hp played a cytoprotective role via the inhibition of ERK signaling pathway, which could be abrogated by Ro 67-7476, the ERK phosphorylation agonist. The results suggested that paracrine action may be the main mechanism for BMSCs-ISL1 to exert protective effects. As an important anti-apoptotic and antioxidant factor in ISL1-CM, Hp may serve as a new therapeutic agent for treating IRI, providing new insights for overcoming the long-term adverse effects of stem cell therapy.
Collapse
Affiliation(s)
- Jiale Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Jingwen Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Cuinan Lu
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Ying Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Huanjing Bi
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Jin Zheng
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
50
|
Ye Z, Zhang J, Xu Z, Li Z, Huang G, Tong B, Xia P, Shen Y, Hu H, Yu P, Xi X. Pioglitazone ameliorates ischemia/reperfusion-induced acute kidney injury via oxidative stress attenuation and NLRP3 inflammasome. Hum Cell 2024; 37:959-971. [PMID: 38607518 DOI: 10.1007/s13577-024-01059-w] [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: 07/25/2023] [Accepted: 03/27/2024] [Indexed: 04/13/2024]
Abstract
Acute kidney injury (AKI) induced by renal ischemia/reperfusion injury (IRI) is a severe clinical condition. ROS accumulation, antioxidant pathways deficiency, and inflammation are involved in IRI. Pioglitazone (Pio) exerts anti-inflammatory and antioxidant effects. The aim of this study was to explore the protective effects of pioglitazone against IRI-induced AKI. Pathogen-free Sprague-Dawley (SD) rats were arbitrarily divided into four groups: Sham operation group Control (CON) group, CON + Pio group, I/R + Saline group, and I/R + Pio group. In addition, HK-2 cells were subjected to hypoxia and reoxygenation to develop an H/R model for investigation of the protective mechanism of Pio. Pretreatment with pioglitazone in the model rats reduced urea nitrogen and creatinine levels, histopathological scores, and cytotoxicity after IRI. Pioglitazone treatment significantly attenuated renal cell apoptosis, decreased cytotoxicity, increased Bcl-2 expression, and downregulated Bax expression. Besides, the levels of ROS and inflammatory factors, including NLRP3, ASC, pro-IL-1β, pro-caspase-1, cleaved-caspase-1, TNF-α, IL-6, and IL-1β, in I/R rats and H/R cells were normalized by the pioglitazone treatment. Pioglitazone improved IRI-induced AKI by attenuating oxidative stress and NLRP3 inflammasome activation. Therefore, pioglitazone has the potential to serve as a novel agent for renal IRI treatment and prevention.
Collapse
Affiliation(s)
- Zhenfeng Ye
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China
| | - Zhou Xu
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Zhangwang Li
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Gaomin Huang
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China
| | - Bin Tong
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Jiangxi, 330006, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, Nanchang, China.
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Jiangxi, 330006, Nanchang, China.
| | - Xiaoqing Xi
- Department of Urology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1st Minde Road, Jiangxi, 330006, Nanchang, China.
| |
Collapse
|