1
|
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
|
2
|
Liu Y, Yi R, Zhang X, Sun X, Li J, Wang N, Yao X, Zhang C, Deng H, Wang S, Yang G. The mitochondrial dysfunction regulated by JAK2/STAT3 pathway leads to the necroptosis in the renal cells under patulin exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118202. [PMID: 40249973 DOI: 10.1016/j.ecoenv.2025.118202] [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: 11/25/2024] [Revised: 04/09/2025] [Accepted: 04/13/2025] [Indexed: 04/20/2025]
Abstract
Patulin (PAT) is a common mycotoxin widely found in various agricultural products and fruits, which has obvious toxic effects on animals and humans. Some studies have shown that PAT can cause nephrotoxicity, but the exact mechanism remains to be elucidated. In the present study, we investigated PAT-induced nephrotoxicity and the possible molecular mechanisms involved in its action. In vivo, the results showed that PAT affected the integrity of the glomerular basement membrane and peduncles, leading to necroptosis. We further demonstrated that PAT up-regulated the expression of JAK2, STAT3, RIPK1, RIPK3, and MLKL. This observation was also confirmed in MPC-5 cells. In vitro, pretreatment with Nec-1 (a specific inhibitor of necroptosis) or si-STAT3 resulted in a significant reduction in necroptosis and improved mitochondrial dysfunction. Notably, the pharmacological protection of mitochondrial function by SS-31 significantly attenuated the onset of PAT-induced necroptosis. Taken together, our study suggested that STAT3 activation, and mitochondrial dysfunction played critical roles in PAT-induced necroptosis in the kidney. These findings revealed the mechanisms by which PAT triggered necroptosis, potentially providing a new therapeutic strategy for PAT poisoning.
Collapse
Affiliation(s)
- Yun Liu
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Ruhan Yi
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Xu Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Xiance Sun
- Department of Occupational & Environmental Health, Dalian Medical University, Dalian 116044, China
| | - Jing Li
- Department of Pathology, Dalian Medical University, Dalian 116044, China
| | - Ningning Wang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Xiaofeng Yao
- Department of Occupational & Environmental Health, Dalian Medical University, Dalian 116044, China
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Haoyuan Deng
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China
| | - Shaopeng Wang
- Department of Cardiology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Guang Yang
- Department of Food Nutrition and Safety, Dalian Medical University, No. 9W. Lushun South Road, Dalian 116044, China.
| |
Collapse
|
3
|
Taha M, Abdelbagi O, Baokbah TAS, Bagadood RM, Jalal NA, Obaid R, Al-Hazmi NE, Qusty NF. Insights into the protective effect of omega-3 nanoemulsion against colistin-induced nephrotoxicity in experimental rats: regulation of autophagy and necroptosis via AMPK/mTOR and RIPK1/RIPK3/MLKL signaling pathways. Ren Fail 2024; 46:2429686. [PMID: 39584420 PMCID: PMC11590192 DOI: 10.1080/0886022x.2024.2429686] [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: 08/17/2024] [Revised: 10/24/2024] [Accepted: 11/10/2024] [Indexed: 11/26/2024] Open
Abstract
Colistin is considered one of the most effective antibiotics against gram-negative bacteria. However, nephrotoxicity is one of the dose-limiting factors in its treatment. This study aimed to evaluate the outcome of omega-3 nanoemulsion against colistin-induced nephrotoxicity and its possible underlying mechanism. Four rat groups were involved in the present research; each group containing ten rats was divided as follows: Group I (control) rats received normal saline; Group II (omega-3 nanoemulsion) rats received a dose of 500 mg/kg/body weight orally; Group III (colistin) rats received colistin intraperitoneally (300.000 IU/kg/day); and Group IV (colistin/omega-3 nanoemulsion) rats were treated for six days. The results revealed that colistin administration caused deterioration in renal functions such as creatinine, blood urea nitrogen, 24 h proteinuria, and kidney injury molecule-1 with decrease in creatinine clearance, resulting in histological alternation and tubular damage with diffuse interstitial inflammation. Additionally, colistin significantly increased the lipid peroxidation marker malonaldehyde, proinflammatory cytokines tumor necrosis alpha, interleukin-6, interleukin-1 beta. Also, autophagy influx marker microtubule-associated protein light chain 3B, Beclin-1, and necroptotic related proteins, receptor-interacting protein kinase-3 (RIPK-3), RIPK-1, mixed lineage kinase domain-like protein, and autophagy pathway regulatory kinase AMP-activated protein kinase, with a decrease in antioxidant enzymes catalase, superoxide dismutase, and total antioxidant capacity, autophagic marker ubiquitin-binding protein (p62), and regulator Mammalian target of rapamycin. Interestingly, omega-3 nanoemulsion reversed the results above, dramatically improving renal function and histological picture. Thus, omega-3 nanoemulsion provided a notable method for suppressing colistin-induced nephrotoxicity via its antioxidant and anti-inflammatory power, inhibiting pathological autophagy and necroptosis.
Collapse
Affiliation(s)
- Medhat Taha
- Department of Anatomy, Al-Qunfudah Medical College, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Omer Abdelbagi
- Department of Pathology, Qunfudah Faculty of Medicine, Umm-Al-Qura University, Al-Qunfudhah, Saudi Arabia
| | - Tourki A. S. Baokbah
- Department of Medical Emergency Services, College of Health Sciences-AlQunfudah, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia
| | - Rehab M. Bagadood
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al–Qura University, Makkah, Saudi Arabia
| | - Naif A. Jalal
- Department of Microbiology and Parasitology, Faculty of Medicine, Umm Al‐Qura University, Makkah, Saudi Arabia
| | - Rami Obaid
- Department of Medical Genetics, Faculty of Medicine at Al-Qunfudah, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia
| | - Nawal E. Al-Hazmi
- Department of Chemistry, Division of Biology (Microbiology), University College of Qunfudah, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia
| | - Naeem F. Qusty
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al–Qura University, Makkah, Saudi Arabia
| |
Collapse
|
4
|
Noh MR, Padanilam BJ. Cell death induced by acute renal injury: a perspective on the contributions of accidental and programmed cell death. Am J Physiol Renal Physiol 2024; 327:F4-F20. [PMID: 38660714 PMCID: PMC11390133 DOI: 10.1152/ajprenal.00275.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/11/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
The involvement of cell death in acute kidney injury (AKI) is linked to multiple factors including energy depletion, electrolyte imbalance, reactive oxygen species, inflammation, mitochondrial dysfunction, and activation of several cell death pathway components. Since our review in 2003, discussing the relative contributions of apoptosis and necrosis, several other forms of cell death have been identified and are shown to contribute to AKI. Currently, these various forms of cell death can be fundamentally divided into accidental cell death and regulated or programmed cell death based on functional aspects. Several death initiator and effector molecules switch molecules that may act as signaling components triggering either death or protective mechanisms or alternate cell death pathways have been identified as part of the machinery. Intriguingly, several of these cell death pathways share components and signaling pathways suggesting complementary or compensatory functions. Thus, defining the cross talk between distinct cell death pathways and identifying the unique molecular effectors for each type of cell death may be required to develop novel strategies to prevent cell death. Furthermore, depending on the multiple forms of cell death simultaneously induced in different AKI settings, strategies for combination therapies that block multiple cell death pathways need to be developed to completely prevent injury, cell death, and renal function. This review highlights the various cell death pathways, cross talk, and interactions between different cell death modalities in AKI.
Collapse
Affiliation(s)
- Mi Ra Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Babu J Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| |
Collapse
|
5
|
El Latif AA, Zahra AEA, Badr A, Elbialy ZI, Alghamdi AAA, Althobaiti NA, Assar DH, Abouzed TK. The potential role of upregulated PARP-1/RIPK1 expressions in amikacin-induced oxidative damage and nephrotoxicity in Wistar rats. Toxicol Res (Camb) 2023; 12:979-989. [PMID: 37915468 PMCID: PMC10615830 DOI: 10.1093/toxres/tfad091] [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: 02/03/2023] [Revised: 09/03/2023] [Accepted: 09/09/2023] [Indexed: 11/03/2023] Open
Abstract
This study aimed to investigate the gene expression levels associated with nephrotoxic action of amikacin, as well as the post-treatment effect of diuretics on its nephrotoxic effects. Sixty male rats were divided equally into six groups, including the control group receiving saline intra-peritoneally (ip), and the five treated groups including therapeutic and double therapeutic dose groups, injected ip (15 and 30 mg/kg b.wt./day) respectively for seven days, and another two rat groups treated as therapeutic and double therapeutic dose groups then administered the diuretic orally for seven days and the last group received amikacin ip at a rate of 15 mg/kg/day for seven days, then given free access to water without diuretics for another seven days and was kept as a self-recovery group. Amikacin caused kidney injury, which was exacerbated by the double therapeutic dose, as evidenced by abnormal serum renal injury biomarkers, elevated renal MDA levels, inhibition of renal catalase and SOD enzyme activities, with renal degenerative and necrotic changes. Moreover, comet assays also revealed renal DNA damage. Interestingly, amikacin administration markedly elevated expression levels of the PARP-1, RIP1, TNF-α, IL-1β, and iNOS genes as compared to the control group. However, compared to the self-recovery group, post-amikacin diuretic treatment modulates amikacin-induced altered findings and alleviates amikacin nephrotoxic effects more efficiently. Our findings suggested the potential role of PARP-1 and RIPK1 expressions that influence the expression of proinflammatory cytokines such as IL-1β and TNF-α by exaggerating oxidative stress which may contribute to the pathogenesis of amikacin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Amera Abd El Latif
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, El-Gish Street, Kafr El Sheikh 33516, Egypt
| | - Abo Elnasr A Zahra
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, El-Gish Street, Kafr El Sheikh 33516, Egypt
| | - AlShimaa Badr
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, El-Gish Street, Kafr El Sheikh 33516, Egypt
| | - Zizy I Elbialy
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, El-Gish Street, Kafr El Sheikh 33516, Egypt
| | - Abdullah A A Alghamdi
- Department of Biology, Faculty of Science, Albaha University, Kafrelsheikh University, El-Gish Street, Albaha 1988, Kingdom of Saudi Arabia
| | - Norah A Althobaiti
- Biology Department, College of Science and Humanities-Al Quwaiiyah, Shaqra University, Kafrelsheikh University, El-Gish Street, El-Gish Street, Al Quwaiiyah 19257, Kingdom of Saudi Arabia
| | - Doaa H Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, El-Gish Street, Kafr El Sheikh 33516, Egypt
| | - Tarek kamal Abouzed
- Biochemistry Department, Faculty of Veterinary Medicine, Kafrelsheikh University, El-Gish Street, Kafr El Sheikh, 33516, Egypt
| |
Collapse
|
6
|
Yang D, Fan Y, Xiong M, Chen Y, Zhou Y, Liu X, Yuan Y, Wang Q, Zhang Y, Petersen RB, Su H, Yue J, Zhang C, Chen H, Huang K, Zheng L. Loss of renal tubular G9a benefits acute kidney injury by lowering focal lipid accumulation via CES1. EMBO Rep 2023; 24:e56128. [PMID: 37042626 PMCID: PMC10240209 DOI: 10.15252/embr.202256128] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/14/2023] [Accepted: 03/27/2023] [Indexed: 04/13/2023] Open
Abstract
Surgery-induced renal ischemia and reperfusion (I/R) injury and nephrotoxic drugs like cisplatin can cause acute kidney injury (AKI), for which there is no effective therapy. Lipid accumulation is evident following AKI in renal tubules although the mechanisms and pathological effects are unclear. Here, we report that Ehmt2-encoded histone methyltransferase G9a is upregulated in patients and mouse kidneys after AKI. Renal tubular specific knockout of G9a (Ehmt2Ksp ) or pharmacological inhibition of G9a alleviates lipid accumulation associated with AKI. Mechanistically, G9a suppresses transcription of the lipolytic enzyme Ces1; moreover, G9a and farnesoid X receptor (FXR) competitively bind to the same promoter regions of Ces1. Ces1 is consistently observed to be downregulated in the kidney of AKI patients. Pharmacological inhibition of Ces1 increases lipid accumulation, exacerbates renal I/R-injury and eliminates the beneficial effects on AKI observed in Ehmt2Ksp mice. Furthermore, lipid-lowering atorvastatin and an FXR agonist alleviate AKI by activating Ces1 and reducing renal lipid accumulation. Together, our results reveal a G9a/FXR-Ces1 axis that affects the AKI outcome via regulating renal lipid accumulation.
Collapse
Affiliation(s)
- Dong Yang
- School of Pharmacy, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yu Fan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhanChina
| | - Mingrui Xiong
- School of Pharmacy, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuchen Chen
- School of Pharmacy, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yihao Zhou
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhanChina
| | - Xikai Liu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhanChina
| | - Yangmian Yuan
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhanChina
| | - Qing Wang
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhanChina
| | - Yu Zhang
- School of Pharmacy, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Robert B Petersen
- Foundational SciencesCentral Michigan University College of MedicineMt. PleasantMIUSA
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Junqiu Yue
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hong Chen
- School of Pharmacy, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kun Huang
- School of Pharmacy, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhanChina
| |
Collapse
|
7
|
Liu WC, Chiu HW, Chou CL, Chiu YJ, Lee YH. Lactoferrin attenuated urban particulate matter-induced nephrotoxicity by regulating the CSF2/CENPE axis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 318:120871. [PMID: 36528199 DOI: 10.1016/j.envpol.2022.120871] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Several epidemiological studies regarding the adverse effect of air pollution have notably accelerated in recent years. Urban particulate matter (PM) gains access to the respiratory system and translocates into the circulation to affect several tissues, such as the liver and kidneys. Lactoferrin is a substance belonging to the non-heme iron-binding glycoprotein which is present in breast milk and other exocrine fluids. Lactoferrin is protective against many pathophysiological conditions. In the present study, we explored the potential influence of lactoferrin on PM-induced nephrotoxicity. We found that lactoferrin rescued PM-induced cell death but did not affect apoptosis in human kidney cells. Lactoferrin decreased necroptosis and fibrosis but increased autophagy in human kidney cells. Furthermore, the gene expression profiles of PM and lactoferrin were analyzed by RNA sequencing. The transcriptional profiles were uploaded and analyzed by ingenuity pathway analysis software and gene set enrichment analysis. The results showed that the crucial role of the CSF2/CENPE pathway was involved in human kidney cells treated with PM and lactoferrin. In a mouse model, lactoferrin ameliorates PM-induced nephrotoxicity by regulating necroptosis, fibrosis, autophagy and the CSF2/CENPE axis. In summary, these findings showed that lactoferrin could be a novel therapeutic or preventive agent for renal disorders caused by airborne PM pollution.
Collapse
Affiliation(s)
- Wen-Chih Liu
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan; Section of Nephrology, Department of Medicine, Antai Medical Care Corporation Antai Tian-Sheng Memorial Hospital, Pingtung, Taiwan
| | - Hui-Wen Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Chu-Lin Chou
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City, Taiwan; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yu-Jhe Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung, Taiwan.
| |
Collapse
|
8
|
Alassaf N, Attia H. Autophagy and necroptosis in cisplatin-induced acute kidney injury: Recent advances regarding their role and therapeutic potential. Front Pharmacol 2023; 14:1103062. [PMID: 36794281 PMCID: PMC9922871 DOI: 10.3389/fphar.2023.1103062] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Cisplatin (CP) is a broad-spectrum antineoplastic agent, used to treat many different types of malignancies due to its high efficacy and low cost. However, its use is largely limited by acute kidney injury (AKI), which, if left untreated, may progress to cause irreversible chronic renal dysfunction. Despite substantial research, the exact mechanisms of CP-induced AKI are still so far unclear and effective therapies are lacking and desperately needed. In recent years, necroptosis, a novel subtype of regulated necrosis, and autophagy, a form of homeostatic housekeeping mechanism have witnessed a burgeoning interest owing to their potential to regulate and alleviate CP-induced AKI. In this review, we elucidate in detail the molecular mechanisms and potential roles of both autophagy and necroptosis in CP-induced AKI. We also explore the potential of targeting these pathways to overcome CP-induced AKI according to recent advances.
Collapse
Affiliation(s)
- Noha Alassaf
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,*Correspondence: Noha Alassaf,
| | - Hala Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia,Department of Biochemistry, College of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
9
|
Contreras CJ, Mukherjee N, Branco RCS, Lin L, Hogan MF, Cai EP, Oberst AA, Kahn SE, Templin AT. RIPK1 and RIPK3 regulate TNFα-induced β-cell death in concert with caspase activity. Mol Metab 2022; 65:101582. [PMID: 36030035 PMCID: PMC9464965 DOI: 10.1016/j.molmet.2022.101582] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE Type 1 diabetes (T1D) is characterized by autoimmune-associated β-cell loss, insulin insufficiency, and hyperglycemia. Although TNFα signaling is associated with β-cell loss and hyperglycemia in non-obese diabetic mice and human T1D, the molecular mechanisms of β-cell TNF receptor signaling have not been fully characterized. Based on work in other cell types, we hypothesized that receptor interacting protein kinase 1 (RIPK1) and receptor interacting protein kinase 3 (RIPK3) regulate TNFα-induced β-cell death in concert with caspase activity. METHODS We evaluated TNFα-induced cell death, caspase activity, and TNF receptor pathway molecule expression in immortalized NIT-1 and INS-1 β-cell lines and primary mouse islet cells in vitro. Our studies utilized genetic and small molecule approaches to alter RIPK1 and RIPK3 expression and caspase activity to interrogate mechanisms of TNFα-induced β-cell death. We used the β-cell toxin streptozotocin (STZ) to determine the susceptibility of Ripk3+/+ and Ripk3-/- mice to hyperglycemia in vivo. RESULTS Expression of TNF receptor signaling molecules including RIPK1 and RIPK3 was identified in NIT-1 and INS-1 β cells and isolated mouse islets at the mRNA and protein levels. TNFα treatment increased NIT-1 and INS-1 cell death and caspase activity after 24-48 h, and BV6, a small molecule inhibitor of inhibitor of apoptosis proteins (IAPs) amplified this TNFα-induced cell death. RIPK1 deficient NIT-1 cells were protected from TNFα- and BV6-induced cell death and caspase activation. Interestingly, small molecule inhibition of caspases with zVAD-fmk (zVAD) did not prevent TNFα-induced cell death in either NIT-1 or INS-1 cells. This caspase-independent cell death was increased by BV6 treatment and decreased in RIPK1 deficient NIT-1 cells. RIPK3 deficient NIT-1 cells and RIPK3 kinase inhibitor treated INS-1 cells were protected from TNFα+zVAD-induced cell death, whereas RIPK3 overexpression increased INS-1 cell death and promoted RIPK3 and MLKL interaction under TNFα+zVAD treatment. In mouse islet cells, BV6 or zVAD treatment promoted TNFα-induced cell death, and TNFα+zVAD-induced cell death was blocked by RIPK3 inhibition and in Ripk3-/- islet cells in vitro. Ripk3-/- mice were also protected from STZ-induced hyperglycemia and glucose intolerance in vivo. CONCLUSIONS RIPK1 and RIPK3 regulate TNFα-induced β-cell death in concert with caspase activity in immortalized and primary islet β cells. TNF receptor signaling molecules such as RIPK1 and RIPK3 may represent novel therapeutic targets to promote β-cell survival and glucose homeostasis in T1D.
Collapse
Affiliation(s)
- Christopher J Contreras
- Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, IN, USA
| | - Noyonika Mukherjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Renato C S Branco
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Li Lin
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Meghan F Hogan
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA, USA
| | - Erica P Cai
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Andrew A Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA, USA
| | - Andrew T Templin
- Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
10
|
Fan Y, Lu J, Yu Z, Qu X, Guan S. 1,3-Dichloro-2-propanol-Induced Renal Tubular Cell Necroptosis through the ROS/RIPK3/MLKL Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10847-10857. [PMID: 36000575 DOI: 10.1021/acs.jafc.2c02619] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
1,3-Dichloro-2-propanol (1,3-DCP), as a food pollutant, exists in a variety of foods. Studies have shown that it has nephrotoxicity. In the study, we found that 1,3-DCP caused renal injury with necroptosis in C57BL/6J mice. The mechanism of 1,3-DCP-caused nephrotoxicity was further explored in NRK-52E cells in vitro. We found that 1,3-DCP caused cell necroptosis with the increase in lactate dehydrogenase (LDH) levels and the expressions of RIPK3 and MLKL. But pretreatment with a ROS inhibitor N-acetyl-l-cysteine (NAC), a RIPK3 inhibitor GSK'872, or RIPK3 gene silencing alleviated 1,3-DCP-induced cell necroptosis. The data indicated that 1,3-DCP induced necroptosis through the ROS/RIPK3/MLKL pathway in NRK-52E cells. In further mechanistic studies, we explored how 1,3-DCP induced ROS production. We found that 1,3-DCP inhibited the expressions of nuclear and cytoplasmic Nrf2. But pretreatment with an Nrf2 activator dimethyl fumarate (DMF) up-regulated the expressions of nuclear and cytoplasmic Nrf2 and down-regulated ROS levels and RIPK3 and MLKL expressions. We also examined the effects of mitophagy on 1,3-DCP-induced ROS. The data manifested that 1,3-DCP suppressed mitophagy in NRK-52E cells by decreasing LC3-II, Pink1, and Parkin levels, increasing p62 levels, and decreasing colocalization of LC3 and Mito-Tracker Red. Pretreatment with an autophagy activator rapamycin (Rapa) decreased 1,3-DCP-induced ROS. Taken together, our data identified that 1,3-DCP caused renal necroptosis through the ROS/RIPK3/MLKL pathway.
Collapse
Affiliation(s)
- Yong Fan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Jing Lu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Zelin Yu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Xiao Qu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin 130062, People's Republic of China
- Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, People's Republic of China
| |
Collapse
|
11
|
Du Y, Hao H, Ma H, Liu H. Macrophage migration inhibitory factor in acute kidneyinjury. Front Physiol 2022; 13:945827. [PMID: 36117692 PMCID: PMC9478040 DOI: 10.3389/fphys.2022.945827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Acute kidney injury (AKI) is a complex clinical syndrome with multiple etiologies and pathogenesis, which lacks early biomarkers and targeted therapy. Recently, macrophage migration inhibitory factor (MIF) family protein have received increasing attention owing to its pleiotropic protein molecule character in acute kidney injury, where it performed a dual role in the pathological process. macrophage migration inhibitory factor and macrophage migration inhibitory factor-2 are released into the peripheral circulation when Acute kidney injury occurs and interact with various cellular pathways. On the one hand, macrophage migration inhibitory factor exerts a protective effect in anti-oxidation and macrophage migration inhibitory factor-2 promotes cell proliferation and ameliorates renal fibrosis. On the other hand, macrophage migration inhibitory factor aggravates renal injury as an upstream inflammation factor. Herein, we provide an overview on the biological role and possible mechanisms of macrophage migration inhibitory factor and macrophage migration inhibitory factor-2 in the process of Acute kidney injury and the clinical application prospects of macrophage migration inhibitory factor family proteins as a potential therapeutic target.
Collapse
Affiliation(s)
- Yiwei Du
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Hao Hao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
- *Correspondence: Hongbao Liu, ; Heng Ma,
| | - Hongbao Liu
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
- *Correspondence: Hongbao Liu, ; Heng Ma,
| |
Collapse
|
12
|
Hepokoski M, Singh P. Mitochondria as mediators of systemic inflammation and organ cross talk in acute kidney injury. Am J Physiol Renal Physiol 2022; 322:F589-F596. [PMID: 35379000 PMCID: PMC9054254 DOI: 10.1152/ajprenal.00372.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acute kidney injury (AKI) is a systemic inflammatory disease that contributes to remote organ failures. Multiple organ failure is the leading cause of death due to AKI, and lack of understanding of the mechanisms involved has precluded the development of novel therapies. Mitochondrial injury in AKI leads to mitochondrial fragmentation and release of damage-associated molecular patterns, which are known to active innate immune pathways and systemic inflammation. This review presents current evidence suggesting that extracellular mitochondrial damage-associated molecular patterns are mediators of remote organ failures during AKI that have the potential to be modifiable.
Collapse
Affiliation(s)
- Mark Hepokoski
- 1Veterans Affairs San Diego Healthcare System, San Diego, California,2Division of Pulmonary and Critical Care Medicine, University of California, San Diego, California
| | - Prabhleen Singh
- 1Veterans Affairs San Diego Healthcare System, San Diego, California,3Division of Nephrology and Hypertension, University of California, San Diego, California
| |
Collapse
|
13
|
Al-Brakati A, Alsharif KF, Alzahrani KJ, Kabrah S, Al-Amer O, Oyouni AA, Habotta OA, Lokman MS, Bauomy AA, Kassab RB, Abdel Moneim AE. Using Green Biosynthesized Lycopene-Coated Selenium Nanoparticles to Rescue Renal Damage in Glycerol-Induced Acute Kidney Injury in Rats. Int J Nanomedicine 2021; 16:4335-4349. [PMID: 34234429 PMCID: PMC8254550 DOI: 10.2147/ijn.s306186] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/09/2021] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Selenium nanoparticles (SeNPs) have recently gained much attention in nanomedicine applications owing to their unique biological properties. Biosynthesis of SeNPs using nutraceuticals as lycopene (LYC) maximizes their stability and bioactivities. In this context, this study aimed to elucidate the renoprotective activity of SeNPs coated with LYC (LYC-SeNPs) in the acute kidney injury (AKI) model. METHODS Rats were divided into six groups: control, AKI (glycerol-treated), AKI+sodium selenite (Na2SeO3; 0.5 mg/kg), AKI+LYC (10 mg/kg), AKI+LYC-SeNPs (0.5 mg/kg) and treated for 14 days. RESULTS Glycerol treatment evoked significant increases in rhabdomyolysis-related markers (creatine kinase and LDH). Furthermore, relative kidney weight, Kim-1, neutrophil gelatinase-associated lipocalin (NGAL), serum urea, and creatinine in the AKI group were elevated. Glycerol-injected rats displayed declines in reduced glutathione level, and superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase activities, paralleled with downregulations in Nfe2l2 and Hmox-1 expressions and high renal MDA and NO contents. Glycerol-induced renal inflammation was evident by rises in TNF-α, IL-1β, IL-6, and upregulated Nos2 expression. Also, apoptotic (elevated caspase-3, Bax, and cytochrome-c with lowered Bcl-2) and necroptotic (elevated Pipk3 expression) changes were reported in damaged renal tissue. Co-treatment with Na2SeO3, LYC, or LYC-SeNPs restored the biochemical, molecular, and histological alterations in AKI. In comparison with Na2SeO3 or LYC treatment, LYC-SeNPs had the best nephroprotective profile. CONCLUSION Our findings authentically revealed that LYC-SeNPs co-administration could be a prospective candidate against AKI-mediated renal damage via antioxidant, anti-inflammatory, anti-apoptotic and anti-necroptotic activities.
Collapse
Affiliation(s)
- Ashraf Al-Brakati
- Department of Human Anatomy, College of Medicine, Taif University, Taif, 21944, Saudi Arabia
| | - Khalaf F Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia
| | - Khalid J Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia
| | - Saeed Kabrah
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm AlQura University, Makkah, Saudi Arabia
| | - Osama Al-Amer
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Atif Abdulwahab Oyouni
- Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
- Department of Biology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Maha S Lokman
- Biology Department, College of Science and Humanities, Prince Sattam bin Abdul Aziz University, Alkharj, Saudi Arabia
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Amira A Bauomy
- Department of Science Laboratories, College of Science and Arts, Qassim University, ArRass, 52719, Saudi Arabia
| | - Rami B Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
- Department of Biology, Faculty of Science and Arts, Al Baha University, Almakhwah, Al Baha, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
14
|
Yang Q, Zang HM, Xing T, Zhang SF, Li C, Zhang Y, Dong YH, Hu XW, Yu JT, Wen JG, Jin J, Li J, Zhao R, Ma TT, Meng XM. Gypenoside XLIX protects against acute kidney injury by suppressing IGFBP7/IGF1R-mediated programmed cell death and inflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153541. [PMID: 33773190 DOI: 10.1016/j.phymed.2021.153541] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 02/23/2021] [Accepted: 03/04/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Acute kidney injury (AKI), characterised by excessive inflammatory cell recruitment and programmed cell death, has a high morbidity and mortality; however, effective and specific therapies for AKI are still lacking. OBJECTIVE This study aimed to evaluate the renoprotective effects of gypenoside XLIX (Gyp XLIX) in AKI. METHODS The protective effects of Gyp XLIX were tested in two AKI mouse models established using male C57BL/6 mice (aged 6-8 weeks) by a single intraperitoneal injection of cisplatin (20 mg/kg) or renal ischemia-reperfusion for 40 min. Gyp XLIX was administered intraperitoneally before cisplatin administration or renal ischemia-reperfusion. Renal function, tubular injury, renal inflammation and programmed cell death were evaluated. In addition, the renoprotective effects of Gyp XLIX were also evaluated in cisplatin- or hypoxia-treated tubular epithelial cells. The mechanisms underlying these effects were then explored using RNA sequencing. RESULTS In vivo, Gyp XLIX substantially suppressed the increase in serum creatinine and blood urea nitrogen levels. Moreover, tubular damage was alleviated by Gyp XLIX as shown by periodic acid-Schiff staining, electron microscopy and molecular analysis of KIM-1. Consistently, we found that Gyp XLIX suppressed renal necroptosis though the RIPK1/RIPK3/MLKL pathway. The anti-inflammatory and antinecroptotic effects were further confirmed in vitro. Mechanistically, RNA sequencing showed that Gyp XLIX markedly suppressed the levels of IGF binding protein 7 (IGFBP7). Co-immunoprecipitation and western blot analysis further showed that Gyp XLIX reduced the binding of IGFBP7 to IGF1 receptor (IGF1R). Additionally, picropodophyllin, an inhibitor of IGF1R, abrogated the therapeutic effects of Gyp XLIX on cisplatin-induced renal cell injury; this finding indicated that Gyp XLIX may function by activating IGF1R-mediated downstream signalling Additionally, we also detected the metabolic distribution of Gyp XLIX after injection; Gyp XLIX had a high concentration in the kidney and exhibited a long retention time. These findings may shed light on the application of Gyp XLIX for AKI treatment clinically. CONCLUSION Gyp XLIX may serve as a potential therapeutic agent for AKI treatment via IGFBP7/ IGF1R-dependent mechanisms.
Collapse
Affiliation(s)
- Qin Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Hong-Mei Zang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Tian Xing
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Shao-Fei Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China; School of Life Sciences, Huaibei Normal University, 100 Dongshan Road, Huaibei 235000, Anhui Province, China
| | - Chao Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yao Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xiao-Wei Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Ju-Tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Juan Jin
- Department of Pharmacology, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Ren Zhao
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Tao-Tao Ma
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
15
|
Zhang S, Sun H, Kong W, Zhang B. Functional role of microRNA-500a-3P-loaded liposomes in the treatment of cisplatin-induced AKI. IET Nanobiotechnol 2021; 14:465-469. [PMID: 32755955 DOI: 10.1049/iet-nbt.2019.0247] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cisplatin treatment results in acute kidney injury (AKI) by the phosphorylation of mixed lineage kinase domain-like protein (MLKL). The knockout of MLKL, which is a principle mediator of necroptosis, is believed to alleviate the AKI symptoms. The present study was aimed to improve the therapeutic efficacy in AKI. For this purpose, miR-500a-3P was identified as appropriate miRNA therapeutics and loaded in liposome delivery carrier. The authors have showed that the miR-LIP directly controls the expression of RIPK3 and MLKL - a modulator of necroptosis and thereby reduces the severity of kidney injury. The miR-LIP significantly controlled the phosphorylation of MLKL compared to that of CDDP-treated HK2 cells. Similar results are observed with RIPK3. The miR-LIP has also been demonstrated to control the inflammatory response in tubular cells. Western blot analysis further revealed that the phosphorylation of P-65 was mainly responsible for the inflammatory response and miR-LIP significantly decreased the CDDP-induced NF-kB phosphorylation. Overall, the present study explored the molecular mechanism behind the necroptosis in AKI and potential of miRNA in targeting MLKL pathways. Study further highlights the potential advantage of liposome as a delivery carrier for miRNA therapeutics.
Collapse
Affiliation(s)
- Suhua Zhang
- Department of Kidney Disease, Suzhou Kowloon Hospital Affiliated to Medical College of Shanghai Jiaotong University, Suzhou, Jiangsu 215028, People's Republic of China
| | - Huaixin Sun
- Department of Kidney Disease, Suzhou Kowloon Hospital Affiliated to Medical College of Shanghai Jiaotong University, Suzhou, Jiangsu 215028, People's Republic of China
| | - Weixin Kong
- Department of Kidney Disease, Suzhou Kowloon Hospital Affiliated to Medical College of Shanghai Jiaotong University, Suzhou, Jiangsu 215028, People's Republic of China
| | - Bo Zhang
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China.
| |
Collapse
|
16
|
Grivei A, Giuliani KTK, Wang X, Ungerer J, Francis L, Hepburn K, John GT, Gois PFH, Kassianos AJ, Healy H. Oxidative stress and inflammasome activation in human rhabdomyolysis-induced acute kidney injury. Free Radic Biol Med 2020; 160:690-695. [PMID: 32942024 DOI: 10.1016/j.freeradbiomed.2020.09.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Acute kidney injury (AKI) is a life-threatening complication of rhabdomyolysis. The pathophysiological mechanisms of rhabdomyolysis-induced AKI (RIAKI) have been extensively studied in the murine system, yet clinical translation of this knowledge to humans is lacking. In this study, we investigated the cellular and molecular pathways of human RIAKI. Renal biopsy tissue from a RIAKI patient was examined by quantitative immunohistochemistry (Q-IHC) and compared to healthy kidney cortical tissue. We identified myoglobin casts and uric acid localised to sites of histological tubular injury, consistent with the diagnosis of RIAKI. These pathological features were associated with tubular oxidative stress (4-hydroxynonenal staining), regulated necrosis/necroptosis (phosphorylated mixed-lineage kinase domain-like protein staining) and inflammation (tumour necrosis factor (TNF)-α staining). Expression of these markers was significantly elevated in the RIAKI tissue compared to the healthy control. A tubulointerstitial inflammatory infiltrate accumulated adjacent to these sites of RIAKI oxidative injury, consisting of macrophages (CD68), dendritic cells (CD1c) and T lymphocytes (CD3). Foci of inflammasome activation were co-localised with these immune cell infiltrate, with significantly increased staining for adaptor protein ASC (apoptosis-associated speck-like protein containing a caspase activation and recruitment domain) and active caspase-1 in the RIAKI tissue compared to the healthy control. Our clinical findings identify multiple pathophysiological pathways previously only reported in murine RIAKI, providing first evidence in humans linking deposition of myoglobin and presence of uric acid to tubular oxidative stress/necroptosis, inflammasome activation and necroinflammation.
Collapse
Affiliation(s)
- Anca Grivei
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia
| | - Kurt T K Giuliani
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Xiangju Wang
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia
| | - Jacobus Ungerer
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Leo Francis
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia
| | - Kirsten Hepburn
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - George T John
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Pedro F H Gois
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Andrew J Kassianos
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia; Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Helen Healy
- Kidney Health Service, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia; Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
17
|
Huang H, Jin WW, Huang M, Ji H, Capen DE, Xia Y, Yuan J, Păunescu TG, Lu HAJ. Gentamicin-Induced Acute Kidney Injury in an Animal Model Involves Programmed Necrosis of the Collecting Duct. J Am Soc Nephrol 2020; 31:2097-2115. [PMID: 32641397 DOI: 10.1681/asn.2019020204] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/11/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Gentamicin is a potent aminoglycoside antibiotic that targets gram-negative bacteria, but nephrotoxicity limits its clinical application. The cause of gentamicin-induced AKI has been attributed mainly to apoptosis of the proximal tubule cells. However, blocking apoptosis only partially attenuates gentamicin-induced AKI in animals. METHODS Mice treated with gentamicin for 7 days developed AKI, and programmed cell death pathways were examined using pharmacologic inhibitors and in RIPK3-deficient mice. Effects in porcine and murine kidney cell lines were also examined. RESULTS Gentamicin caused a low level of apoptosis in the proximal tubules and significant ultrastructural alterations consistent with necroptosis, occurring predominantly in the collecting ducts (CDs), including cell and organelle swelling and rupture of the cell membrane. Upregulation of the key necroptotic signaling molecules, mixed lineage kinase domain-like pseudokinase (MLKL) and receptor-interacting serine/threonine-protein kinase 3 (RIPK3), was detected in gentamicin-treated mice and in cultured renal tubule cells. In addition, gentamicin induced apical accumulation of total and phosphorylated MLKL (pMLKL) in CDs in mouse kidney. Inhibiting a necroptotic protein, RIPK1, with necrostatin-1 (Nec-1), attenuated gentamicin-induced necrosis and upregulation of MLKL and RIPK3 in mice and cultured cells. Nec-1 also alleviated kidney inflammation and fibrosis, and significantly improved gentamicin-induced renal dysfunction in mice. Furthermore, deletion of RIPK3 in the Ripk3 -/- mice significantly attenuated gentamicin-induced AKI. CONCLUSIONS A previously unrecognized role of programmed necrosis in collecting ducts in gentamicin-induced kidney injury presents a potential new therapeutic strategy to alleviate gentamicin-induced AKI through inhibiting necroptosis.
Collapse
Affiliation(s)
- Huihui Huang
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - William W Jin
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Ming Huang
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Heyu Ji
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Diane E Capen
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Yin Xia
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - Teodor G Păunescu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology and Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts .,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Ishii S, Yamada M, Koibuchi N. Chicken ovalbumin upstream promoter-transcription factor II protects against cisplatin-induced acute kidney injury. Endocr J 2020; 67:283-293. [PMID: 31801919 DOI: 10.1507/endocrj.ej19-0459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) plays essential roles in organogenesis of embryos. Recently COUP-TFII is also implicated in several diseases in adults. Here we focus on the role of COUP-TFII in cisplatin-induced acute kidney injury (AKI). COUP-TFII was the most abundantly expressed in the kidney among organs. Male tamoxifen-inducible COUP-TFII-knockout mice or control mice were intraperitoneally treated with 30 mg/kg body weight of cisplatin at 12 weeks old to induce AKI. The kidney samples were subject to morphological studies, terminal deoxynucleotidyl transferase-mediated deoxyuridine nick-end labeling (TUNEL) assay, immunohistochemistry and RT-qPCR. Serum levels of creatinine, blood urea nitrogen (BUN) and tumor necrosis factor alpha (TNF-α) were measured. Administration of cisplatin induced a more severe AKI in adult COUP-TFII-knockout mice. An increase in dead cells in both the proximal tubules and thick ascending limb of Henle's loop (TAL) was observed in the knockout mouse kidney. The expression levels of COUP-TFII decreased in the TAL by cisplatin administration. There was no difference in the expression levels of transporter mRNAs responsible for cellular cisplatin uptake between control and knockout mouse kidney. COUP-TFII-knockout mice and COUP-TFII-depleted cells exhibited an elevation in TNF-α levels, suggesting the involvement of the TNF-α pathway. Chromatin immunoprecipitation showed that COUP-TFII was enriched in the potential binding site, suggesting that COUP-TFII might directly suppress the TNF-α gene at transcriptional level. These results indicate the involvement of COUP-TFII in the pathophysiology of AKI and COUP-TFII may be a potential therapeutic target for AKI.
Collapse
Affiliation(s)
- Sumiyasu Ishii
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Masanobu Yamada
- Department of Internal Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
19
|
Hashish EA, Elgaml SA, El-Fattah A, Shalaby SI, Abdelaziz S. β-Amyrin supplementation ameliorates the toxic effect of glycerol in the kidney of rat model. Hum Exp Toxicol 2020; 39:930-937. [PMID: 32081053 DOI: 10.1177/0960327120907136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute kidney injury (AKI) is a common life-threatening complication. In this study, β-amyrin is hypothesized to exert a potential nephroprotective effect against glycerol-induced nephrotoxicity in rats. Thirty-two female Sprague-Dawley rats were divided into four groups: normal control, β-amyrin treated (50 mg kg-1 body weight) for 14 days, glycerol 25% (10 ml kg-1 BW volume/volume in sterile saline, intramuscular), and β-amyrin + glycerol-treated rats. Assessing kidney function was done through the measurement of serum urea and creatinine (SCr). Real-time quantitative polymerase chain reaction analysis was done to measure the changes in the gap junction protein and intermediate filament proteins (IFPs) messenger RNA (mRNA) levels. Renal tissue histopathology was also observed. Glycerol exhibited significant elevation in the SCr and urea with significant upregulation of connexin43, vimentin, and nestin. The levels of all disrupted parameters were improved by the pre-administration of β-amyrin. The β-amyrin exerts significant improvement of the biochemical parameters with a restoration of the renal tissue histopathological picture. Significant downregulation of the expression levels of the gap junction protein and IFPs mRNA was also seen. Collectively, the administration of β-amyrin showed a promising effect for a protection against glycerol-induced AKI in rats.
Collapse
Affiliation(s)
- E A Hashish
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkyia, Egypt
| | - S A Elgaml
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkyia, Egypt
| | - Aha El-Fattah
- Department of Veterinary Genetics and Genetic Engineering, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkyia, Egypt
| | - S I Shalaby
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Sharkyia, Egypt
| | - S Abdelaziz
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkyia, Egypt
| |
Collapse
|
20
|
Zang H, Yang Q, Li J. Eleutheroside B Protects against Acute Kidney Injury by Activating IGF Pathway. Molecules 2019; 24:E3876. [PMID: 31661774 PMCID: PMC6864713 DOI: 10.3390/molecules24213876] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/22/2019] [Accepted: 10/24/2019] [Indexed: 01/17/2023] Open
Abstract
Acute kidney injury (AKI) is a common, complex, and severe clinical syndrome characterized by rapid decline in renal function, combined with tissue damage. Currently, the prevention and treatment of AKI are focused on symptomatic treatment, rather than treating the underlying causes. Therefore, there is no specific treatment to prevent renal injury except for renal dialysis. In this study, we used cisplatin-induced AKI mouse and human kidney-2 (HK-2) cell models to evaluate the renal protective effect of eleutheroside B, an active compound in traditional Chinese medicines. MTT assay was used to detect the effect of eleutheroside B on proliferation of human HK-2 cells in presence and in absence of cisplatin. Western blot and immunostaining were used to detect the protein level of kidney injury molecule-1 (KIM-1), cleaved caspase-3, receptor-interacting protein kinase (RIPK)-1, and RIPK-3. Real-time PCR was used to detect the mRNA levels of chemokines (like monocyte chemotactic protein 1, MCP-1) and pro-inflammatory cytokines including interleukin-6 (IL-6) and tumor necrosis factor (TNF-α). Flow cytometry assay was used to detect apoptosis of HK-2 cells. In vivo results showed that eleutheroside B reduced the increase in serum creatinine and blood urea nitrogen (BUN) levels in the AKI model. Periodic acid-Schiff staining and Western blot analysis of KIM-1 showed that eleutheroside B alleviated tubular cell injury. Further, eleutheroside B reduced macrophage infiltration and production of inflammatory cytokines, inhibited the activation of nuclear factor (NF)-κB, and inhibited apoptosis and programmed necrosis. The mechanism may be that eleutheroside B can activate the insulin-like growth factor (IGF) pathway and its downstream pathway by downregulating the expression of IGFBP-7, thus promoting cell proliferation. Therefore, our results suggest that eleutheroside B is a potential drug for AKI treatment.
Collapse
Affiliation(s)
- Hongmei Zang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
- Anhui Institute of Innovative Drugs, Hefei 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Qin Yang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
- Anhui Institute of Innovative Drugs, Hefei 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
21
|
Priante G, Gianesello L, Ceol M, Del Prete D, Anglani F. Cell Death in the Kidney. Int J Mol Sci 2019; 20:E3598. [PMID: 31340541 PMCID: PMC6679187 DOI: 10.3390/ijms20143598] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Apoptotic cell death is usually a response to the cell's microenvironment. In the kidney, apoptosis contributes to parenchymal cell loss in the course of acute and chronic renal injury, but does not trigger an inflammatory response. What distinguishes necrosis from apoptosis is the rupture of the plasma membrane, so necrotic cell death is accompanied by the release of unprocessed intracellular content, including cellular organelles, which are highly immunogenic proteins. The relative contribution of apoptosis and necrosis to injury varies, depending on the severity of the insult. Regulated cell death may result from immunologically silent apoptosis or from immunogenic necrosis. Recent advances have enhanced the most revolutionary concept of regulated necrosis. Several modalities of regulated necrosis have been described, such as necroptosis, ferroptosis, pyroptosis, and mitochondrial permeability transition-dependent regulated necrosis. We review the different modalities of apoptosis, necrosis, and regulated necrosis in kidney injury, focusing particularly on evidence implicating cell death in ectopic renal calcification. We also review the evidence for the role of cell death in kidney injury, which may pave the way for new therapeutic opportunities.
Collapse
Affiliation(s)
- Giovanna Priante
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy.
| | - Lisa Gianesello
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Monica Ceol
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Dorella Del Prete
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| | - Franca Anglani
- Kidney Histomorphology and Molecular Biology Laboratory, Clinical Nephrology, Department of Medicine - DIMED, University of Padua, via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
22
|
Dizaji R, Sharafi A, Pourahmad J, Abdollahifar MA, Vatanpour H, Hosseini MJ. Induction of two independent immunological cell death signaling following hemoglobinuria -induced acute kidney injury: In vivo study. Toxicon 2019; 163:23-31. [DOI: 10.1016/j.toxicon.2019.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 02/08/2023]
|
23
|
A pan-RAF inhibitor LY3009120 inhibits necroptosis by preventing phosphorylation of RIPK1 and alleviates dextran sulfate sodium-induced colitis. Clin Sci (Lond) 2019; 133:919-932. [PMID: 30944150 DOI: 10.1042/cs20181081] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 12/13/2022]
Abstract
Abstract
A dramatic increase in the incidence of inflammatory bowel disease (IBD) has been observed in the past two decades, mainly in developed countries and also in developing regions. Necroptosis has been found to play an important role in the pathogenesis of IBD, suggesting its inhibitors are promising in clinic. However, clinical drugs targeting necroptosis are seriously lacking. Through screening a clinical compound library that contains 611 inhibitors, a pan-RAF inhibitor LY3009120 was found to be promising as a necroptosis inhibitor. LY3009120 inhibited necroptosis in vitro, and its inhibition against necroptosis was independent of its well-known activity to inhibit RAF. Surprisingly, LY3009120 prevented phosphorylation of receptor interacting serine/threonine kinase 1 (RIPK1) and subsequently phosphorylation of receptor interacting serine/threonine kinase 3 (RIPK3) and mixed lineage kinase domain like pseudokinase (MLKL) which happened during necroptosis. In vivo, LY3009120 significantly alleviated dextran sulfate sodium (DSS)-induced colitis as indicated by prevention of body weight loss, colon shortening, and decreased mortality. Furthermore, LY3009120 inhibited necroptosis of intestinal epithelial cells (IECs) and prevented intestinal barrier function loss. Consistently, LY3009120 decreased DSS-induced colonic inflammation, as indicated by decreased infiltration of macrophages and neutrophils, and decreased colonic TNF-α, IL-6, and IL-1β level in DSS treated mice. These results indicate that an anti-cancer pan-RAF inhibitor LY3009120 is a necroptosis inhibitor and may serve as a potential therapeutic drug for colitis.
Collapse
|
24
|
|
25
|
Abstract
Acute kidney injury (AKI) is a severe and frequent condition in hospitalized patients. Currently, no efficient therapy of AKI is available. Therefore, efforts focus on early prevention and potentially early initiation of renal replacement therapy to improve the outcome in AKI. The detection of AKI in hospitalized patients implies the need for early, accurate, robust, and easily accessible biomarkers of AKI evolution and outcome prediction because only a narrow window exists to implement the earlier-described measures. Even more challenging is the multifactorial origin of AKI and the fact that the changes of molecular expression induced by AKI are difficult to distinguish from those of the diseases associated or causing AKI as shock or sepsis. During the past decade, a considerable number of protein biomarkers for AKI have been described and we expect from recent advances in the field of omics technologies that this number will increase further in the future and be extended to other sorts of biomolecules, such as RNAs, lipids, and metabolites. However, most of these biomarkers are poorly defined by their AKI-associated molecular context. In this review, we describe the state-of-the-art tissue and biofluid proteomic and metabolomic technologies and new bioinformatics approaches for proteomic and metabolomic pathway and molecular interaction analysis. In the second part of the review, we focus on AKI-associated proteomic and metabolomic biomarkers and briefly outline their pathophysiological context in AKI.
Collapse
|
26
|
Yang C, Qi R, Yang B. Pathogenesis of Chronic Allograft Dysfunction Progress to Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:101-116. [PMID: 31399963 DOI: 10.1007/978-981-13-8871-2_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Kidney transplantation is a life-change measurement for the patients of end-stage renal disease (ESRD). However, the renal allograft cannot avoid initial acute kidney injury (AKI) and subsequent chronic allograft dysfunction (CAD), gradually develops fibrosis and eventually loses function. It is imperative to disclose the pathogenesis of AKI and CAD in order to facilitate interventions. We have studied the involvement of immunity, inflammation, and apoptosis in ischemia-reperfusion injury (IRI) and/or immunosuppressant induced AKI models, with associated chronic damage. Our research mainly focused on tubular epithelial cells (TECs) that are passive victims and also active participators in injury and mediate following repair or fibrosis. Targeting not only fibroblasts/myofibroblasts, but also TECs, might be a fundamental strategy to prevent and treat renal fibrosis. We have also evaluated the potential application of siRNA targeting caspase-3 and tissue protective erythropoietin derivatives, HBSP and CHBP, aiming to treat AKI and prevent CAD. Significant improvements have been obtained, but timely diagnosis and precise therapy of AKI and prevention of CAD progressing to ESRD are still very challenging. Modern technologies such as microarray and sequencing analysis have been used to identify biomarkers and potentially facilitate individual cell target treatment for transplant patients.
Collapse
Affiliation(s)
- Cheng Yang
- Department of Urology, Zhongshan Hospital, Zhangjiang Technology Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruochen Qi
- Department of Urology, Zhongshan Hospital, Zhangjiang Technology Institute, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Bin Yang
- Nantong-Leicester Joint Institute of Kidney Science, Department of Nephrology, Affiliated Hospital of Nantong University, Renal Group, Basic Medical Research Centre, Medical College of Nantong University, Nantong, 226001, Jiangsu, China. .,Department of Cardiovascular Sciences, University of Leicester, University Hospitals of Leicester, Leicester, LE1 7RH, UK.
| |
Collapse
|
27
|
Azad MAK, Nation RL, Velkov T, Li J. Mechanisms of Polymyxin-Induced Nephrotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1145:305-319. [PMID: 31364084 DOI: 10.1007/978-3-030-16373-0_18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Polymyxin-induced nephrotoxicity is the major dose-limiting factor and can occur in up to 60% of patients after intravenous administration. This chapter reviews the latest literature on the mechanisms of polymyxin-induced nephrotoxicity and its amelioration. After filtration by glomeruli, polymyxins substantially accumulate in renal proximal tubules via receptor-mediated endocytosis mainly by megalin and PEPT2. It is believed that subsequently, a cascade of interconnected events occur, including the activation of death receptor and mitochondrial apoptotic pathways, mitochondrial damage, endoplasmic reticulum stress, oxidative stress and cell cycle arrest. The current literature shows that oxidative stress plays a key role in polymyxin-induced kidney damage. Use of antioxidants have a potential in the attenuation of polymyxin-induced nephrotoxicity, thereby widening the therapeutic window. Mechanistic findings on polymyxin-induced nephrotoxicity are critical for the optimization of their use in the clinic and the discovery of safer polymyxin-like antibiotics.
Collapse
Affiliation(s)
- Mohammad A K Azad
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton Campus, Melbourne, VIC, Australia
| | - Roger L Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville Campus, Melbourne, VIC, Australia
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Jian Li
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton Campus, Melbourne, VIC, Australia.
| |
Collapse
|
28
|
Exclusive expression of transmembrane TNF aggravates acute glomerulonephritis despite reduced leukocyte infiltration and inflammation. Kidney Int 2018; 95:75-93. [PMID: 30389199 DOI: 10.1016/j.kint.2018.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 07/29/2018] [Accepted: 08/02/2018] [Indexed: 02/03/2023]
Abstract
Tumor necrosis factor-α (TNF) is a cytokine mediating inflammatory kidney diseases such as immune complex glomerulonephritis. Its two receptors, TNFR1 and TNFR2, play distinct roles in this process, with TNFR2 strongly required for induction of disease. In contrast to soluble TNF (sTNF), transmembrane TNF robustly activates TNFR2. Thus, we examined the functional role of transmembrane TNF by inducing heterologous nephrotoxic serum nephritis in wild-type and transgenic TNFΔ1-9,K11E knock-in mice expressing transmembrane TNF but no sTNF (memTNF mice). Compared to wild-type, nephritis was exacerbated in memTNF mice on day 5, indicated by increased albuminuria, higher serum urea levels, and more pronounced glomerular deposits, together with higher numbers of dying and proliferating glomerular cells. This was associated with greater loss of glomerular endothelial cells, increased podocyte stress, and signs of augmented necroptosis in memTNF kidneys. Aggravation of nephritis was dependent on transmembrane TNF expression in parenchymal cells, but not leukocytes. Surprisingly, increased kidney injury was associated with reduced renal leukocyte infiltration in memTNF mice, which correlated with decreased renal mRNA expression of pro-inflammatory mediators. This effect was also present in isolated memTNF glomeruli stimulated with interleukin-1β in vitro. Thus, uncleaved transmembrane TNF is an important mediator of renal tissue damage characterized by increased renal cell death and loss of glomerular endothelial cells in murine glomerulonephritis. In contrast, sTNF predominantly mediates renal leukocyte recruitment and inflammation. These findings highlight the importance of transmembrane TNF in inflammatory kidney disease as a possible therapeutic target.
Collapse
|
29
|
Jiang L, Liu XQ, Ma Q, Yang Q, Gao L, Li HD, Wang JN, Wei B, Wen J, Li J, Wu YG, Meng XM. hsa-miR-500a-3P alleviates kidney injury by targeting MLKL-mediated necroptosis in renal epithelial cells. FASEB J 2018; 33:3523-3535. [PMID: 30365367 DOI: 10.1096/fj.201801711r] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MLKL is a central mediator for necroptosis. Its knockout significantly relieves acute kidney injury (AKI). However, its upstream regulatory mechanism in AKI has not been fully elucidated. We recently reviewed how microRNAs (miRNAs), a type of well-studied epigenetic regulator, play critical roles in AKI. Here, we evaluated miRNAs that potentially target MLKL and evaluated their function in human tubular epithelial cells in response to toxic and ischemic insults. TargetScan analysis showed that miR-194-5P, miR-338-3P, miR-500a-3P, and miR-577 had MLKL binding sites. Although all 4 miRNAs are reduced in AKI, our data show that only hsa-miR-500a-3P was significantly suppressed in cisplatin-treated human tubular epithelial (HK2) cells. We further found that hsa-miR-500a-3P alleviated cisplatin-induced HK2 cell death, which was confirmed by transmission electron microscopy and flow cytometry. In addition, overexpression of hsa-miR-500a-3P decreased kidney injury molecule-1 mRNA and protein levels. Real-time PCR, ELISA, and immunofluorescence data show that hsa-miR-500a-3P protected against inflammatory response, evidenced by decreased monocyte chemotactic protein-1 and proinflammatory cytokines TNF-α and IL-8. Further, hsa-miR-500a-3P attenuated P65 NF-κB phosphorylation and promoter activity. Mechanistically, luciferase reporter assay showed that hsa-miR-500a-3P bound the 3'UTR of MLKL, thereby suppressing phosphorylation and membrane translocation of MLKL. In agreement with these findings, we identified that overexpression of hsa-miR-500a-3P attenuated cell injury and the inflammatory response in response to sodium azide treatment in an in vitro model. Results show that circulating exosomes from patients with AKI down-regulated miR-500a-3P, which suppressed cell injury and inflammation in HK2 cells. hsa-miR-500a-3P alleviated toxic and ischemic insults that were triggered by cell necroptosis and the inflammatory response in human HK2 cells by targeting MLKL. This may serve as a novel therapeutic target for treatment of AKI.-Jiang, L., Liu, X.-Q., Ma, Q., Yang, Q., Gao, L., Li, H.-D., Wang, J.-N., Wei, B., Wen, J., Li, J., Wu, Y.-G., Meng, X.-M. hsa-miR-500a-3P alleviates kidney injury by targeting MLKL-mediated necroptosis in renal epithelial cells.
Collapse
Affiliation(s)
- Ling Jiang
- Department of Nephrology, The First Affiliated Hospital, Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xue-Qi Liu
- Department of Nephrology, The First Affiliated Hospital, Anhui Medical University, Hefei, China.,School of Pharmacy, Anhui Medical University, Hefei, China
| | - Qiuying Ma
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Qin Yang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Li Gao
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hai-Di Li
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jia-Nan Wang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Biao Wei
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jiagen Wen
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China; and.,The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China; and.,The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| | - Yong-Gui Wu
- Department of Nephrology, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.,Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China; and.,The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, China
| |
Collapse
|
30
|
Barton CD, Pizer B, Jones C, Oni L, Pirmohamed M, Hawcutt DB. Identifying cisplatin-induced kidney damage in paediatric oncology patients. Pediatr Nephrol 2018; 33:1467-1474. [PMID: 28821959 PMCID: PMC6061670 DOI: 10.1007/s00467-017-3765-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 11/06/2022]
Abstract
Cisplatin is one chemotherapeutic agent used to treat childhood cancer in numerous treatment protocols, including as a single agent. It is likely to remain in clinical use over the long term. However, cisplatin-related toxicities, including neurotoxicity and nephrotoxicity, are common, affecting treatment, day-to-day life and survival of such children. With one in 700 young adults having survived childhood cancer, patients who have completed chemotherapy that includes cisplatin can experience long-term morbidity due to treatment-related adverse reactions. A better understanding of these toxicities is essential to facilitate prevention, surveillance and management. This review article discusses the effect of cisplatin-induced nephrotoxicity (Cis-N) in children and considers the underlying mechanisms. We focus on clinical features and identification of Cis-N (e.g. investigations and biomarkers) and the importance of magnesium homeostasis and supplementation.
Collapse
Affiliation(s)
- Chris D Barton
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Paediatric Oncology, Alder Hey Children's Hospital, Liverpool, UK
| | - Barry Pizer
- Department of Paediatric Oncology, Alder Hey Children's Hospital, Liverpool, UK
| | - Caroline Jones
- Department of Paediatric Nephrology, Alder Hey Children's Hospital, Liverpool, UK
| | - Louise Oni
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Paediatric Nephrology, Alder Hey Children's Hospital, Liverpool, UK
| | - Munir Pirmohamed
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Daniel B Hawcutt
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
- NIHR Alder Hey Clinical Research Facility, University of Liverpool, Liverpool, UK.
| |
Collapse
|
31
|
Wnt4 is significantly upregulated during the early phases of cisplatin-induced acute kidney injury. Sci Rep 2018; 8:10555. [PMID: 30002385 PMCID: PMC6043520 DOI: 10.1038/s41598-018-28595-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/25/2018] [Indexed: 02/07/2023] Open
Abstract
Wnt4 is a secreted growth factor associated with renal tubulogenesis. Our previous studies identified that renal and urinary Wnt4 are upregulated following ischemia-reperfusion injury in mice, but the roles of Wnt4 in other forms of acute kidney injury (AKI) remain unclear. Here, we investigated the changes in Wnt4 expression using a cisplatin-induced AKI model. We found that renal and urinary Wnt4 expression increased as early as 12 hours, peaked at day 4 following cisplatin-induced AKI and was closely correlated with histopathological alterations. By contrast, the serum creatinine level was significantly elevated until day 3, indicating that Wnt4 is more sensitive to early tubular injury than serum creatinine. In addition, renal Wnt4 was co-stained with aquaporin-1 and thiazide-sensitive NaCl cotransporter, suggesting that Wnt4 can detect both proximal and distal tubular injuries. These data were further confirmed in a clinical study. Increased urinary Wnt4 expression was detected earlier than serum creatinine and eGFR in patients with contrast-induced AKI after vascular intervention. This study is the first to demonstrate that increased expression of renal and urinary Wnt4 can be detected earlier than serum creatinine after drug-induced AKI. In particular, urinary Wnt4 can potentially serve as a noninvasive biomarker for monitoring patients with tubular injury.
Collapse
|
32
|
Cardinal H, Dieudé M, Hébert MJ. Endothelial Dysfunction in Kidney Transplantation. Front Immunol 2018; 9:1130. [PMID: 29875776 PMCID: PMC5974048 DOI: 10.3389/fimmu.2018.01130] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/04/2018] [Indexed: 12/17/2022] Open
Abstract
Kidney transplantation entails a high likelihood of endothelial injury. The endothelium is a target of choice for injury by ischemia-reperfusion, alloantibodies, and autoantibodies. A certain degree of ischemia-reperfusion injury inevitably occurs in the immediate posttransplant setting and can manifest as delayed graft function. Acute rejection episodes, whether T-cell or antibody-mediated, can involve the graft micro- and macrovasculature, leading to endothelial injury and adverse long-term consequences on graft function and survival. In turn, caspase-3 activation in injured and dying endothelial cells favors the release of extracellular vesicles (apoptotic bodies and apoptotic exosome-like vesicles) that further enhance autoantibody production, complement deposition, and microvascular rarefaction. In this review, we present the evidence for endothelial injury, its causes and long-term consequences on graft outcomes in the field of kidney transplantation.
Collapse
Affiliation(s)
- Héloïse Cardinal
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada.,University of Montreal, Montreal, QC, Canada
| | - Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada.,University of Montreal, Montreal, QC, Canada
| |
Collapse
|
33
|
Imamura M, Moon JS, Chung KP, Nakahira K, Muthukumar T, Shingarev R, Ryter SW, Choi AM, Choi ME. RIPK3 promotes kidney fibrosis via AKT-dependent ATP citrate lyase. JCI Insight 2018; 3:94979. [PMID: 29415885 DOI: 10.1172/jci.insight.94979] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 12/27/2017] [Indexed: 01/17/2023] Open
Abstract
Renal fibrosis is a common pathogenic response to injury in chronic kidney disease (CKD). The receptor-interacting protein kinase-3 (RIPK3), a regulator of necroptosis, has been implicated in disease pathogenesis. In mice subjected to unilateral ureteral obstruction-induced (UUO-induced) or adenine diet-induced (AD-induced) renal fibrosis, models of progressive kidney fibrosis, we demonstrate increased kidney expression of RIPK3. Mice genetically deficient in RIPK3 displayed decreased kidney fibrosis and improved kidney function relative to WT mice when challenged with UUO or AD. In contrast, mice genetically deficient in mixed-lineage kinase domain-like protein (MLKL), a downstream RIPK3 target, were not protected from UUO-induced kidney fibrosis. We demonstrate a pathway by which RIPK3 promotes fibrogenesis through the AKT-dependent activation of ATP citrate lyase (ACL). Genetic or chemical inhibition of RIPK3 suppressed the phosphorylation of AKT and ACL in response to TGF-β1 in fibroblasts. Inhibition of AKT or ACL suppressed TGF-β1-dependent extracellular matrix production and myofibroblast differentiation in fibroblasts. Pharmacological inhibition of ACL suppressed UUO-induced kidney fibrosis. RIPK3 expression was highly regulated in human CKD kidney. In conclusion, we identify a pathway by which RIPK3 promotes kidney fibrosis independently of MLKL-dependent necroptosis as a promising therapeutic target in CKD.
Collapse
Affiliation(s)
- Mitsuru Imamura
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jong-Seok Moon
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA.,Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | - Kuei-Pin Chung
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Kiichi Nakahira
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA.,Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, New York, USA
| | - Roman Shingarev
- Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stefan W Ryter
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Augustine Mk Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA.,Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, New York, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA.,Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, New York, USA
| |
Collapse
|
34
|
Necrostatin-1 Improves Long-term Functional Recovery Through Protecting Oligodendrocyte Precursor Cells After Transient Focal Cerebral Ischemia in Mice. Neuroscience 2018; 371:229-241. [DOI: 10.1016/j.neuroscience.2017.12.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 11/22/2022]
|
35
|
Wogonin protects against cisplatin-induced acute kidney injury by targeting RIPK1-mediated necroptosis. J Transl Med 2018; 98:79-94. [PMID: 29200200 DOI: 10.1038/labinvest.2017.115] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 11/08/2022] Open
Abstract
Acute kidney injury (AKI), characterized by aggressive inflammatory responses and destruction of renal resident cells, can cause abrupt kidney dysfunction. To date, effective therapy for AKI is lacking. In this study, we evaluated the renoprotective effect of wogonin, an herbal active compound, using a cisplatin-induced AKI mouse model. In vivo results show that wogonin substantially suppressed the increased levels of serum creatinine and blood urea nitrogen (BUN) almost to the normal level. Wogonin also attenuated tubular damage, shown by PAS staining, electron microscopy and molecular analysis of KIM-1. In addition, wogonin suppressed kidney inflammation as indicated by a >60% decrease in macrophage infiltration, a >50% reduction in inflammatory cytokine production and inhibited NF-κB activation in the injured kidney. Mechanistically, molecular docking results show that wogonin effectively inhibited RIPK1 by occupying the ATP-binding pocket of the enzyme, which is a key regulator of necroptosis. Moreover, inhibition of RIPK1, or RIPK3, reversed the protective effects of wogonin in cisplatin-treated HK2 cells, indicating wogonin works in a RIPK1/RIPK3-dependent manner. Surprisingly, wogonin enhanced the anti-proliferative effect of cisplatin on human hepatoma HepG2 cells. Thus, our findings suggest wogonin may be a renoprotective adjuvant for cisplatin-based anticancer therapy.
Collapse
|
36
|
Extracellular vesicles from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect against renal ischemia/reperfusion injury via delivering specificity protein (SP1) and transcriptional activating of sphingosine kinase 1 and inhibiting necroptosis. Cell Death Dis 2017; 8:3200. [PMID: 29233979 PMCID: PMC5870585 DOI: 10.1038/s41419-017-0041-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
Renal ischemia-reperfusion is a main cause of acute kidney injury (AKI), which is associated with high mortality. Here we show that extracellular vesicles (EVs) secreted from hiPSC-MSCs play a critical role in protection against renal I/R injury. hiPSC-MSCs-EVs can fuse with renal cells and deliver SP1 into target cells, subsequently active SK1 expression and increase S1P formation. Chromatin immunoprecipitation (ChIP) analyses and luciferase assay were used to confirm SP1 binds directly to the SK1 promoter region and promote promoter activity. Moreover, SP1 inhibition (MIT) or SK1 inhibition (SKI-II) completely abolished the renal protective effect of hiPSC-MSCs-EVs in rat I/R injury mode. However, pre-treatment of necroptosis inhibitor Nec-1 showed no difference with the administration of hiPSC-MSCs-EVs only. We then generated an SP1 knockout hiPSC-MSC cell line by CRISPR/Cas9 system and found that SP1 knockout failed to show the protective effect of hiPSC-MSCs-EVs unless restoring the level of SP1 by Ad-SP1 in vitro and in vivo. In conclusion, this study describes an anti-necroptosis effect of hiPSC-MSCs-EVs against renal I/R injury via delivering SP1 into target renal cells and intracellular activating the expression of SK1 and the generation of S1P. These findings suggest a novel mechanism for renal protection against I/R injury, and indicate a potential therapeutic approach for a variety of renal diseases and renal transplantation.
Collapse
|
37
|
Zhang C, Lin S, Li T, Jiang Y, Huang Z, Wen J, Cheng W, Li H. Mechanical force-mediated pathological cartilage thinning is regulated by necroptosis and apoptosis. Osteoarthritis Cartilage 2017; 25:1324-1334. [PMID: 28396243 DOI: 10.1016/j.joca.2017.03.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/15/2017] [Accepted: 03/31/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study aimed to identify the mechanisms underlying mandibular chondrocyte cell death and cartilage thinning in response to mechanical force. MATERIAL AND METHODS An in vivo model (compressive mechanical force) and an in vitro model (TNF-α+cycloheximide) were used to induce mandibular chondrocyte necroptosis. Hematoxylin and eosin staining and transmission electron microscopy were used to assess histological and subcellular changes in mandibular chondrocyte. Immunohistochemistry, western blotting, and real-time PCR were performed to evaluate changes in necroptotic protein markers. Cell activity, mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) were examined in vitro. RESULTS The expression of RIP1, RIP3 and Caspase-8 in mandibular chondrocytes significantly increased after 4 days of compressive mechanical force. Furthermore, the inhibition of necroptosis by Necrostatin-1 (Nec-1) or the inhibition of apoptosis by N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (Z-VAD) partially restored mechanical force-mediated mandibular cartilage thinning and chondrocyte death. Moreover, a synergistic effect on cell death inhibition and mandibular cartilage thickness restoration were found when treated with Nec-1+Z-VAD. The results of the in vitro model were in line with the in vivo ones, indicating that the changes in MMP and ROS generation contributed to mandibular chondrocyte apoptosis and necroptosis. CONCLUSION In addition to apoptosis, necroptosis also plays critical roles in pathological changes in mandibular cartilage after compressive mechanical force stimulation, implying RIP1, a master protein that mediates both necroptosis and apoptosis, as a potential therapeutic target in temporal mandibular osteoarthritis.
Collapse
Affiliation(s)
- C Zhang
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - S Lin
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - T Li
- Department of Biostatistics, Robert Stempel College of Public Health and Social Work, Florida International University, USA
| | - Y Jiang
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Z Huang
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - J Wen
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - W Cheng
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - H Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
38
|
Cippà PE, Fehr T. Pharmacological modulation of cell death in organ transplantation. Transpl Int 2017; 30:851-859. [PMID: 28480540 DOI: 10.1111/tri.12977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/20/2017] [Accepted: 04/29/2017] [Indexed: 12/22/2022]
Abstract
New options to pharmacologically modulate fundamental mechanisms of regulated cell death are rapidly evolving and found first clinical applications in cancer therapy. Here, we present an overview on how the recent advances in the understanding of the biology and pharmacology of cell death might influence research and clinical practice in solid organ transplantation. Of particular interest are the novel opportunities related to organ preservation and immunomodulation, which might contribute to promote organ repair and to develop more selective ways to modulate allogeneic immune responses to prevent rejection and induce immunological tolerance.
Collapse
Affiliation(s)
- Pietro E Cippà
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Fehr
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland.,Department of Internal Medicine, Cantonal Hospital Graubuenden, Chur, Switzerland
| |
Collapse
|
39
|
Adedoyin O, Boddu R, Traylor A, Lever JM, Bolisetty S, George JF, Agarwal A. Heme oxygenase-1 mitigates ferroptosis in renal proximal tubule cells. Am J Physiol Renal Physiol 2017; 314:F702-F714. [PMID: 28515173 DOI: 10.1152/ajprenal.00044.2017] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI because of its antiapoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of antioxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared with vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Cotreatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-l-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important antiferroptotic role of HO-1 in renal epithelial cells.
Collapse
Affiliation(s)
- Oreoluwa Adedoyin
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Ravindra Boddu
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Amie Traylor
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Jeremie M Lever
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Subhashini Bolisetty
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - James F George
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Department of Surgery, University of Alabama at Birmingham , Birmingham, Alabama
| | - Anupam Agarwal
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama.,Birmingham VA Medical Center , Birmingham, Alabama
| |
Collapse
|
40
|
Wang S, Zhang C, Li J, Niyazi S, Zheng L, Xu M, Rong R, Yang C, Zhu T. Erythropoietin protects against rhabdomyolysis-induced acute kidney injury by modulating macrophage polarization. Cell Death Dis 2017; 8:e2725. [PMID: 28383559 PMCID: PMC5477572 DOI: 10.1038/cddis.2017.104] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/02/2017] [Accepted: 02/15/2017] [Indexed: 12/15/2022]
Abstract
Erythropoietin (EPO) is a well-known hormone that is clinically used for the treatment of anemia. Very recently, an increasing body of evidence showed that EPO could still regulate bioactivities of macrophages. However, the details about the immunomodulatory effect of EPO on macrophages are not fully delineated, particularly in the setting of renal damages. Therefore, in the present study, we determined whether EPO could exert an impact on the dynamics of macrophages in a well-established model of rhabdomyolysis-induced acute kidney injury and explored the potential mechanisms. EPO was found to ameliorate kidney injuries by reducing macrophages recruitment and promoting phenotype switch toward M2 macrophages in vivo. It was also confirmed that EPO could directly suppress pro-inflammatory responses of M1 macrophages and promote M2 marker expression in vitro. Data indicated the possible involvement of Jak2/STAT3/STAT6 pathway in the augmentation of EPO on M2 polarization. These results improved the understanding of the immunoregulatory capacity of EPO on macrophages, which might optimize the therapeutic modalities of EPO.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Chao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jiawei Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Sidikejiang Niyazi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Long Zheng
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Oh CJ, Ha CM, Choi YK, Park S, Choe MS, Jeoung NH, Huh YH, Kim HJ, Kweon HS, Lee JM, Lee SJ, Jeon JH, Harris RA, Park KG, Lee IK. Pyruvate dehydrogenase kinase 4 deficiency attenuates cisplatin-induced acute kidney injury. Kidney Int 2016; 91:880-895. [PMID: 28040265 DOI: 10.1016/j.kint.2016.10.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 09/29/2016] [Accepted: 10/06/2016] [Indexed: 02/01/2023]
Abstract
Clinical prescription of cisplatin, one of the most widely used chemotherapeutic agents, is limited by its side effects, particularly tubular injury-associated nephrotoxicity. Since details of the underlying mechanisms are not fully understood, we investigated the role of pyruvate dehydrogenase kinase (PDK) in cisplatin-induced acute kidney injury. Among the PDK isoforms, PDK4 mRNA and protein levels were markedly increased in the kidneys of mice treated with cisplatin, and c-Jun N-terminal kinase activation was involved in cisplatin-induced renal PDK4 expression. Treatment with the PDK inhibitor sodium dichloroacetate (DCA) or genetic knockout of PDK4 attenuated the signs of cisplatin-induced acute kidney injury, including apoptotic morphology of the kidney tubules along with numbers of TUNEL-positive cells, cleaved caspase-3, and renal tubular injury markers. Cisplatin-induced suppression of the mitochondrial membrane potential, oxygen consumption rate, expression of electron transport chain components, cytochrome c oxidase activity, and disruption of mitochondrial morphology were noticeably improved in the kidneys of DCA-treated or PDK4 knockout mice. Additionally, levels of the oxidative stress marker 4-hydroxynonenal and mitochondrial reactive oxygen species were attenuated, whereas superoxide dismutase 2 and catalase expression and glutathione synthetase and glutathione levels were recovered in DCA-treated or PDK4 knockout mice. Interestingly, lipid accumulation was considerably attenuated in DCA-treated or PDK4 knockout mice via recovered expression of peroxisome proliferator-activated receptor-α and coactivator PGC-1α, which was accompanied by recovery of mitochondrial biogenesis. Thus, PDK4 mediates cisplatin-induced acute kidney injury, suggesting that PDK4 might be a therapeutic target for attenuating cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Chang Joo Oh
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Chae-Myeong Ha
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Republic of Korea
| | - Young-Keun Choi
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Sungmi Park
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Medical Center, Daegu, Republic of Korea
| | - Mi Sun Choe
- Department of Pathology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Nam Ho Jeoung
- Department of Pharmaceutical Science and Technology, College of Health and Medical Science, Catholic University of Daegu, Gyeongbuk, Republic of Korea
| | - Yang Hoon Huh
- Nano-Bio Electron Microscopy Research Group, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Hyo-Jeong Kim
- Nano-Bio Electron Microscopy Research Group, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Hee-Seok Kweon
- Nano-Bio Electron Microscopy Research Group, Korea Basic Science Institute, Daejeon, Republic of Korea
| | - Ji-Min Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Republic of Korea
| | - Sun Joo Lee
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jae-Han Jeon
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Robert A Harris
- Roudebush VA Medical Center and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Keun-Gyu Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
42
|
Abstract
Over a decade ago, it was proposed that the regulation of tubular repair in the kidney might involve the recapitulation of developmental pathways. Although the kidney cannot generate new nephrons after birth, suggesting a low level of regenerative competence, the tubular epithelial cells of the nephrons can proliferate to repair the damage after AKI. However, the debate continues over whether this repair involves a persistent progenitor population or any mature epithelial cell remaining after injury. Recent reports have highlighted the expression of Sox9, a transcription factor critical for normal kidney development, during postnatal epithelial repair in the kidney. Indeed, the proliferative response of the epithelium involves expression of several pathways previously described as being involved in kidney development. In some instances, these pathways are also apparently involved in the maladaptive responses observed after repeated injury. Whether development and repair in the kidney are the same processes or we are misinterpreting the similar expression of genes under different circumstances remains unknown. Here, we review the evidence for this link, concluding that such parallels in expression may more correctly represent the use of the same pathways in a distinct context, likely triggered by similar stressors.
Collapse
Affiliation(s)
- Melissa Helen Little
- Murdoch Children's Research Institute, Melbourne, Australia; and .,Department of Pediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Pamela Kairath
- Murdoch Children's Research Institute, Melbourne, Australia; and.,Department of Pediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| |
Collapse
|