1
|
Rasmussen SB, Boyko Y, Ranucci M, de Somer F, Ravn HB. Cardiac surgery-Associated acute kidney injury - A narrative review. Perfusion 2024; 39:1516-1530. [PMID: 37905794 DOI: 10.1177/02676591231211503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cardiac Surgery-Associated Acute Kidney Injury (CSA-AKI) is a serious complication seen in approximately 20-30% of cardiac surgery patients. The underlying pathophysiology is complex, often involving both patient- and procedure related risk factors. In contrast to AKI occurring after other types of major surgery, the use of cardiopulmonary bypass comprises both additional advantages and challenges, including non-pulsatile flow, targeted blood flow and pressure as well as the ability to manipulate central venous pressure (congestion). With an increasing focus on the impact of CSA-AKI on both short and long-term mortality, early identification and management of high-risk patients for CSA-AKI has evolved. The present narrative review gives an up-to-date summary on definition, diagnosis, underlying pathophysiology, monitoring and implications of CSA-AKI, including potential preventive interventions. The review will provide the reader with an in-depth understanding of how to identify, support and provide a more personalized and tailored perioperative management to avoid development of CSA-AKI.
Collapse
Affiliation(s)
- Sebastian Buhl Rasmussen
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Yuliya Boyko
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
| | - Marco Ranucci
- Department of Cardiovascular Anaesthesiology and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | | | - Hanne Berg Ravn
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
2
|
Wang F, Huang X, Wang S, Wu D, Zhang M, Wei W. The main molecular mechanisms of ferroptosis and its role in chronic kidney disease. Cell Signal 2024; 121:111256. [PMID: 38878804 DOI: 10.1016/j.cellsig.2024.111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
The term ferroptosis, coined in 2012, has been widely applied in various disease research fields. Ferroptosis is a newly regulated form of cell death distinct from apoptosis, necrosis, and autophagy, the mechanisms of which have been extensively studied. Chronic kidney disease, characterized by renal dysfunction, is a common disease severely affecting human health, with its occurrence and development influenced by multiple factors and leading to dysfunction in multiple systems. It often lacks obvious clinical symptoms in the early stages, and thus, diagnosis is typically made in the later stages, complicating treatment. While research on ferroptosis and acute kidney injury has made continuous progress, studies on the association between ferroptosis and chronic kidney disease remain limited. This review aims to summarize chronic kidney disease, investigate the mechanism and regulation of ferroptosis, and attempt to elucidate the role of ferroptosis in the occurrence and development of chronic kidney disease.
Collapse
Affiliation(s)
- Fulin Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xuesong Huang
- Department of Urology, Jilin People's Hospital, Jilin, China
| | - Shaokun Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Dawei Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | | | - Wei Wei
- Department of Urology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
3
|
Grujcic M, Milovanovic M, Nedeljkovic J, Jovanovic D, Arsenijevic D, Solovjova N, Stankovic V, Tanaskovic I, Arsenijevic A, Milovanovic J. The Possible Effects of Galectin-3 on Mechanisms of Renal and Hepatocellular Injury Induced by Intravascular Hemolysis. Int J Mol Sci 2024; 25:8129. [PMID: 39125698 PMCID: PMC11311984 DOI: 10.3390/ijms25158129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Intravascular hemolysis is a central feature of congenital and acquired hemolytic anemias, complement disorders, infectious diseases, and toxemias. Massive and/or chronic hemolysis is followed by the induction of inflammation, very often with severe damage of organs, which enhances the morbidity and mortality of hemolytic diseases. Galectin-3 (Gal-3) is a β-galactoside-binding lectin that modulates the functions of many immune cells, thus affecting inflammatory processes. Gal-3 is also one of the main regulators of fibrosis. The role of Gal-3 in the development of different kidney and liver diseases and the potential of therapeutic Gal-3 inhibition have been demonstrated. Therefore, the objective of this review is to discuss the possible effects of Gal-3 on the process of kidney and liver damage induced by intravascular hemolysis, as well as to shed light on the potential therapeutic targeting of Gal-3 in intravascular hemolysis.
Collapse
Affiliation(s)
- Mirjana Grujcic
- Institute for Transfusiology and Hemobiology of Military Medical Academy, 11000 Belgrade, Serbia;
| | - Marija Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Nedeljkovic
- Department of Medical Statistics and Informatics, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Danijela Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Dragana Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Natalija Solovjova
- Academy of Applied Studies Belgrade, The College of Health Science, Cara Dušana 254, 11080 Belgrade, Serbia;
| | - Vesna Stankovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Irena Tanaskovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Jelena Milovanovic
- Center for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia; (M.M.); (D.A.); (V.S.); (A.A.)
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
4
|
Xie T, Yao L, Li X. Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases. Antioxidants (Basel) 2024; 13:659. [PMID: 38929098 PMCID: PMC11200795 DOI: 10.3390/antiox13060659] [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: 05/06/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Kidney diseases pose a significant global health issue, frequently resulting in the gradual decline of renal function and eventually leading to end-stage renal failure. Abnormal iron metabolism and oxidative stress-mediated cellular dysfunction facilitates the advancement of kidney diseases. Iron homeostasis is strictly regulated in the body, and disturbance in this regulatory system results in abnormal iron accumulation or deficiency, both of which are associated with the pathogenesis of kidney diseases. Iron overload promotes the production of reactive oxygen species (ROS) through the Fenton reaction, resulting in oxidative damage to cellular molecules and impaired cellular function. Increased oxidative stress can also influence iron metabolism through upregulation of iron regulatory proteins and altering the expression and activity of key iron transport and storage proteins. This creates a harmful cycle in which abnormal iron metabolism and oxidative stress perpetuate each other, ultimately contributing to the advancement of kidney diseases. The crosstalk of iron metabolism and oxidative stress involves multiple signaling pathways, such as hypoxia-inducible factor (HIF) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. This review delves into the functions and mechanisms of iron metabolism and oxidative stress, along with the intricate relationship between these two factors in the context of kidney diseases. Understanding the underlying mechanisms should help to identify potential therapeutic targets and develop novel and effective therapeutic strategies to combat the burden of kidney diseases.
Collapse
Affiliation(s)
- Tiancheng Xie
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Li S, Han Q, Liu C, Wang Y, Liu F, Pan S, Zuo L, Gao D, Chen K, Feng Q, Liu Z, Liu D. Role of ferroptosis in chronic kidney disease. Cell Commun Signal 2024; 22:113. [PMID: 38347570 PMCID: PMC10860320 DOI: 10.1186/s12964-023-01422-8] [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: 08/14/2023] [Accepted: 12/03/2023] [Indexed: 02/15/2024] Open
Abstract
Chronic kidney disease (CKD) has historically been a significant global health concern, profoundly impacting both life and well-being. In the process of CKD, with the gradual loss of renal function, the incidence of various life-threatening complications, such as cardiovascular diseases, cerebrovascular accident, infection and stroke, is also increasing rapidly. Unfortunately, existing treatments exhibit limited ability to halt the progression of kidney injury in CKD, emphasizing the urgent need to delve into the precise molecular mechanisms governing the occurrence and development of CKD while identifying novel therapeutic targets. Renal fibrosis, a typical pathological feature of CKD, plays a pivotal role in disrupting normal renal structures and the loss of renal function. Ferroptosis is a recently discovered iron-dependent form of cell death characterized by lipid peroxide accumulation. Ferroptosis has emerged as a potential key player in various diseases and the initiation of organ fibrosis. Substantial evidence suggests that ferroptosis may significantly contribute to the intricate interplay between CKD and its progression. This review comprehensively outlines the intricate relationship between CKD and ferroptosis in terms of iron metabolism and lipid peroxidation, and discusses the current landscape of pharmacological research on ferroptosis, shedding light on promising avenues for intervention. It further illustrates recent breakthroughs in ferroptosis-related regulatory mechanisms implicated in the progression of CKD, thereby providing new insights for CKD treatment. Video Abstract.
Collapse
Affiliation(s)
- Shiyang Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Qiuxia Han
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Chang Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yixue Wang
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Fengxun Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Shaokang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Dan Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China
| | - Kai Chen
- Kaifeng Renmin Hospital, Kaifeng, 475000, Henan, People's Republic of China
| | - Qi Feng
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| | - Dongwei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, Henan, People's Republic of China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|
6
|
Shao L, Fang Q, Shi C, Zhang Y, Xia C, Zhang Y, Wang J, Chen F. Bone marrow mesenchymal stem cells inhibit ferroptosis via regulating the Nrf2-keap1/p53 pathway to ameliorate chronic kidney disease injury in the rats. J Recept Signal Transduct Res 2023; 43:9-18. [PMID: 36883687 DOI: 10.1080/10799893.2023.2185083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
PURPOSE Although bone marrow mesenchymal stem cells (BMMSCs) have been reported to exhibit a protective effect on animal models of chronic kidney disease (CKD), the exact mechanisms involved require further investigation. This study aims to investigate the underlying molecular mechanisms of BMMSCs in inhibiting ferroptosis and preventing an Adriamycin (ADR)-induced CKD injury. METHODS A rat model of long-term CKD induced through the injection of ADR administered twice weekly via the tail vein was used in this study. After BMMSCs were systemically administered through the renal artery, pathological staining, western blotting, ELISA, and transmission electron microscopy were used to analyze ferroptosis. RESULTS Analyses of renal function and histopathological findings indicated that ADR-mediated renal dysfunction improved in response to the BMMSC treatment, which was also sufficient to mediate the partial reversal of renal injury and mitochondrial pathological changes. BMMSCs decreased the ferrous iron (Fe2+) and reactive oxygen species and elevated glutathione (GSH) and GSH peroxidase 4. Moreover, the BMMSC treatment activated the expression of ferroptosis-related regulator NF-E2-related factor 2 (Nrf2) and inhibited Keap1 and p53 in CKD rat kidney tissues. CONCLUSIONS BMMSCs alleviate CKD, possibly resulting from the inhibition of kidney ferroptosis by regulating the Nrf2-Keap1/p53 pathway.
Collapse
Affiliation(s)
- Lishi Shao
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Qixiang Fang
- Department of Urology, The First Affiliated Hospital of the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi
| | - Chen Shi
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Ya Zhang
- Department of Radiology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan
| | - Chunjuan Xia
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Yifan Zhang
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Jiaping Wang
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan
| |
Collapse
|
7
|
He L, Wang B, Wang X, Liu Y, Song X, Zhang Y, Li X, Yang H. Uncover diagnostic immunity/hypoxia/ferroptosis/epithelial mesenchymal transformation-related CCR5, CD86, CD8A, ITGAM, and PTPRC in kidney transplantation patients with allograft rejection. Ren Fail 2022; 44:1850-1865. [PMID: 36330810 PMCID: PMC9639483 DOI: 10.1080/0886022x.2022.2141648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to identify predictive immunity/hypoxia/ferroptosis/epithelial mesenchymal transformation (EMT)-related biomarkers, pathways and new drugs in allograft rejection in kidney transplant patients. First, gene expression data were downloaded followed by identification of differentially expressed genes (DEGs), weighted gene co-expression network analysis (WGCNA) and protein–protein interaction (PPI) analysis. Second, diagnostic model was construction based on key genes, followed by correlation analysis between immune/hypoxia/ferroptosis/EMT and key diagnostic genes. Finally, drug prediction of diagnostic key genes was carried out. Five diagnostic genes were further identified, including CCR5, CD86, CD8A, ITGAM, and PTPRC, which were positively correlated with allograft rejection after the kidney transplant. Highly infiltrated immune cells, highly expression of hypoxia-related genes and activated status of EMT were significantly positively correlated with five diagnostic genes. Interestingly, suppressors of ferroptosis (SOFs) and drivers of ferroptosis (DOFs) showed a complex regulatory relationship between ferroptosis and five diagnostic genes. CD86, CCR5, and ITGAM were respectively drug target of ABATACEPT, MARAVIROC, and CLARITHROMYCIN. PTPRC was drug target of both PREDNISONE and EPOETIN BETA. In conclusion, the study could be useful in understanding changes in the microenvironment within transplantation, which may promote or sustain the development of allograft rejection after kidney transplantation.
Collapse
Affiliation(s)
- Long He
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| | - Boqian Wang
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| | - Xueyi Wang
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| | - Yuewen Liu
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| | - Xing Song
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| | - Yijian Zhang
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| | - Xin Li
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| | - Hongwei Yang
- Organ Transplantation Center, General Hospital of Northern Theater Command, Shenyang City, China
| |
Collapse
|
8
|
Kobayashi H, Yamada Y. Efficacy of oral ferric citrate hydrate treatment for anemia caused by niraparib: a case report. J Med Case Rep 2022; 16:440. [PMID: 36424618 PMCID: PMC9694605 DOI: 10.1186/s13256-022-03666-3] [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: 02/18/2022] [Accepted: 11/02/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Maintenance therapy using poly(adenosine diphosphate-ribose)polymerase inhibitors may have adverse events, including hematological toxicity, and may limit therapeutic potential in patients with cancer. Niraparib-induced anemia negatively impacts one's quality of life. Its amelioration by ferrous iron (for example, sodium ferrous citrate), folic acid, or vitamin B12 has not been supported. Oral ferric citrate hydrate increases circulating levels of iron and hepatic iron accumulation, improving renal anemia in patients with kidney failure receiving hemodialysis. The uptake of ferric iron is considered to be much higher than that of ferrous iron. CASE PRESENTATION The admitted patient was a 57-year-old Japanese woman with stage IIIB ovarian cancer who underwent primary debulking surgery and standard carboplatin-paclitaxel chemotherapy combined with bevacizumab, followed by niraparib (200 mg/day) maintenance therapy. The patient started oral SFC (100 mg/day) to treat niraparib-related anemia. However, she required two units of packed red blood cell transfusions three times within 3 months after starting niraparib treatment. The patient was diagnosed with niraparib-related anemia. The blood test results after 1 month from the start of niraparib treatment were as follows: red blood cells, 211 × 104/μL; hemoglobin, 7.0 g/dL; hematocrit, 20.8%; reticulocyte, 0.2%; platelet count, 18.0 × 104/μL. She was switched to oral ferric citrate hydrate with a dose of 500 mg per day and resumed niraparib treatment. She did not experience grade 3 niraparib-related hematological toxicity and achieved blood transfusion independence. CONCLUSIONS Ferric citrate hydrate may be a safe, effective, and well-tolerated oral drug for treating patients with niraparib-related anemia.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Ms.Clinic MayOne, Kashihara, Nara 634-0813 Japan ,grid.410814.80000 0004 0372 782XDepartment of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara 634-8522 Japan
| | - Yuki Yamada
- grid.410814.80000 0004 0372 782XDepartment of Obstetrics and Gynecology, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara 634-8522 Japan
| |
Collapse
|
9
|
Zhuo WQ, Wen Y, Luo HJ, Luo ZL, Wang L. Mechanisms of ferroptosis in chronic kidney disease. Front Mol Biosci 2022; 9:975582. [PMID: 36090053 PMCID: PMC9448928 DOI: 10.3389/fmolb.2022.975582] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a newly identified form of regulated cell death characterized by iron accumulation and lipid peroxidation. Ferroptosis plays an essential role in the pathology of numerous diseases and has emerged as a key area of focus in studies of chronic kidney disease (CKD). CKD is a major public health problem with high incidence and mortality that is characterized by a gradual loss of kidney function over time. The severity and complexity of CKD combined with the limited knowledge of its underlying molecular mechanism(s) have led to increased interest in this disease area. Here, we summarize recent advances in our understanding of the regulatory mechanism(s) of ferroptosis and highlight recent studies describing its role in the pathogenesis and progression of CKD. We further discuss the potential therapeutic benefits of targeting ferroptosis for the treatment of CKD and the major hurdles to overcome for the translation of in vitro studies into the clinic.
Collapse
Affiliation(s)
- Wen-Qing Zhuo
- Department of Nephrology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Yi Wen
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu, Sichuan, China
| | - Hui-Jun Luo
- Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Zhu-Lin Luo
- Department of General Surgery and Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command (Chengdu Military General Hospital), Chengdu, Sichuan, China
| | - Li Wang
- Department of Nephrology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
10
|
Zhang Y, Mou Y, Zhang J, Suo C, Zhou H, Gu M, Wang Z, Tan R. Therapeutic Implications of Ferroptosis in Renal Fibrosis. Front Mol Biosci 2022; 9:890766. [PMID: 35655759 PMCID: PMC9152458 DOI: 10.3389/fmolb.2022.890766] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/12/2022] [Indexed: 12/13/2022] Open
Abstract
Renal fibrosis is a common feature of chronic kidney disease (CKD), and can lead to the destruction of normal renal structure and loss of kidney function. Little progress has been made in reversing fibrosis in recent years. Ferroptosis is more immunogenic than apoptosis due to the release and activation of damage-related molecular patterns (DAMPs) signals. In this paper, the relationship between renal fibrosis and ferroptosis was reviewed from the perspective of iron metabolism and lipid peroxidation, and some pharmaceuticals or chemicals associated with both ferroptosis and renal fibrosis were summarized. Other programmed cell death and ferroptosis in renal fibrosis were also firstly reviewed for comparison and further investigation.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhua Mou
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Jianjian Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanjian Suo
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai Zhou
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Gu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zengjun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Ruoyun Tan,
| |
Collapse
|
11
|
Saurage E, Davis PR, Meek R, Pollock DM, Kasztan M. Endothelin A receptor antagonist attenuated renal iron accumulation in iron overload heme oxygenase-1 knockout mice. Can J Physiol Pharmacol 2022; 100:637-650. [PMID: 35413222 PMCID: PMC10164438 DOI: 10.1139/cjpp-2022-0038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Progressive iron accumulation and renal impairment are prominent in both patients and mouse models of sickle cell disease (SCD). Endothelin A receptor (ETA) antagonism prevents this iron accumulation phenotype and reduces renal iron deposition in proximal tubules of SCD mice. To better understand the mechanisms of iron metabolism in the kidney and the role of ETA receptor in iron chelation and transport, we studied renal iron handling in a non-sickle cell iron overload model, heme oxygenase-1 (Hmox-1-/-) knockout mice. We found that Hmox-1-/- mice had elevated plasma endothelin-1 (ET-1), cortical ET-1 mRNA expression, and renal iron content compared to Hmox-1+/+ controls. The ETA receptor antagonist, ambrisentan, attenuated renal iron deposition, without any changes to anemia status in Hmox-1-/- mice. This was accompanied by reduced urinary iron excretion. Finally, ambrisentan had an important iron recycling effect by increasing expression of cellular iron exporter, ferroportin-1 (FPN-1) and circulating total iron levels in Hmox-1-/- mice. These findings suggest the ET-1/ETA signaling pathway contributes to in renal iron trafficking in a murine model of iron overload.
Collapse
Affiliation(s)
- Elizabeth Saurage
- The University of Alabama at Birmingham School of Medicine, 9967, Medicine, Division of Nephrology, Birmingham, Alabama, United States;
| | - Parker Ross Davis
- The University of Alabama at Birmingham Department of Medicine, 164494, Medicine, Division of Nephrology, Birmingham, Alabama, United States;
| | - Rachel Meek
- The University of Alabama at Birmingham School of Medicine, 9967, Medicine, Devision of Nephrology, Birmingham, Alabama, United States;
| | - David M Pollock
- The University of Alabama at Birmingham Department of Medicine, 164494, Medicine, Division of Nephrology, Birmingham, Alabama, United States;
| | - Malgorzata Kasztan
- The University of Alabama at Birmingham School of Medicine, 9967, Pediatrics, Division of Hematology-Oncology, Birmingham, Alabama, United States;
| |
Collapse
|
12
|
Zhang X, Li X. Abnormal Iron and Lipid Metabolism Mediated Ferroptosis in Kidney Diseases and Its Therapeutic Potential. Metabolites 2022; 12:58. [PMID: 35050181 PMCID: PMC8779729 DOI: 10.3390/metabo12010058] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 12/15/2022] Open
Abstract
Ferroptosis is a newly identified form of regulated cell death driven by iron-dependent phospholipid peroxidation and oxidative stress. Ferroptosis has distinct biological and morphology characteristics, such as shrunken mitochondria when compared to other known regulated cell deaths. The regulation of ferroptosis includes different molecular mechanisms and multiple cellular metabolic pathways, including glutathione/glutathione peroxidase 4(GPX4) signaling pathways, which are involved in the amino acid metabolism and the activation of GPX4; iron metabolic signaling pathways, which are involved in the regulation of iron import/export and the storage/release of intracellular iron through iron-regulatory proteins (IRPs), and lipid metabolic signaling pathways, which are involved in the metabolism of unsaturated fatty acids in cell membranes. Ferroptosis plays an essential role in the pathology of various kidneys diseases, including acute kidney injury (AKI), chronic kidney disease (CKD), autosomal dominant polycystic kidney disease (ADPKD), and renal cell carcinoma (RCC). Targeting ferroptosis with its inducers/initiators and inhibitors can modulate the progression of kidney diseases in animal models. In this review, we discuss the characteristics of ferroptosis and the ferroptosis-based mechanisms, highlighting the potential role of the main ferroptosis-associated metabolic pathways in the treatment and prevention of various kidney diseases.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
13
|
Cheng Z, Akatsuka S, Li GH, Mori K, Takahashi T, Toyokuni S. Ferroptosis resistance determines high susceptibility of murine A/J strain to iron-induced renal carcinogenesis. Cancer Sci 2021; 113:65-78. [PMID: 34699654 PMCID: PMC8748236 DOI: 10.1111/cas.15175] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer susceptibility is a critical factor in the understanding of carcinogenesis. Intraperitoneal (i.p.) injection of an iron chelate, ferric nitrilotriacetate (Fe‐NTA), produces hydroxyl radicals via Fenton reaction to induce ferroptosis in renal proximal tubules. Rats or mice subjected to repeated i.p. injections of Fe‐NTA develop renal cell carcinoma (RCC). To elucidate the molecular mechanisms that cause susceptibility to renal carcinogenesis, we first established an inter‐strain difference in the susceptibility to Fe‐NTA‐induced renal carcinogenesis in mice. Based on a previous observation of a low incidence of RCC with this model in C57BL/6J strain mice, we investigated A/J strain mice here, which demonstrated significantly higher susceptibility to Fe‐NTA‐induced renal carcinogenesis. Homozygous deletion of the Cdkn2a/2b tumor suppressor locus was detected for the first time in A/J strain mice. Focusing on ferroptosis and iron metabolism, we explored the mechanisms involved that lead to the difference in RCC development. We compared the protective responses in the kidney of A/J and C57BL/6J strains after Fe‐NTA treatment. After 3‐week Fe‐NTA treatment, A/J mice maintained higher levels of expression of glutathione peroxidase 4 and xCT (SLC7A11), leading to a lower level of lipid peroxidation. Simultaneously, A/J mice had decreased expression of transferrin receptor and increased expression of ferritin to greater degrees than C57BL/6 mice. After a single Fe‐NTA injection, higher levels of oxidative cell damage and cytosolic catalytic Fe(II) were observed in C57BL/6J mice, accompanied by a greater increase in lipocalin‐2. Lipocalin‐2 deficiency significantly decreased oxidative renal damage. Our results suggest that a genetic trait favoring ferroptosis resistance contributes to high susceptibility to Fe‐NTA‐induced RCC in A/J strain.
Collapse
Affiliation(s)
- Zhen Cheng
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinya Akatsuka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Guang Hua Li
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyoshi Mori
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan.,Department of Molecular and Clinical Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.,Department of Nephrology and Kidney Research, Shizuoka General Hospital, Shizuoka, Japan
| | | | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
14
|
Yadav AK, Ghosh A, Divyaveer S, Mukhopadhyay B, Kundu M, Kumar V, Lele SS, Rajapurkar MM, Jha V. Serum catalytic iron and progression of chronic kidney disease: findings from the ICKD study. Nephrol Dial Transplant 2021; 37:gfab271. [PMID: 34534345 DOI: 10.1093/ndt/gfab271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The non-transferrin bound catalytic iron moiety catalyses production of toxic reactive oxygen species and is associated with adverse outcomes. We hypothesized that serum catalytic iron (SCI) is associated with progression of chronic kidney disease (CKD). METHODS Baseline samples of the Indian Chronic Kidney Disease participants with at least one follow up visit were tested for total iron, iron binding capacity, transferrin saturation, SCI, ferritin and hepcidin. SCI was measured using the bleomycin-detectable iron assay that detects biologically active iron. Association with the incidence of major kidney endpoints, (MAKE, a composite of kidney death, kidney failure or > 40% loss of eGFR) was examined using Cox proportional hazards model adjusted for sex and age. RESULTS 2002 subjects (49.9 ± 11.6 years, 68.1% males, baseline eGFR 41.01 ml/min/1.73m2) were enrolled. After a median follow up of 12.6 (12.2, 16.7) months, the composite MAKE occurred in 280 (14%). After adjusting for age and sex, increase from 25th to 75th percentile in SCI, transferrin saturation, ferritin and hepcidin were associated with 78% (43-122%), 34% (10-62%), 57% (24-100%) and 74% (35-124%) increase in hazard of MAKE, respectively. SCI was associated with MAKE and kidney failure after adjustment for occupational exposure, hypertension, diabetes, tobacco, alcohol use, history of AKI, baseline eGFR, uACR, and allowing baseline hazard to vary by centre. CONCLUSIONS SCI is strongly and independently associated with composite MAKE in patients with mild to moderate CKD. Confirmation in other studies will allow consideration of SCI as a risk marker and treatment target.
Collapse
Affiliation(s)
- Ashok K Yadav
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh
| | - Arpita Ghosh
- George Institute for Global Health India, New Delhi
| | - Smita Divyaveer
- Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh
| | | | - Monica Kundu
- George Institute for Global Health India, New Delhi
| | - Vivek Kumar
- Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh
| | - Suhas S Lele
- Department of Cardiology, Bhailal Amin General Hospital, Baroda
| | | | - Vivekanand Jha
- George Institute for Global Health India, New Delhi
- School of Public Health, Imperial College, London, UK
- Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
15
|
Zhao S, Wang X, Zheng X, Liang X, Wang Z, Zhang J, Zhao X, Zhuang S, Pan Q, Sun F, Shang W, Barasch J, Qiu A. Iron deficiency exacerbates cisplatin- or rhabdomyolysis-induced acute kidney injury through promoting iron-catalyzed oxidative damage. Free Radic Biol Med 2021; 173:81-96. [PMID: 34298093 PMCID: PMC9482792 DOI: 10.1016/j.freeradbiomed.2021.07.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 02/05/2023]
Abstract
Iron deficiency is the most common micronutrient deficiency worldwide. While iron deficiency is known to suppress embryonic organogenesis, its effect on the adult organ in the context of clinically relevant damage has not been considered. Here we report that iron deficiency is a risk factor for nephrotoxic intrinsic acute kidney injury of the nephron (iAKI). Iron deficiency exacerbated cisplatin-induced iAKI by markedly increasing non-heme catalytic iron and Nox4 protein which together catalyze production of hydroxyl radicals followed by protein and DNA oxidation, apoptosis and ferroptosis. Crosstalk between non-heme catalytic iron/Nox4 and downstream oxidative damage generated a mutual amplification cycle that facilitated rapid progression of cisplatin-induced iAKI. Iron deficiency also exacerbated a second model of iAKI, rhabdomyolysis, via increasing catalytic heme-iron. Heme-iron induced lipid peroxidation and DNA oxidation by interacting with Nox4-independent mechanisms, promoting p53/p21 activity and cellular senescence. Our data suggests that correcting iron deficiency and/or targeting specific catalytic iron species are strategies to mitigate iAKI in a wide range of patients with diverse forms of kidney injury.
Collapse
Affiliation(s)
- Shifeng Zhao
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xueqiao Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoqing Zheng
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiu Liang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhigang Wang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Juanlian Zhang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xudong Zhao
- Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shougang Zhuang
- Division of Nephrology, Department of Medicine, Brown University, Providence, USA
| | - Qiuhui Pan
- Department of Clinical Laboratory Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wenjun Shang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Columbia University, New York, USA.
| | - Andong Qiu
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
16
|
Kovacevic S, Ivanov M, Zivotic M, Brkic P, Miloradovic Z, Jeremic R, Mihailovic-Stanojevic N, Vajic UJ, Karanovic D, Jovovic D, Nesovic Ostojic J. Immunohistochemical Analysis of 4-HNE, NGAL, and HO-1 Tissue Expression after Apocynin Treatment and HBO Preconditioning in Postischemic Acute Kidney Injury Induced in Spontaneously Hypertensive Rats. Antioxidants (Basel) 2021; 10:antiox10081163. [PMID: 34439411 PMCID: PMC8388865 DOI: 10.3390/antiox10081163] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/07/2023] Open
Abstract
Oxidative stress has been considered as a central aggravating factor in the development of postischemic acute kidney injury (AKI). The aim of this study was to perform the immunohistochemical analysis of 4-hydroxynonenal (4-HNE), neutrophil gelatinase-associated lipocalin (NGAL), and heme oxygenase-1 (HO-1) tissue expression after apocynin (APO) treatment and hyperbaric oxygenation (HBO) preconditioning, applied as single or combined protocol, in postischemic acute kidney injury induced in spontaneously hypertensive rats (SHR). Twenty-four hours before AKI induction, HBO preconditioning was carried out by exposing to pure oxygen (2.026 bar) twice a day, for 60 min in two consecutive days. Acute kidney injury was induced by removal of the right kidney while the left renal artery was occluded for 45 min by atraumatic clamp. Apocynin was applied in a dose of 40 mg/kg body weight, intravenously, 5 min before reperfusion. We showed increased 4-HNE renal expression in postischemic AKI compared to Sham-operated (SHAM) group. Apocynin treatment, with or without HBO preconditioning, improved creatinine and phosphate clearances, in postischemic AKI. This improvement in renal function was accompanied with decreased 4-HNE, while HO-1 kidney expression restored close to the control group level. NGAL renal expression was also decreased after apocynin treatment, and HBO preconditioning, with or without APO treatment. Considering our results, we can say that 4-HNE tissue expression can be used as a marker of oxidative stress in postischemic AKI. On the other hand, apocynin treatment and HBO preconditioning reduced oxidative damage, and this protective effect might be expected even in experimental hypertensive condition.
Collapse
Affiliation(s)
- Sanjin Kovacevic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milan Ivanov
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia; (M.I.); (Z.M.); (N.M.-S.); (U.J.V.); (D.K.); (D.J.)
| | - Maja Zivotic
- Institute of Pathology, Medical Faculty, University of Belgrade, 11000 Belgrade, Serbia;
| | - Predrag Brkic
- Department of Medical Physiology, Medical Faculty, University of Belgrade, 11000 Belgrade, Serbia; (P.B.); (R.J.)
| | - Zoran Miloradovic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia; (M.I.); (Z.M.); (N.M.-S.); (U.J.V.); (D.K.); (D.J.)
| | - Rada Jeremic
- Department of Medical Physiology, Medical Faculty, University of Belgrade, 11000 Belgrade, Serbia; (P.B.); (R.J.)
| | - Nevena Mihailovic-Stanojevic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia; (M.I.); (Z.M.); (N.M.-S.); (U.J.V.); (D.K.); (D.J.)
| | - Una Jovana Vajic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia; (M.I.); (Z.M.); (N.M.-S.); (U.J.V.); (D.K.); (D.J.)
| | - Danijela Karanovic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia; (M.I.); (Z.M.); (N.M.-S.); (U.J.V.); (D.K.); (D.J.)
| | - Djurdjica Jovovic
- Department of Cardiovascular Physiology, Institute for Medical Research, University of Belgrade, 11129 Belgrade, Serbia; (M.I.); (Z.M.); (N.M.-S.); (U.J.V.); (D.K.); (D.J.)
| | - Jelena Nesovic Ostojic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, 11000 Belgrade, Serbia;
- Correspondence: ; Tel./Fax: +381-11-2685-340
| |
Collapse
|
17
|
The Cross-Link between Ferroptosis and Kidney Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6654887. [PMID: 34007403 PMCID: PMC8110383 DOI: 10.1155/2021/6654887] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/31/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
Acute and chronic kidney injuries result from structural dysfunction and metabolic disorders of the kidney in various etiologies, which significantly affect human survival and social wealth. Nephropathies are often accompanied by various forms of cell death and complex microenvironments. In recent decades, the study of kidney diseases and the traditional forms of cell death have improved. Nontraditional forms of cell death, represented by ferroptosis and necroptosis, have been discovered in the field of kidney diseases, which have reshuffled the role of traditional cell death in nephropathies. Although interactions between ferroptosis and acute kidney injury (AKI) have been continuously explored, studies on ferroptosis and chronic kidney disease (CKD) remain limited. Here, we have reviewed the therapeutic significance of ferroptosis in AKI and anticipated the curative potential of ferroptosis for CKD in the hope of providing insights into ferroptosis and CKD.
Collapse
|
18
|
Annamalai C, Seth R, Viswanathan P. Ferrotoxicity and Its Amelioration by Calcitriol in Cultured Renal Cells. Anal Cell Pathol (Amst) 2021. [DOI: https://doi.org/10.1155/2021/6634429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Globally, acute kidney injury (AKI) is associated with significant mortality and an enormous economic burden. Whereas iron is essential for metabolically active renal cells, it has the potential to cause renal cytotoxicity by promoting Fenton chemistry-based oxidative stress involving lipid peroxidation. In addition, 1,25-dihydroxyvitamin D3 (calcitriol), the active form of vitamin D, is reported to have an antioxidative role. In this study, we intended to demonstrate the impact of vitamin D on iron-mediated oxidant stress and cytotoxicity of Vero cells exposed to iohexol, a low osmolar iodine-containing contrast media in vitro. Cultured Vero cells were pretreated with 1,25-dihydroxyvitamin D3 dissolved in absolute ethanol (0.05%, 2.0 mM) at a dose of 1 mM for 6 hours. Subsequently, iohexol was added at a concentration of 100 mg iodine per mL and incubated for 3 hours. Total cellular iron content was analysed by a flame atomic absorption spectrophotometer at 372 nm. Lipid peroxidation was determined by TBARS (thiobarbituric acid reactive species) assay. Antioxidants including total thiol content were assessed by Ellman’s method, catalase by colorimetric method, and superoxide dismutase (SOD) by nitroblue tetrazolium assay. The cells were stained with DAPI (4
,6-diamidino-2-phenylindole), and the cytotoxicity was evaluated by viability assay (MTT assay). The results indicated that iohexol exposure caused a significant increase of the total iron content in Vero cells. A concomitant increase of lipid peroxidation and decrease of total thiol protein levels, catalase, and superoxide dismutase activity were observed along with decreased cell viability in comparison with the controls. Furthermore, these changes were significantly reversed when the cells were pretreated with vitamin D prior to incubation with iohexol. Our findings of this in vitro model of iohexol-induced renotoxicity lend further support to the nephrotoxic potential of iron and underpin the possible clinical utility of vitamin D for the treatment and prevention of AKI.
Collapse
Affiliation(s)
- Chandrashekar Annamalai
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632 014 Tamil Nadu, India
| | - Rohit Seth
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, 495009 Chhattisgarh, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632 014 Tamil Nadu, India
| |
Collapse
|
19
|
Annamalai C, Seth R, Viswanathan P. Ferrotoxicity and Its Amelioration by Calcitriol in Cultured Renal Cells. Anal Cell Pathol (Amst) 2021; 2021:6634429. [PMID: 33680716 PMCID: PMC7925041 DOI: 10.1155/2021/6634429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/22/2021] [Accepted: 02/15/2021] [Indexed: 02/05/2023] Open
Abstract
Globally, acute kidney injury (AKI) is associated with significant mortality and an enormous economic burden. Whereas iron is essential for metabolically active renal cells, it has the potential to cause renal cytotoxicity by promoting Fenton chemistry-based oxidative stress involving lipid peroxidation. In addition, 1,25-dihydroxyvitamin D3 (calcitriol), the active form of vitamin D, is reported to have an antioxidative role. In this study, we intended to demonstrate the impact of vitamin D on iron-mediated oxidant stress and cytotoxicity of Vero cells exposed to iohexol, a low osmolar iodine-containing contrast media in vitro. Cultured Vero cells were pretreated with 1,25-dihydroxyvitamin D3 dissolved in absolute ethanol (0.05%, 2.0 mM) at a dose of 1 mM for 6 hours. Subsequently, iohexol was added at a concentration of 100 mg iodine per mL and incubated for 3 hours. Total cellular iron content was analysed by a flame atomic absorption spectrophotometer at 372 nm. Lipid peroxidation was determined by TBARS (thiobarbituric acid reactive species) assay. Antioxidants including total thiol content were assessed by Ellman's method, catalase by colorimetric method, and superoxide dismutase (SOD) by nitroblue tetrazolium assay. The cells were stained with DAPI (4',6-diamidino-2-phenylindole), and the cytotoxicity was evaluated by viability assay (MTT assay). The results indicated that iohexol exposure caused a significant increase of the total iron content in Vero cells. A concomitant increase of lipid peroxidation and decrease of total thiol protein levels, catalase, and superoxide dismutase activity were observed along with decreased cell viability in comparison with the controls. Furthermore, these changes were significantly reversed when the cells were pretreated with vitamin D prior to incubation with iohexol. Our findings of this in vitro model of iohexol-induced renotoxicity lend further support to the nephrotoxic potential of iron and underpin the possible clinical utility of vitamin D for the treatment and prevention of AKI.
Collapse
Affiliation(s)
- Chandrashekar Annamalai
- 1Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632 014 Tamil Nadu, India
| | - Rohit Seth
- 2Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, 495009 Chhattisgarh, India
| | - Pragasam Viswanathan
- 1Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632 014 Tamil Nadu, India
| |
Collapse
|
20
|
Cavdar Z, Oktan MA, Ural C, Calisir M, Kocak A, Heybeli C, Yildiz S, Arici A, Ellidokuz H, Celik A, Yilmaz O, Sarioglu S, Cavdar C. Renoprotective Effects of Alpha Lipoic Acid on Iron Overload-Induced Kidney Injury in Rats by Suppressing NADPH Oxidase 4 and p38 MAPK Signaling. Biol Trace Elem Res 2020; 193:483-493. [PMID: 31025242 DOI: 10.1007/s12011-019-01733-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/11/2019] [Indexed: 10/27/2022]
Abstract
We aimed to investigate the protective effect of alpha lipoic acid (ALA), a powerful antioxidant, against oxidative kidney damage induced by iron overload in rats. Male Wistar albino rats were separated into groups: control (n = 7), ALA (100 mg/kg (n = 7), iron sucrose (IS) (40 mg/kg) (n = 7), and IS + ALA (40 mg/kg IS administration followed by 100 mg/kg ALA) (n = 7). IS and ALA were injected weekly for 4 weeks via the tail vein. Blood and kidneys were collected at sacrification on day 29. Serum creatinine and iron levels were analyzed. Tubular injury and iron deposits were evaluated histopathologically. Additionally, iron, malondialdehyde (MDA), superoxide dismutase (SOD), catalase, and glutathione (GSH) levels and mRNA expressions of the subunits of NADPH oxidase, known as NOX4 and p22phox, tumor necrosis factor (TNF)-α, kidney injury molecule-1 (KIM-1), and also p38 MAPK signaling in the kidneys, were evaluated biochemically. In the IS group, serum creatinine and iron level, tubular dilation, and loss of brush border in the kidneys were significantly higher than those of the control. Although those changes were reduced by ALA, the differences were not statistically significant. However, ALA reduced significantly MDA level and increased SOD activity in the kidney during IS administration. ALA also significantly reduced mRNA expressions of NOX4 and p22phox induced by IS, which was parallel to significant decreases of TNF-α and KIM-1 mRNA expressions. Moreover, ALA could suppress the activation of p38 MAPK during IS administration. In conclusion, ALA may be an effective strategy to attenuate in IS-induced oxidative kidney injury.
Collapse
Affiliation(s)
- Zahide Cavdar
- Department of Molecular Medicine, Health Sciences Institute, Dokuz Eylul University, 35340, Izmir, Turkey.
| | - Mehmet Asi Oktan
- Department of Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Cemre Ural
- Department of Molecular Medicine, Health Sciences Institute, Dokuz Eylul University, 35340, Izmir, Turkey
| | - Meryem Calisir
- Department of Laboratory Animal Science, Health Sciences Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ayse Kocak
- Department of Molecular Medicine, Health Sciences Institute, Dokuz Eylul University, 35340, Izmir, Turkey
| | - Cihan Heybeli
- Department of Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Serkan Yildiz
- Department of Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Aylin Arici
- Department of Medical Pharmacology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Hulya Ellidokuz
- Department of Biostatistics, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Ali Celik
- Department of Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Osman Yilmaz
- Department of Laboratory Animal Science, Health Sciences Institute, Dokuz Eylul University, Izmir, Turkey
| | - Sulen Sarioglu
- Department of Pathology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Caner Cavdar
- Department of Nephrology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
21
|
Decker G, Sprinkart AM, Wolter K, Schild HH, Thomas DK. The impact of rheolytic percutaneous mechanical thrombectomy on glomerular filtration rate levels. J Vasc Surg Venous Lymphat Disord 2020; 8:545-550. [PMID: 31928956 DOI: 10.1016/j.jvsv.2019.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/25/2019] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Rheolytic percutaneous mechanical thrombectomy (PMT) has been established as an endovascular technique for thrombus removal. Initial studies reporting on postinterventional kidney dysfunction have surfaced. The aim of this study was to investigate glomerular filtration rate (GFR) changes after PMT. METHODS A total of 45 interventions were included; 21 were performed in the venous system and 24 in the arterial system. Renal function was evaluated through assessment of GFR value changes from baseline to a minimum of two postinterventional values, and RIFLE criteria (Risk, Injury, Failure, Loss of kidney function, End-stage kidney disease) were applied. RESULTS The univariate analysis of variance revealed a significant association of GFR increase between time points and the type of intervention (arterious or venous; P = .002), whereas there was no significant association of intervention duration (P = .382), quantity of administered contrast medium (P = .544), or use of urokinase (P = .377). Repeated measures analysis of variance revealed a significant difference in GFR values between the four time points for venous interventions (P = .008) but not for arterial interventions (P = .908). In venous interventions, postinterventional GFR values were significantly lower compared with preinterventional values (P = .008) and the two measurements after intervention (P = .017 and P = .014, respectively). According to the RIFLE criteria, 1 of the 21 patients in the venous group had a complete loss of kidney function and 2 patients progressed to the risk group (GFR decreases >25%). CONCLUSIONS PMT in the venous system has a significant impact on GFR levels, although there is only a low risk for clinically important renal dysfunction. The occurrence of renal impairment should be taken into account in evaluating PMT treatment, especially because of the associated morbidity.
Collapse
Affiliation(s)
- Georges Decker
- Department of Radiology, University of Bonn, Bonn, Germany.
| | | | - Karsten Wolter
- Department of Radiology, University of Bonn, Bonn, Germany
| | - Hans H Schild
- Department of Radiology, University of Bonn, Bonn, Germany
| | | |
Collapse
|
22
|
Zhao L, Zou Y, Zhang J, Zhang R, Ren H, Li L, Guo R, Zhang J, Liu F. Serum transferrin predicts end-stage Renal Disease in Type 2 Diabetes Mellitus patients. Int J Med Sci 2020; 17:2113-2124. [PMID: 32922172 PMCID: PMC7484672 DOI: 10.7150/ijms.46259] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/16/2020] [Indexed: 02/05/2023] Open
Abstract
Background: To investigate the relationship between serum iron status and renal outcome in patients with type 2 diabetes mellitus (T2DM). Methods: Chinese patients (n=111) with T2DM and biopsy-proven diabetic nephropathy (DN) were surveyed in a longitudinal, retrospective study. Serum iron, total iron-binding capacity, ferritin, and transferrin were measured at the time of renal biopsy. Iron deposition and transferrin staining were performed with renal biopsy specimens of DN patients and potential kidney donors. End-stage renal disease (ESRD) was the end-point. ESRD was defined as an estimated glomerular filtration rate <15 mL/min/1.73 m2 or the need for chronic renal replacement therapy. Cox proportional hazard models were used to estimate the hazard ratios (HRs) for the influence of serum iron metabolism on ESRD. Results: During a median follow up of 30.9 months, 66 (59.5%) patients progressed to ESRD. After adjusting for age, sex, baseline systolic blood pressure, renal functions, hemoglobin, HbA1c, and pathological findings, lower serum transferrin concentrations were significantly associated with higher ESRD in multivariate models. Compared with patients in the highest transferrin quartile (≥1.65 g/L), patients in the lowest quartile (≤1.15 g/L) had multivariable-adjusted HR (95% confidence interval) of 7.36 (1.40-38.65) for ESRD. Moreover, tubular epithelial cells in DN exhibited a higher deposition of iron and transferrin expression compared with healthy controls. Conclusions: Low serum transferrin concentration was associated with diabetic ESRD in patients with T2DM. Free iron nephrotoxicity and poor nutritional status with accumulated iron or transferrin deposition might contribute to ESRD.
Collapse
Affiliation(s)
- Lijun Zhao
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China.,Division of General Practice, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yutong Zou
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junlin Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Honghong Ren
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lin Li
- Division of Pathology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ruikun Guo
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jie Zhang
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, Regenerative Medicine Research Center, Chengdu China
| | - Fang Liu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
23
|
Pan S, Qian ZM, Cui S, Zhao D, Lan W, Wang X, Chen X. Local hepcidin increased intracellular iron overload via the degradation of ferroportin in the kidney. Biochem Biophys Res Commun 2019; 522:322-327. [PMID: 31761321 DOI: 10.1016/j.bbrc.2019.11.066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 11/10/2019] [Indexed: 02/09/2023]
Abstract
BACKGROUND Hepcidin is a key regulator of iron homeostasis. Some studies showed that exogenous hepcidin decreased the expression of divalent metal transporter (DMT1) rather than ferroportin(FPN1) to regulate renal iron metabolism. This study explored the effects of hepcidin synthesized by the kidney and its mechanism of iron regulation. METHODS In the in vivo experiments, mice were divided into a unilateral ureter obstruction (UUO) model group and a sham operation group, and mice in the UUO model group were sacrificed on days 1, 3, 5 and 7. The expression of renal hepcidin, FPN1, DMT1 and the retention of renal iron were studied. In the in vitro experiments, we overexpressed hepcidin in HK-2 cells. Then we tested the expression of renal hepcidin, FPN1, DMT1 and observed the production of intracellular ferrous ions. RESULTS Renal hepcidin expression was consistently higher in the UUO group than in the sham group from the first day. The expression of FPN1 gradually decreased, and the expression of DMT1 gradually increased in the UUO model. Intracellular ferrous ions significantly increased on the first day of the UUO model. In hepcidin overexpressed HK-2 cells, the expression of FPN1 was decreased, while the expression of DMT1 has no significant change. In addition, production of intracellular ferrous ions increased. CONCLUSION local hepcidin can regulate iron metabolism in the kidney by adjusting the expression of FPN1.
Collapse
Affiliation(s)
- Sai Pan
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, People's Republic of China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, People's Republic of China
| | - Shaoyuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, People's Republic of China
| | - Delong Zhao
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, People's Republic of China
| | - Weiren Lan
- The Second Affiliated Hospital of Army Medical University, Chongqing, People's Republic of China
| | - Xu Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, People's Republic of China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, People's Republic of China.
| |
Collapse
|
24
|
Abstract
Iron is an essential element that is indispensable for life. The delicate physiological body iron balance is maintained by both systemic and cellular regulatory mechanisms. The iron-regulatory hormone hepcidin assures maintenance of adequate systemic iron levels and is regulated by circulating and stored iron levels, inflammation and erythropoiesis. The kidney has an important role in preventing iron loss from the body by means of reabsorption. Cellular iron levels are dependent on iron import, storage, utilization and export, which are mainly regulated by the iron response element-iron regulatory protein (IRE-IRP) system. In the kidney, iron transport mechanisms independent of the IRE-IRP system have been identified, suggesting additional mechanisms for iron handling in this organ. Yet, knowledge gaps on renal iron handling remain in terms of redundancy in transport mechanisms, the roles of the different tubular segments and related regulatory processes. Disturbances in cellular and systemic iron balance are recognized as causes and consequences of kidney injury. Consequently, iron metabolism has become a focus for novel therapeutic interventions for acute kidney injury and chronic kidney disease, which has fuelled interest in the molecular mechanisms of renal iron handling and renal injury, as well as the complex dynamics between systemic and local cellular iron regulation.
Collapse
|
25
|
Van Avondt K, Nur E, Zeerleder S. Mechanisms of haemolysis-induced kidney injury. Nat Rev Nephrol 2019; 15:671-692. [PMID: 31455889 DOI: 10.1038/s41581-019-0181-0] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
Intravascular haemolysis is a fundamental feature of chronic hereditary and acquired haemolytic anaemias, including those associated with haemoglobinopathies, complement disorders and infectious diseases such as malaria. Destabilization of red blood cells (RBCs) within the vasculature results in systemic inflammation, vasomotor dysfunction, thrombophilia and proliferative vasculopathy. The haemoprotein scavengers haptoglobin and haemopexin act to limit circulating levels of free haemoglobin, haem and iron - potentially toxic species that are released from injured RBCs. However, these adaptive defence systems can fail owing to ongoing intravascular disintegration of RBCs. Induction of the haem-degrading enzyme haem oxygenase 1 (HO1) - and potentially HO2 - represents a response to, and endogenous defence against, large amounts of cellular haem; however, this system can also become saturated. A frequent adverse consequence of massive and/or chronic haemolysis is kidney injury, which contributes to the morbidity and mortality of chronic haemolytic diseases. Intravascular destruction of RBCs and the resulting accumulation of haemoproteins can induce kidney injury via a number of mechanisms, including oxidative stress and cytotoxicity pathways, through the formation of intratubular casts and through direct as well as indirect proinflammatory effects, the latter via the activation of neutrophils and monocytes. Understanding of the detailed pathophysiology of haemolysis-induced kidney injury offers opportunities for the design and implementation of new therapeutic strategies to counteract the unfavourable and potentially fatal effects of haemolysis on the kidney.
Collapse
Affiliation(s)
- Kristof Van Avondt
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany.
| | - Erfan Nur
- Department of Haematology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Sacha Zeerleder
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands. .,Department of Haematology and Central Haematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
26
|
Unthank JL, Ortiz M, Trivedi H, Pelus LM, Sampson CH, Sellamuthu R, Fisher A, Chua HL, Plett A, Orschell CM, Cohen EP, Miller SJ. Cardiac and Renal Delayed Effects of Acute Radiation Exposure: Organ Differences in Vasculopathy, Inflammation, Senescence and Oxidative Balance. Radiat Res 2019; 191:383-397. [PMID: 30901530 DOI: 10.1667/rr15130.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have previously shown significant pathology in the heart and kidney of murine hematopoietic-acute radiation syndrome (H-ARS) survivors of 8.7-9.0 Gy total-body irradiation (TBI). The goal of this study was to determine temporal relationships in the development of vasculopathy and the progression of renal and cardiovascular delayed effects of acute radiation exposure (DEARE) at TBI doses less than 9 Gy and to elucidate the potential roles of senescence, inflammation and oxidative stress. Our results show significant loss of endothelial cells in coronary arteries by 4 months post-TBI (8.53 or 8.72 Gy of gamma radiation). This loss precedes renal dysfunction and interstitial fibrosis and progresses to abnormalities in the arterial media and adventitia and loss of coronary arterioles. Major differences in radiation-induced pathobiology exist between the heart and kidney in terms of vasculopathy progression and also in indices of inflammation, senescence and oxidative imbalance. The results of this work suggest a need for different medical countermeasures for multiple targets in different organs and at various times after acute radiation injury to prevent the progression of DEARE.
Collapse
Affiliation(s)
- Joseph L Unthank
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Miguel Ortiz
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hina Trivedi
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Louis M Pelus
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carol H Sampson
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Rajendran Sellamuthu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alexa Fisher
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hui Lin Chua
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Artur Plett
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christie M Orschell
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Eric P Cohen
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Steven J Miller
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
27
|
Iron uptake by ZIP8 and ZIP14 in human proximal tubular epithelial cells. Biometals 2019; 32:211-226. [PMID: 30806852 PMCID: PMC6437295 DOI: 10.1007/s10534-019-00183-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/14/2018] [Indexed: 12/11/2022]
Abstract
In patients with iron overload disorders, increasing number of reports of renal dysfunction and renal iron deposition support an association between increased iron exposure and renal injury. In systemic iron overload, elevated circulating levels of transferrin-bound (TBI) and non-transferrin-bound iron (NTBI) are filtered to the renal proximal tubules, where they may cause injury. However, the mechanisms of tubular iron handling remain elusive. To unravel molecular renal proximal tubular NTBI and TBI handling, human conditionally immortalized proximal tubular epithelial cells (ciPTECs) were incubated with 55Fe as NTBI and fluorescently labeled holo-transferrin as TBI. Ferrous iron importers ZIP8 and ZIP14 were localized in the ciPTEC plasma membrane. Whereas silencing of either ZIP8 or ZIP14 alone did not affect 55Fe uptake, combined silencing significantly reduced 55Fe uptake compared to control (p < 0.05). Furthermore, transferrin receptor 1 (TfR1) and ZIP14, but not ZIP8, colocalized with early endosome antigen 1 (EEA1). TfR1 and ZIP14 also colocalized with uptake of fluorescently labeled transferrin. Furthermore, ZIP14 silencing decreased 55Fe uptake after 55Fe-Transferrin exposure (p < 0.05), suggesting ZIP14 could be involved in early endosomal transport of TBI-derived iron into the cytosol. Our data suggest that human proximal tubular epithelial cells take up TBI and NTBI, where ZIP8 and ZIP14 are both involved in NTBI uptake, but ZIP14, not ZIP8, mediates TBI-derived iron uptake. This knowledge provides more insights in the mechanisms of renal iron handling and suggests that ZIP8 and ZIP14 could be potential targets for limiting renal iron reabsorption and enhancing urinary iron excretion in systemic iron overload disorders.
Collapse
|
28
|
van Raaij SEG, Rennings AJ, Biemond BJ, Schols SEM, Wiegerinck ETG, Roelofs HMJ, Hoorn EJ, Walsh SB, Nijenhuis T, Swinkels DW, van Swelm RPL. Iron handling by the human kidney: glomerular filtration and tubular reabsorption both contribute to urinary iron excretion. Am J Physiol Renal Physiol 2019; 316:F606-F614. [PMID: 30623722 DOI: 10.1152/ajprenal.00425.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In physiological conditions, circulating iron can be filtered by the glomerulus and is almost completely reabsorbed by the tubular epithelium to prevent urinary iron wasting. Increased urinary iron concentrations have been associated with renal injury. However, it is not clear whether increased urinary iron concentrations in patients are the result of increased glomerular iron filtration and/or insufficient tubular iron reabsorption and if these processes contribute to renal injury. We measured plasma and urine iron parameters and urinary tubular injury markers in healthy human subjects ( n = 20), patients with systemic iron overload ( n = 20), and patients with renal tubular dysfunction ( n = 18). Urinary iron excretion parameters were increased in both patients with systemic iron overload and tubular dysfunction, whereas plasma iron parameters were only increased in patients with systemic iron overload. In patients with systemic iron overload, increased urinary iron levels were associated with elevated circulating iron, as indicated by transferrin saturation (TSAT), and increased body iron, as suggested by plasma ferritin concentrations. In patients with tubular dysfunction, enhanced urinary iron and transferrin excretion were associated with distal tubular injury as indicated by increased urinary glutathione S-transferase pi 1-1 (GSTP1-1) excretion. In systemic iron overload, elevated urinary iron and transferrin levels were associated with increased injury to proximal tubules, indicated by increased urinary kidney injury marker 1 (KIM-1) excretion. Our explorative study demonstrates that both glomerular filtration of elevated plasma iron levels and insufficient tubular iron reabsorption could increase urinary iron excretion and cause renal injury.
Collapse
Affiliation(s)
- Sanne E G van Raaij
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Alexander J Rennings
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Bart J Biemond
- Department of Hematology, Academic Medical Center , Amsterdam , The Netherlands
| | - Saskia E M Schols
- Department of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Erwin T G Wiegerinck
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Hennie M J Roelofs
- Department of Gastroenterology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam , Rotterdam , The Netherlands
| | - Stephen B Walsh
- University College London Centre for Nephrology , London , United Kingdom
| | - Tom Nijenhuis
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Dorine W Swinkels
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Rachel P L van Swelm
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands
| |
Collapse
|
29
|
Palmer WC, Vishnu P, Sanchez W, Aqel B, Riegert-Johnson D, Seaman LAK, Bowman AW, Rivera CE. Diagnosis and Management of Genetic Iron Overload Disorders. J Gen Intern Med 2018; 33:2230-2236. [PMID: 30225768 PMCID: PMC6258594 DOI: 10.1007/s11606-018-4669-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 05/23/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022]
Abstract
Iron overload disorders lead to excess iron deposition in the body, which can occur as a result of genetic or secondary causes. Genetic iron overload, referred to as hereditary hemochromatosis, may present as a common autosomal recessive mutation or as one of several uncommon mutations. Secondary iron overload may result from frequent blood transfusions, exogenous iron intake, or certain hematological diseases such as dyserythropoietic syndrome or chronic hemolytic anemia. Iron overload may be asymptomatic, or may present with significant diseases of the liver, heart, endocrine glands, joints, or other organs. If treated appropriately prior to end-organ damage, life expectancy has been shown to be similar compared to matched populations. Alongside clinical assessment, diagnostic studies involve blood tests, imaging, and in some cases liver biopsy. The mainstay of therapy is periodic phlebotomy, although oral chelation is an option for selected patients.
Collapse
Affiliation(s)
- William C Palmer
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA.
| | - Prakash Vishnu
- Department of Hematology/Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - William Sanchez
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Bashar Aqel
- Department of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, AZ, USA
| | - Doug Riegert-Johnson
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Candido E Rivera
- Department of Hematology/Oncology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
30
|
Shi J, Wu G, Zou X, Jiang K. Enteral Baicalin, a Flavone Glycoside, Reduces Indicators of Cardiac Surgery-Associated Acute Kidney Injury in Rats. Cardiorenal Med 2018; 9:31-40. [DOI: 10.1159/000492159] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/16/2018] [Indexed: 11/19/2022] Open
Abstract
Background/Aims: Cardiac surgery-associated acute kidney injury (CSA-AKI) is one of the most common postoperative complications in intensive care medicine. Baicalin has been shown to have anti-inflammatory and antioxidant roles in various disorders. We aimed to test the protective effects of baicalin on CSA-AKI using a rat model. Methods: Sprague-Dawley rats underwent 75 min of cardiopulmonary bypass (CPB) with 45 min of cardioplegic arrest (CA) to establish the AKI model. Baicalin was administered at different doses intragastrically 1 h before CPB. The control and treated rats were subjected to the evaluation of different kidney injury index and inflammation biomarkers. Results: Baicalin significantly attenuated CPB/CA-induced AKI in rats, as evidenced by the lower levels of serum creatinine, serum NGAL, and Kim1. Baicalin remarkably inhibited oxidative stress, reflected in the decreased malondialdehyde and myeloperoxidase activity, and enhanced superoxide dismutase activity and glutathione in renal tissue. Baicalin suppressed the expression of IL-18 and iNOS, and activated the Nrf2/HO-1 pathway. Conclusion: Our data indicated that baicalin mediated CPB/CA-induced AKI by decreasing the oxidative stress and inflammation in the renal tissues, and that baicalin possesses the potential to be developed as a therapeutic tool in clinical use for CSA-AKI.
Collapse
|
31
|
Liu J, Wang L, Tang X, Fu S, Tian Y, Ma L. Analysis of differentially expressed advanced glycation end product-modified proteins in diabetic rat kidney. Int J Diabetes Dev Ctries 2018. [DOI: 10.1007/s13410-018-0616-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
32
|
van Raaij S, Masereeuw R, Swinkels D, van Swelm R. Inhibition of Nrf2 alters cell stress induced by chronic iron exposure in human proximal tubular epithelial cells. Toxicol Lett 2018; 295:179-186. [DOI: 10.1016/j.toxlet.2018.06.1218] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/18/2018] [Accepted: 06/26/2018] [Indexed: 01/19/2023]
|
33
|
Wang X, Zheng X, Zhang J, Zhao S, Wang Z, Wang F, Shang W, Barasch J, Qiu A. Physiological functions of ferroportin in the regulation of renal iron recycling and ischemic acute kidney injury. Am J Physiol Renal Physiol 2018; 315:F1042-F1057. [PMID: 29923765 DOI: 10.1152/ajprenal.00072.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Renal iron recycling preserves filtered iron from urinary excretion. However, it remains debated whether ferroportin (FPN), the only known iron exporter, is functionally involved in renal iron recycling and whether renal iron recycling is required for systemic iron homeostasis. We deleted FPN in whole nephrons by use of a Nestin-Cre and in the distal nephrons and collecting ducts, using a Ksp-Cre, and investigated its impacts on renal iron recycling and systemic iron homeostasis. FPN deletion by Nestin-Cre, but not by Ksp-Cre, caused excess iron retention and increased ferritin heavy chain (FTH1) specifically in the proximal tubules and resulted in the reduction of serum and hepatic iron. The systemic iron redistribution was aggravated, resulting in anemia and the marked downregulation of hepatic hepcidin in elderly FPN knockout (KO)/Nestin-Cre mice. Similarly, in iron-deficient FPN KO/Nestin-Cre mice, the renal iron retention worsened anemia with the activation of the erythropoietin-erythroferrone-hepcidin pathway and the downregulation of hepatic hepcidin. Hence, FPN likely located at the basolateral membrane of the proximal tubules to export iron into the circulation and was required for renal iron recycling and systemic iron homeostasis particularly in elderly and iron-deficient mice. Moreover, FPN deletion in the proximal tubules alleviated ischemic acute kidney injury, possibly by upregulating FTH1 to limit catalytic iron and by priming antioxidant mechanisms, indicating that FPN could be deleterious in the pathophysiology of ischemic acute kidney injury (AKI) and thus may be a potential target for the prevention and mitigation of ischemic AKI.
Collapse
Affiliation(s)
- Xueqiao Wang
- School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University , Shanghai , China
| | - Xiaoqing Zheng
- School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University , Shanghai , China
| | - Juanlian Zhang
- School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University , Shanghai , China
| | - Shifeng Zhao
- School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University , Shanghai , China
| | - Zhigang Wang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Fudi Wang
- Department of Nutrition, Nutrition Discovery Innovation Center, Institute of Nutrition and Food Safety, School of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , China
| | - Wenjun Shang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University , Zhengzhou , China
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Columbia University , New York, New York
| | - Andong Qiu
- School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University , Shanghai , China.,Department of Laboratory Medicine, The First People's Hospital of Ninghai , Ningbo, China
| |
Collapse
|
34
|
Tubular iron deposition and iron handling proteins in human healthy kidney and chronic kidney disease. Sci Rep 2018; 8:9353. [PMID: 29921869 PMCID: PMC6008459 DOI: 10.1038/s41598-018-27107-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/23/2018] [Indexed: 02/07/2023] Open
Abstract
Iron is suggested to play a detrimental role in the progression of chronic kidney disease (CKD). The kidney recycles iron back into the circulation. However, the localization of proteins relevant for physiological tubular iron handling and their potential role in CKD remain unclear. We examined associations between iron deposition, expression of iron handling proteins and tubular injury in kidney biopsies from CKD patients and healthy controls using immunohistochemistry. Iron was deposited in proximal (PT) and distal tubules (DT) in 33% of CKD biopsies, predominantly in pathologies with glomerular dysfunction, but absent in controls. In healthy kidney, PT contained proteins required for iron recycling including putative iron importers ZIP8, ZIP14, DMT1, iron storage proteins L- and H-ferritin and iron exporter ferroportin, while DT only contained ZIP8, ZIP14, and DMT1. In CKD, iron deposition associated with increased intensity of iron importers (ZIP14, ZIP8), storage proteins (L-, H-ferritin), and/or decreased ferroportin abundance. This demonstrates that tubular iron accumulation may result from increased iron uptake and/or inadequate iron export. Iron deposition associated with oxidative injury as indicated by heme oxygenase-1 abundance. In conclusion, iron deposition is relatively common in CKD, and may result from altered molecular iron handling and may contribute to renal injury.
Collapse
|
35
|
Hönig E, Ringer K, Dewes J, von Mach T, Kamm N, Kreitzer G, Jacob R. Galectin-3 modulates the polarized surface delivery of β1-integrin in epithelial cells. J Cell Sci 2018; 131:jcs.213199. [PMID: 29748377 DOI: 10.1242/jcs.213199] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/30/2018] [Indexed: 12/25/2022] Open
Abstract
Epithelial cells require a precise intracellular transport and sorting machinery to establish and maintain their polarized architecture. This machinery includes β-galactoside-binding galectins for targeting of glycoprotein to the apical membrane. Galectin-3 sorts cargo destined for the apical plasma membrane into vesicular carriers. After delivery of cargo to the apical milieu, galectin-3 recycles back into sorting organelles. We analysed the role of galectin-3 in the polarized distribution of β1-integrin in MDCK cells. Integrins are located primarily at the basolateral domain of epithelial cells. We demonstrate that a minor pool of β1-integrin interacts with galectin-3 at the apical plasma membrane. Knockdown of galectin-3 decreases apical delivery of β1-integrin. This loss is restored by supplementation with recombinant galectin-3 and galectin-3 overexpression. Our data suggest that galectin-3 targets newly synthesized β1-integrin to the apical membrane and promotes apical delivery of β1-integrin internalized from the basolateral membrane. In parallel, knockout of galectin-3 results in a reduction in cell proliferation and an impairment in proper cyst development. Our results suggest that galectin-3 modulates the surface distribution of β1-integrin and affects the morphogenesis of polarized cells.
Collapse
Affiliation(s)
- Ellena Hönig
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Karina Ringer
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg 35043, Germany
| | - Jenny Dewes
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Tobias von Mach
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Natalia Kamm
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany
| | - Geri Kreitzer
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, City College of New York, NY 10031, USA
| | - Ralf Jacob
- Department of Cell Biology and Cell Pathology, Philipps-Universität Marburg, Marburg 35037, Germany .,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg 35043, Germany
| |
Collapse
|
36
|
More than a simple biomarker: the role of NGAL in cardiovascular and renal diseases. Clin Sci (Lond) 2018; 132:909-923. [PMID: 29739822 DOI: 10.1042/cs20171592] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/05/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a small circulating protein that is highly modulated in a wide variety of pathological situations, making it a useful biomarker of various disease states. It is one of the best markers of acute kidney injury, as it is rapidly released after tubular damage. However, a growing body of evidence highlights an important role for NGAL beyond that of a biomarker of renal dysfunction. Indeed, numerous studies have demonstrated a role for NGAL in both cardiovascular and renal diseases. In the present review, we summarize current knowledge concerning the involvement of NGAL in cardiovascular and renal diseases and discuss the various mechanisms underlying its pathological implications.
Collapse
|
37
|
Del Greco M F, Foco L, Pichler I, Eller P, Eller K, Benyamin B, Whitfield JB, Pramstaller PP, Thompson JR, Pattaro C, Minelli C. Serum iron level and kidney function: a Mendelian randomization study. Nephrol Dial Transplant 2018; 32:273-278. [PMID: 28186534 DOI: 10.1093/ndt/gfw215] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/25/2016] [Indexed: 01/01/2023] Open
Abstract
Background Iron depletion is a known consequence of chronic kidney disease (CKD), but there is contradicting epidemiological evidence on whether iron itself affects kidney function and whether its effect is protective or detrimental in the general population. While epidemiological studies tend to be affected by confounding and reverse causation, Mendelian randomization (MR) can provide unconfounded estimates of causal effects by using genes as instruments. Methods We performed an MR study of the effect of serum iron levels on estimated glomerular filtration rate (eGFR), using genetic variants known to be associated with iron. MR estimates of the effect of iron on eGFR were derived based on the association of each variant with iron and eGFR from two large genome-wide meta-analyses on 48 978 and 74 354 individuals. We performed a similar MR analysis for ferritin, which measures iron stored in the body, using variants associated with ferritin. Results A combined MR estimate across all variants showed a 1.3% increase in eGFR per standard deviation increase in iron (95% confidence interval 0.4–2.1%; P = 0.004). The results for ferritin were consistent with those for iron. Secondary MR analyses of the effects of iron and ferritin on CKD did not show significant associations but had very low statistical power. Conclusions Our study suggests a protective effect of iron on kidney function in the general population. Further research is required to confirm this causal association, investigate it in study populations at higher risk of CKD and explore its underlying mechanism of action.
Collapse
Affiliation(s)
- Fabiola Del Greco M
- Center for Biomedicine, European Academy of Bolzano/Bozen (EURAC), affiliated with the University of Lübeck, Via Galvani 31, Bolzano, Italy
| | - Luisa Foco
- Center for Biomedicine, European Academy of Bolzano/Bozen (EURAC), affiliated with the University of Lübeck, Via Galvani 31, Bolzano, Italy
| | - Irene Pichler
- Center for Biomedicine, European Academy of Bolzano/Bozen (EURAC), affiliated with the University of Lübeck, Via Galvani 31, Bolzano, Italy
| | | | | | - Beben Benyamin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - John B Whitfield
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | | | - Peter P Pramstaller
- Center for Biomedicine, European Academy of Bolzano/Bozen (EURAC), affiliated with the University of Lübeck, Via Galvani 31, Bolzano, Italy
| | - John R Thompson
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Cristian Pattaro
- Center for Biomedicine, European Academy of Bolzano/Bozen (EURAC), affiliated with the University of Lübeck, Via Galvani 31, Bolzano, Italy
| | - Cosetta Minelli
- Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College, Emmanuel Kaye Building, 1 Manresa Road, London, UK
| |
Collapse
|
38
|
Nakatani S, Nakatani A, Ishimura E, Toi N, Tsuda A, Mori K, Emoto M, Hirayama Y, Saito A, Inaba M. Urinary Iron Excretion is Associated with Urinary Full-Length Megalin and Renal Oxidative Stress in Chronic Kidney Disease. Kidney Blood Press Res 2018; 43:458-470. [PMID: 29590662 DOI: 10.1159/000488470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/16/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Megalin mediates the uptake of glomerular-filtered iron in the proximal tubules. Urinary full length megalin (C-megalin) excretion has been found to be increased in association with megalin-mediated metabolic load to the endo-lysosomal system in proximal tubular epithelial cells (PTECs) of residual nephrons. In the present study, we investigated the association between urinary iron and C-megalin in chronic kidney disease (CKD) patients, and the possible harmful effect of iron in renal tubules. METHODS Urinary levels of iron and C-megalin were measured in 63 CKD patients using automatic absorption spectrometry and a recently-established sandwich ELISA, respectively. RESULTS Although both urinary C-megalin and urinary total protein levels were correlated with urinary iron (C-megalin: ρ = 0.574, p <0.001; total protein: ρ = 0.500, p <0.001, respectively), urinary C-megalin alone emerged as an independent factor positively associated with urinary iron (β = 0.520, p <0.001) (R2 = 0.75, p <0.001). Furthermore, urinary iron was significantly and positively associated with urinary 8-hydroxydeoxyguanosine, an oxidative stress marker, while no association with other markers of renal tubular injury, i.e., β2-microglobulin and N-acetyl-β-D-glucosaminidase, was noted. CONCLUSIONS Our findings suggest that renal iron handling may be associated with megalin-mediated endo-lysosomal metabolic load in PTECs of residual nephrons and oxidative stress in renal tubules.
Collapse
Affiliation(s)
- Shinya Nakatani
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ayumi Nakatani
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Eiji Ishimura
- Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Norikazu Toi
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akihiro Tsuda
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Katsuhito Mori
- Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masanori Emoto
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yoshiaki Hirayama
- Reagent Research and Development Department, Denka Seiken Co., Ltd., Gosen, Japan
| | - Akihiko Saito
- Department of Applied Molecular Medicine, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masaaki Inaba
- Department of Metabolism, Endocrinology, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Nephrology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
39
|
Wang Y, Bellomo R. Cardiac surgery-associated acute kidney injury: risk factors, pathophysiology and treatment. Nat Rev Nephrol 2017; 13:697-711. [DOI: 10.1038/nrneph.2017.119] [Citation(s) in RCA: 413] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Abstract
Lipocalin 2 (Lcn2), an innate immune protein, has emerged as a critical iron regulatory protein during physiological and inflammatory conditions. As a bacteriostatic factor, Lcn2 obstructs the siderophore iron-acquiring strategy of bacteria and thus inhibits bacterial growth. As part of host nutritional immunity, Lcn2 facilitates systemic, cellular, and mucosal hypoferremia during inflammation, in addition to stabilizing the siderophore-bound labile iron pool. In this review, we summarize recent advances in understanding the interaction between Lcn2 and iron, and its effects in various inflammatory diseases. Lcn2 exerts mostly a protective role in infectious and inflammatory bowel diseases, whereas both beneficial and detrimental functions have been documented in neurodegenerative diseases, metabolic syndrome, renal disorders, skin disorders, and cancer. Further animal and clinical studies are necessary to unveil the multifaceted roles of Lcn2 in iron dysregulation during inflammation and to explore its therapeutic potential for treating inflammatory diseases.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802;
| | - Beng San Yeoh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802;
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802; .,Department of Medicine, The Pennsylvania State University Medical Center, Hershey, Pennsylvania 17033
| |
Collapse
|
41
|
Williams AN, Conway BN. Effect of high density lipoprotein cholesterol on the relationship of serum iron and hemoglobin with kidney function in diabetes. J Diabetes Complications 2017; 31:958-963. [PMID: 28410926 PMCID: PMC5560583 DOI: 10.1016/j.jdiacomp.2017.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/20/2017] [Accepted: 03/25/2017] [Indexed: 01/09/2023]
Abstract
Findings of increased hemoglobin inside the HDL proteome among persons with diabetes who have haptoglobin 2-2 genotype suggest that iron-induced lipid peroxidation may be involved in diabetic kidney disease. We investigated the relationships of serum hemoglobin and iron with kidney function, and whether this varied by level of HDLc, in 5296 adults with and 49,161 without diabetes. Estimated eGFR was our marker of kidney function. Hemoglobin was positively associated with eGFR among those with diabetes and inversely among those without diabetes (interaction p-value <0.0001). Iron was inversely associated with eGFR regardless of diabetes status. When stratified by median HDLc and median hemoglobin, among persons with diabetes mean eGFR was highest in those with high hemoglobin and low HDLc and lowest in those with both low hemoglobin and low HDLc. This divergent relationship was not observed in the non-diabetic population. In contrast to hemoglobin, high iron and low HDLc were associated with a lower mean eGFR regardless of diabetes status. Our data suggest that among persons with diabetes, both hemoglobin and iron are harmful to kidney function at high levels. Our data also suggest that HDLc may play a role in the relationship of high hemoglobin in kidney function in diabetes.
Collapse
Affiliation(s)
- Ashley N Williams
- Department of Epidemiology, West Virginia University, Morgantown, WV
| | - Baqiyyah N Conway
- Department of Epidemiology, West Virginia University, Morgantown, WV.
| |
Collapse
|
42
|
Akintunde JK, Oboh G. Nephritic cell damage and antioxidant status in rats exposed to leachate from battery recycling industry. Interdiscip Toxicol 2017; 9:1-11. [PMID: 28652841 PMCID: PMC5458108 DOI: 10.1515/intox-2016-0001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 03/05/2016] [Accepted: 03/07/2016] [Indexed: 01/09/2023] Open
Abstract
Limited studies have assessed the toxic effect of sub-acute and sub-chronic exposure of leachate (mixture of metals) in mammalian kidney. The sub-acute and sub-chronic exposure of mature male Wistar-strain albino rats (200–220 g) were given by oral administration with leachate from Elewi Odo municipal battery recycling industry (EOMABRIL) for period of 7 and 60 days respectively, at different concentrations (20%, 40%, 60%, 80% and 100%). This was to evaluate its toxic effects on male renal functions using biomarkers of oxidative stress and nephro-cellular damage. Control groups were treated equally, but given distilled water instead of the leachate. All the groups were fed with the same standard food and had free access to drinking water. Following the exposure, results showed that the treatment induced systemic toxicity at the doses tested by causing a significant (p<0.05) alteration in enzymatic antioxidants-catalase (CAT) and superoxide dismutase (SOD) in the kidneys which resulted into elevated levels of malonaldehyde (MDA). Reduced glutathione (GSH) levels were found to be significantly (p<0.05) depleted relative to the control group. Considerable renal cortical congestion and numerous tubules with protein casts were observed in the lumen of EOMABRIL-treated rats. These findings conclude that possible mechanism by which EOMABRIL at the investigated concentrations elicits nephrotoxicity could be linked to the individual, additive, synergistic or antagonistic interactions of this mixture of metals with the renal bio-molecules, alteration of kidney detoxifying enzymes and necrosis of nephritic tubular epithelial cells.
Collapse
Affiliation(s)
- Jacob K Akintunde
- Toxicology and Safety Unit, Department of Environmental Health Sciences, Faculty of Public Health, College of Medicine, University of Ibadan, Ibadan, Nigeria.,Functional Foods, Nutraceuticals and Phytomedicine Research Laboratory, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Ganiyu Oboh
- Functional Foods, Nutraceuticals and Phytomedicine Research Laboratory, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
43
|
Abstract
Iron overload and enhanced hydroxyl radical (•OH) formation have been implicated as the causative factors of oxidative stress in different organs. Both pro-oxidant and anti-oxidant properties have been reported for nitric oxide (NO) in iron-mediated tissue injury. To determine the contribution of NO to iron-induced renal injury, eight groups of rats (eight in each group) were studied as follows: control (normal saline), L-Arg (L-arginine as a substrate of NO synthase, 400 mg/kg), L-NAME (an inhibitor of NO synthase, 8 mg/kg), Fe (iron dextran, 600 mg/kg), DFO (deferroxamine as a chelator of iron, 150 mg/kg), Fe+L-Arg, Fe+L-NAME, DFO+L-Arg. Twenty-four hours after the injections, blood samples were taken and kidneys removed for biochemical analysis. Plasma creatinine and urea were used to stimulate renal function. Renal tissue and plasma vitamin E levels, the most important endogenous fat soluble antioxidant, were measured by HPLC and UV detection. In this study, renal function was markedly reduced in the Fe group compared to controls (creatinine, 1.02± 0.05 mg/dL versus 0.78±0.04 P <0.05; urea, 49.59±1.69 mg/dL versus 40.75±0.86, P <0.01). Vitamin E levels were significantly lower in the Fe group compared to controls (plasma P <0.01; renal tissue P <0.05). Administration of L-Arg to Fe-treated groups prevented these reductions. L-NAME increased iron-induced toxicity significantly, demonstrated by further reduction in the vitamin E levels and renal function compared to the Fe group alone. We concluded that NO plays an important role in protecting the kidney from iron-induced nephrotoxicity. NO synthase blockade enhances iron-mediated renal toxicity in this model.
Collapse
Affiliation(s)
- M Kadkhodaee
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences University, Tehran, Iran.
| | | |
Collapse
|
44
|
Gardner DS, De Brot S, Dunford LJ, Grau-Roma L, Welham SJM, Fallman R, O'Sullivan SE, Oh W, Devonald MAJ. Remote effects of acute kidney injury in a porcine model. Am J Physiol Renal Physiol 2015; 310:F259-71. [PMID: 26608790 DOI: 10.1152/ajprenal.00389.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/19/2015] [Indexed: 01/09/2023] Open
Abstract
Acute kidney injury (AKI) is a common and serious condition with no specific treatment. An episode of AKI may affect organs distant from the kidney, further increasing the morbidity associated with AKI. The mechanism of organ cross talk after AKI is unclear. The renal and immune systems of pigs and humans are alike. Using a preclinical animal (porcine) model, we tested the hypothesis that early effects of AKI on distant organs is by immune cell infiltration, leading to inflammatory cytokine production, extravasation, and edema. In 29 pigs exposed to either sham surgery or renal ischemia-reperfusion (control, n = 12; AKI, n = 17), we assessed remote organ (liver, lung, brain) effects in the short (from 2- to 48-h reperfusion) and longer term (5 wk later) using immunofluorescence (for leukocyte infiltration, apoptosis), a cytokine array, tissue elemental analysis (e.g., electrolytes), blood hematology and chemistry (e.g., liver enzymes), and PCR (for inflammatory markers). AKI elicited significant, short-term (∼24 h) increments in enzymes indicative of acute liver damage (e.g. , AST: ALT ratio; P = 0.02) and influenced tissue biochemistry in some remote organs (e.g., lung tissue [Ca(2+)] increased; P = 0.04). These effects largely resolved after 48 h, and no further histopathology, edema, apoptosis, or immune cell infiltration was noted in the liver, lung, or hippocampus in the short and longer term. AKI has subtle biochemical effects on remote organs in the short term, including a transient increment in markers of acute liver damage. These effects resolved by 48 h, and no further remote organ histopathology, apoptosis, edema, or immune cell infiltration was noted.
Collapse
Affiliation(s)
- David S Gardner
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom;
| | - Simone De Brot
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Louise J Dunford
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom; Renal and Transplant Unit, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; and
| | - Llorenc Grau-Roma
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Simon J M Welham
- School of Biosciences, University of Nottingham, Loughborough, United Kingdom
| | - Rebecca Fallman
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Saoirse E O'Sullivan
- School of Graduate Entry Medicine and Health, Royal Derby Hospital, Derby, United Kingdom
| | - Weng Oh
- Renal and Transplant Unit, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; and
| | - Mark A J Devonald
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom; Renal and Transplant Unit, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; and
| |
Collapse
|
45
|
Liu Y, Templeton DM. Iron-dependent turnover of IRP-1/c-aconitase in kidney cells. Metallomics 2015; 7:766-75. [PMID: 25652229 DOI: 10.1039/c4mt00315b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The kidney plays an important role in iron homeostasis and actively reabsorbs citrate. The bifunctional iron-regulatory protein IRP-1 potentially regulates iron trafficking and participates in citrate metabolism as a cytosolic (c-) aconitase. We investigated the role of cellular iron status in determining the expression and dynamics of IRP-1 in two renal cell types, with the aim of identifying a role of the protein in cellular ROS levels, citrate metabolism and glutamate production. The effects of iron supplementation and chelation on IRP-1 protein and mRNA levels and protein turnover were compared in cultured primary rat mesangial cells and a porcine renal tubule cell line (LLC-PK1). Levels of ROS were measured in both cell types, and c-aconitase activity, glutamate, and glutathione were measured in LLC-PK1 cells, with and without IRP-1 silencing and in glutamine-supplemented or nominally glutamine-free medium. Iron supplementation decreased IRP-1 levels (e.g., approx. 40% in mesangial cells treated with 10 μg ml(-1) iron for 16 h) and increased ubiquitinated IRP-1 levels in both cells types, with iron chelation having the opposite effect. Although iron increased ROS levels (three-fold with 20 μg ml(-1) iron in mesangial cells and more modestly by about 30% with 50 μg ml(-1) in LLC-PK1 cells, both after 24 h), protein degradation was not ROS-dependent. In LLC-PK1 cells, 10 μg ml(-1) iron (24 h) increased both aconitase activity (30%) and secreted glutamate levels (65%). Silencing did not remove the glutamate response to iron but decreased the c-aconitase activity of the residual protein independent of iron loading (37% and 46% of control levels, without and with iron treatment, respectively). However, in glutamine-free medium, glutamate was still increased by iron, even in IRP-1-silenced cells, and did not correspond to c-aconitase. Silencing decreased the amount of ferritin measured in response to iron loading, decreased the affect of iron on total glutathione by 48%, and increased the response of ROS to iron loading by 38%. We conclude that iron increases turnover of IRP-1 in kidney cells, while increasing aconitase activity, suggesting that the apoprotein (aconitase-inactive) form is not exclusively responsible for turnover. Iron increases glutamate levels in tubule epithelial cells, but this appears to be independent of c-aconitase activity or the availability of extracellular glutamine. IRP-1 protein levels are not regulated by ROS, but IRP-1-dependent ferritin expression may decrease ROS and increase total glutathione levels, suggesting that ferritin levels are more important than citrate metabolism in protecting renal cells against iron.
Collapse
Affiliation(s)
- Ying Liu
- University of Toronto, Laboratory Medicine and Pathobiology, 1 King's College Circle, Toronto, Ont. M5S 1A8, Canada.
| | | |
Collapse
|
46
|
Budak H, Kocpinar EF, Gonul N, Ceylan H, Erol HS, Erdogan O. Stimulation of gene expression and activity of antioxidant related enzyme in Sprague Dawley rat kidney induced by long-term iron toxicity. Comp Biochem Physiol C Toxicol Pharmacol 2014; 166:44-50. [PMID: 25038477 DOI: 10.1016/j.cbpc.2014.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022]
Abstract
The trace elements such as iron are vital for various enzyme activities and for other cellular proteins, but iron toxicity causes the production of reactive oxygen species (ROS) that causes alterations in morphology and function of the nephron. The present study was designed to determine the effect of long-term iron overload on the renal antioxidant system and to determine any possible correlation between enzymatic and molecular levels. Our data showed that reduced glutathione (GSH) levels, which is a marker for oxidative stress, strikingly decreased with a long-term iron overload in rat kidney. While renal mRNA levels of glucose 6-phosphate dehydrogenase (G6pd), 6-phosphogluconate dehydrogenase (6pgd) and glutathione peroxidase (Gpx) were significantly affected in the presence of ferric iron, no changes were seen for glutathione reductase (Gsr) and glutathione S-transferases (Gst). While the iron affected the enzymatic activity of G6PD, GSR, GST, and GPX, it had no significant effect on 6PGD activity in the rat kidney. In conclusion, we reported here that the gene expression of G6pd, 6pgd, Gsr, Gpx, and Gst did not correlate to enzyme activity, and the actual effect of long-term iron overload on renal antioxidant system is observed at protein level. Furthermore, the influence of iron on the renal antioxidant system is different from its effect on the hepatic antioxidant system.
Collapse
Affiliation(s)
- Harun Budak
- Atatürk University, Science Faculty, Department of Molecular Biology and Genetics, 25240 Erzurum, Turkey.
| | - Enver Fehim Kocpinar
- Atatürk University, Science Faculty, Department of Chemistry, 25240 Erzurum, Turkey
| | - Nurdan Gonul
- Atatürk University, Science Faculty, Department of Molecular Biology and Genetics, 25240 Erzurum, Turkey
| | - Hamid Ceylan
- Atatürk University, Science Faculty, Department of Molecular Biology and Genetics, 25240 Erzurum, Turkey
| | - Huseyin Serkan Erol
- Atatürk University, Faculty of Veterinary, Department of Biochemistry, 25240 Erzurum, Turkey
| | - Orhan Erdogan
- Atatürk University, Science Faculty, Department of Molecular Biology and Genetics, 25240 Erzurum, Turkey
| |
Collapse
|
47
|
Leaf DE, Rajapurkar M, Lele SS, Mukhopadhyay B, Waikar SS. Plasma catalytic iron, AKI, and death among critically ill patients. Clin J Am Soc Nephrol 2014; 9:1849-56. [PMID: 25189925 DOI: 10.2215/cjn.02840314] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Catalytic iron has been hypothesized to be a key mediator of AKI. However, the association between plasma catalytic iron levels and AKI has not been well studied in humans. DESIGN, SETTINGS, PARTICIPANTS, & MEASUREMENTS A single-center, prospective, nonconsecutive cohort study of 121 critically ill patients admitted to intensive care units (ICUs) between 2008 and 2012 was performed. Plasma catalytic iron, free hemoglobin, and other iron markers were measured on ICU days 1 and 4. The primary end point was in-hospital mortality or AKI requiring RRT. Secondary end points included mortality (assessed during hospitalization, at 30 days, and 1 year) and incident AKI, defined by modified Kidney Disease Improving Global Outcomes criteria. RESULTS ICU day 1 plasma catalytic iron levels were higher among patients who reached the primary end point (median, 0.74 µmol/l [interquartile range, 0.31-3.65] versus 0.29 µmol/l [0.22-0.46]; P<0.01). ICU day 1 plasma catalytic iron levels were associated with number of packed red blood cell transfusions before ICU arrival (rs=0.29; P<0.001) and plasma free hemoglobin levels on ICU day 1 (rs=0.32; P<0.001). Plasma catalytic iron levels on ICU day 1 were significantly associated with in-hospital mortality or AKI requiring RRT, even after adjusting for age, enrollment eGFR, and number of packed red blood cell transfusions before ICU arrival (13 events; adjusted odds ratio per 1-SD higher ln[catalytic iron], 3.33; 95% confidence interval, 1.79 to 6.20). ICU day 1 plasma catalytic iron levels were also significantly associated with incident AKI, RRT, hospital mortality, and 30-day mortality. CONCLUSIONS Among critically ill patients, elevated plasma catalytic iron levels on arrival to the ICU are associated with a greater risk of incident AKI, RRT, and hospital mortality.
Collapse
Affiliation(s)
- David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts; and
| | | | - Suhas S Lele
- Department of Cardiology, Muljibhai Patel Urological Hospital, Gujarat, India
| | | | - Sushrut S Waikar
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts; and
| |
Collapse
|
48
|
Mahadevappa R, Nielsen R, Christensen EI, Birn H. Megalin in acute kidney injury: foe and friend. Am J Physiol Renal Physiol 2013; 306:F147-54. [PMID: 24197071 DOI: 10.1152/ajprenal.00378.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The kidney proximal tubule is a key target in many forms of acute kidney injury (AKI). The multiligand receptor megalin is responsible for the normal proximal tubule uptake of filtered molecules, including nephrotoxins, cytokines, and markers of AKI. By mediating the uptake of nephrotoxins, megalin plays an essential role in the development of some types of AKI. However, megalin also mediates the tubular uptake of molecules implicated in the protection against AKI, and changes in megalin expression have been demonstrated in AKI in animal models. Thus, modulation of megalin expression in response to AKI may be an important part of the tubule cell adaption to cellular protection and regeneration and should be further investigated as a potential target of intervention. This review explores current evidence linking megalin expression and function to the development, diagnosis, and progression of AKI as well as renal protection against AKI.
Collapse
Affiliation(s)
- Ravikiran Mahadevappa
- Dept. of Biomedicine, Aarhus Univ., Wilhelm Meyers Allé 3, Bldg. 1234, Aarhus DK-8000, Denmark.
| | | | | | | |
Collapse
|
49
|
Urinary hepcidin: an inverse biomarker of acute kidney injury after cardiopulmonary bypass? Curr Opin Crit Care 2013; 16:540-4. [PMID: 20736824 DOI: 10.1097/mcc.0b013e32833ecdcc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW In this review, we discuss the potential role of urinary hepcidin, a 2.8-kDa hormonal regulator of iron metabolism, as a biomarker of acute kidney injury (AKI) after cardiopulmonary bypass. RECENT FINDINGS Hepcidin is one of the novel biomarkers of AKI that have been identified using hypothesis-free, proteomic analysis of urine or plasma in patients who develop AKI. Collectively, these markers promise a new era for the early diagnosis and treatment of AKI in the ICU and an understanding of their biological role may also provide mechanistic insights into the pathogenesis of AKI. Although data confirming the association between urinary hepcidin and AKI are as yet limited, we believe hepcidin is of particular interest because hepcidin may be a biomarker specific to cardiopulmonary bypass-associated AKI; as a central regulator of iron metabolism, hepcidin could play a biological role in the pathogenesis of AKI after cardiopulmonary bypass; and hepcidin displays an intriguing negative association with AKI, in that a smaller increase in hepcidin from baseline after cardiopulmonary bypass appears to predict greater chance of developing AKI. SUMMARY Smaller increases in urinary hepcidin, a central regulator of iron metabolism, may be associated with greater risk of AKI after cardiopulmonary bypass. Further research is required to establish the significance and nature of this association.
Collapse
|
50
|
Martines AMF, Masereeuw R, Tjalsma H, Hoenderop JG, Wetzels JFM, Swinkels DW. Iron metabolism in the pathogenesis of iron-induced kidney injury. Nat Rev Nephrol 2013; 9:385-98. [DOI: 10.1038/nrneph.2013.98] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|