1
|
Bose C, Megyesi J, Karaduta O, Singh S, Swaminathan S, Shah S. Iron Chelation Prevents Age-Related Skeletal Muscle Sarcopenia in Klotho Gene Mutant Mice, a Genetic Model of Aging. J Cachexia Sarcopenia Muscle 2025; 16:e13678. [PMID: 39797505 PMCID: PMC11724147 DOI: 10.1002/jcsm.13678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/23/2024] [Accepted: 11/16/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND A decline in skeletal muscle mass and function known as skeletal muscle sarcopenia is an inevitable consequence of aging. Sarcopenia is a major cause of decreased muscle strength, physical frailty and increased muscle fatigability, contributing significantly to an increased risk of physical disability and functional dependence among the elderly. There remains a significant need for a novel therapy that can improve sarcopenia and related problems in aging. Iron accumulation, especially catalytic iron (labile iron) through increased oxidative stress, could be one of the contributing factors to sarcopenia. Our study aimed to examine the effect of an iron chelator on age-related sarcopenia in mice. METHODS We investigated the effect of iron chelation (deferiprone, DFP) in sarcopenia, using mice with klotho deficiency (kl/kl), an established mouse model for aging. Four weeks old Klotho -/- male mice were treated with 25 mg/kg body weight of iron chelator deferiprone in drinking water for 8-14 weeks (n = 12/group, treated and untreated). At the end of the study, gastrocnemius, quadriceps and bicep muscles were dissected and used for western blot and immunohistochemistry analysis, histopathology and iron staining. Serum total iron, catalytic iron and cytokine ELISAs were performed with established methods. RESULTS Treatment with DFP significantly reduced loss of muscle mass in gastrocnemius and quadriceps muscles (p < 0.0001). Total and catalytic iron content of serum and iron in muscles were significantly (both p < 0.0001) lower in the treated animals. The inhibitory factor of myogenesis, the myostatin protein in gastrocnemius muscles (p = 0.019) and serum (p = 0.003) were downregulated after 8 weeks of therapy accompanied by an increased in muscle contractile protein myosin heavy chain (~2.9 folds, p = 0.0004). Treatment decreased inflammation (serum IL6 and TNFα) (p < 0.0001, p = 0.005), respectively, and elevated insulin-like growth factor levels (p = 0.472). This was associated with reduced DNA damage and reduced 8-hydroxy 2 deoxyguanosine in muscle and HO-1 protein (p < 0.001, p = 079), respectively. Significant weight loss (p < 0.001) and decreased water intake (p = 0.012) were observed in untreated mice compared to treatment group. Kaplan-Meier survival curves show the median life span of treated mice was 108 days as compared to 63 days for untreated mice (p = 0.0002). CONCLUSIONS In summary, our research findings indicate that deferiprone reduced age-related sarcopenia in the muscles of Klotho-/- mice. Our finding suggests chelation of excess iron could be an effective therapy to counter sarcopenia. However, additional studies are needed to evaluate and determine the efficacy in humans.
Collapse
Affiliation(s)
- Chhanda Bose
- Central Arkansas Veterans Healthcare SystemLittle RockArkansasUSA
- University of Arkansas for Medical SciencesLittle RockArkansasUSA
- Pharmacology and Neuroscience DepartmentTexas Tech Health Sciences CenterLubbockTexasUSA
| | - Judit Megyesi
- University of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Oleg Karaduta
- Central Arkansas Veterans Healthcare SystemLittle RockArkansasUSA
- University of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of Biochemistry and Molecular BiologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Sharda P. Singh
- Central Arkansas Veterans Healthcare SystemLittle RockArkansasUSA
- University of Arkansas for Medical SciencesLittle RockArkansasUSA
- Internal Medicine DepartmentTexas Tech Health Sciences CenterLubbockTexasUSA
| | - Sundararaman Swaminathan
- Central Arkansas Veterans Healthcare SystemLittle RockArkansasUSA
- University of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Sudhir V. Shah
- Central Arkansas Veterans Healthcare SystemLittle RockArkansasUSA
- University of Arkansas for Medical SciencesLittle RockArkansasUSA
| |
Collapse
|
2
|
Schaer DJ, Schaer CA, Humar R, Vallelian F, Henderson R, Tanaka KA, Levy JH, Buehler PW. Navigating Hemolysis and the Renal Implications of Hemoglobin Toxicity in Cardiac Surgery. Anesthesiology 2024; 141:1162-1174. [PMID: 39159287 PMCID: PMC11560668 DOI: 10.1097/aln.0000000000005109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Acute kidney injury (AKI) affects 20% to 30% of patients undergoing cardiac surgery with cardiopulmonary bypass (CPB). This review synthesizes clinical evidence indicating that CPB-induced hemolysis plays a pivotal role in the development of AKI. The pathogenesis involves cell-free hemoglobin, which triggers oxidative stress, depletes nitric oxide, and incites inflammation, culminating in renal damage. We highlight emerging interventions, including haptoglobin administration, nitric oxide supplementation, and antioxidants, which are promising in reducing the toxicity of cell-free hemoglobin and the incidence of AKI. Current clinical data support the potential efficacy of these treatments. Our analysis concludes that sufficient proof of concept exists to further develop and test these targeted therapies for preventing hemoglobin-induced AKI in patients undergoing CPB. Cardiopulmonary bypass-induced hemolysis is linked to acute kidney injury in cardiac surgery. Emerging therapies targeting cell-free hemoglobin, like haptoglobin, nitric oxide, and antioxidants, show promise in reducing kidney injury, highlighting the need for further research.
Collapse
Affiliation(s)
- Dominik J Schaer
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Christian A Schaer
- Institute of Anesthesiology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Rok Humar
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Florence Vallelian
- Department of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Reney Henderson
- Division of Cardiovascular Anesthesia, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kenichi A Tanaka
- Department of Anesthesiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care and Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Paul W Buehler
- Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, and Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
3
|
Chen W, Zhang H, Shen X, Hong L, Tao H, Xiao J, Nie S, Wei M, Chen M, Zhang C, Yu W. Iron metabolism indexes as predictors of the incidence of cardiac surgery-associated acute kidney surgery. J Cardiothorac Surg 2024; 19:533. [PMID: 39300557 PMCID: PMC11411798 DOI: 10.1186/s13019-024-03080-4] [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: 06/08/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a major complication following cardiac surgery. We explored the clinical utility of iron metabolism indexes for identification of patients at risk for AKI after cardiac surgery. METHODS This prospective observational study included patients who underwent cardiac surgery between March 2023 and June 2023. Iron metabolism indexes were measured upon admission to the intensive care unit. Multivariable logistic regression analyses were performed to explore the relationship between iron metabolism indexes and cardiac surgery-associated AKI (CSA-AKI). Receiver operating characteristic (ROC) curve was used to assess the predictive ability of iron, APACHE II score and the combination of the two indicators. Restricted cubic splines (RCS) was used to further confirm the linear relationship between iron and CSA-AKI. RESULTS Among the 112 recruited patients, 38 (33.9%) were diagnosed with AKI. Multivariable logistic regression analysis indicated that APACHE II score (odds ratio [OR], 1.208; 95% confidence interval [CI], 1.003-1.455, P = 0.036) and iron (OR 1.069; 95% CI 1.009-1.133, P = 0.036) could be used as independent risk factors to predict CSA-AKI. ROC curve analysis showed that iron (area under curve [AUC] = 0.669, 95% CI 0.572-0.757), APACHE II score (AUC = 0.655, 95% CI 0.557-0.744) and iron and APACHE II score combination (AUC = 0.726, 95% CI 0.632-0.807) were predictive indicators for CSA-AKI. RCS further confirmed the linear relationship between iron and CSA-AKI. CONCLUSIONS Elevated iron levels were independently associated with higher risk of CSA-AKI, and there was a linear relationship between iron and CSA-AKI.
Collapse
Affiliation(s)
- Wenxiu Chen
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Hao Zhang
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xiao Shen
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Liang Hong
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Hong Tao
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Jilai Xiao
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Shuai Nie
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Meng Wei
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Ming Chen
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Cui Zhang
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China.
| | - Wenkui Yu
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
4
|
Xie Q, Wang J, Li R, Liu H, Zhong Y, Xu Q, Ge Y, Li C, Sun L, Zhu J. IL-6 signaling accelerates iron overload by upregulating DMT1 in endothelial cells to promote aortic dissection. Int J Biol Sci 2024; 20:4222-4237. [PMID: 39247821 PMCID: PMC11379073 DOI: 10.7150/ijbs.99511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/22/2024] [Indexed: 09/10/2024] Open
Abstract
Aortic dissection (AD), caused by tearing of the intima and avulsion of the aortic media, is a severe threat to patient life and organ function. Iron is closely related to dissection formation and organ injury, but the mechanism of iron ion transport disorder in endothelial cells (ECs) remains unclear. We identified the characteristic EC of dissection with iron overload by single-cell RNA sequencing data. After intersecting iron homeostasis and differentially expressed genes, it was found that hypoxia-inducible factor-1α (HIF-1α) and divalent metal transporter 1 (DMT1) are key genes for iron ion disorder. Subsequently, IL-6R was identified as an essential reason for the JAK-STAT activation, a classical iron regulation pathway, through further intersection and validation. In in vivo and in vitro, both high IL-6 receptor expression and elevated IL-6 levels promote JAK1-STAT3 phosphorylation, leading to increased HIF-1α protein levels. Elevated HIF-1α binds explicitly to the 5'-UTR sequence of the DMT1 gene and transcriptionally promotes DMT1 expression, thereby increasing Fe2+ accumulation and endoplasmic reticulum stress (ERS). Blocking IL-6R and free iron with deferoxamine and tocilizumab significantly prolonged survival and reduced aortic and organ damage in dissection mice. A comparison of perioperative data between AD patients and others revealed that high free iron, IL-6, and ERS levels are characteristics of AD patients and are correlated with prognosis. In conclusion, activated IL-6/JAK1/STAT3 signaling axis up-regulates DMT1 expression by increasing HIF-1α, thereby increasing intracellular Fe2+ accumulation and tissue injury, which suggests a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Qiang Xie
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianji Wang
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Runqiao Li
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yongliang Zhong
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qinfeng Xu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yipeng Ge
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Chengnan Li
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Lizhong Sun
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Junming Zhu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
5
|
Solomon RJ. Acute Kidney Injury Prevention Following Cardiac Catheterization: The Ins and Outs of Management. Kidney Med 2024; 6:100865. [PMID: 39157195 PMCID: PMC11327454 DOI: 10.1016/j.xkme.2024.100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Affiliation(s)
- Richard J. Solomon
- Department of Medicine, Division of Nephrology, University of Vermont, Larner College of Medicine, Burlington, VT
| |
Collapse
|
6
|
Young AM, Strobel RJ, Rotar EP, Kleiman A, McNeil JS, Teman NR, Hawkins RB, Raphael J, Mehaffey JH. Perioperative acetaminophen is associated with reduced acute kidney injury after cardiac surgery. J Thorac Cardiovasc Surg 2024; 167:1372-1380. [PMID: 36207161 DOI: 10.1016/j.jtcvs.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/19/2022] [Accepted: 09/03/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Cardiac surgery-associated acute kidney injury (AKI) is associated with increased postoperative morbidity and mortality. Evidence suggests an association between perioperative acetaminophen administration and decreased incidence of postoperative AKI in pediatric cardiac surgery patients; however, an effect in adults is unknown. METHODS All patients (n = 6192) undergoing coronary and/or valve surgery with a recorded Society of Thoracic Surgeons (STS) risk score at our institution between 2010 and 2018 were stratified by acetaminophen exposure on the day of surgery using institutional pharmacy records. AKI was determined using the Kidney Disease: Improving Global Outcomes (KDIGO) staging criteria. Logistic regression was used to analyze the association between perioperative acetaminophen and postoperative kidney injury or STS major morbidity. A sensitivity analysis using propensity score matching on the STS predicted risk of renal failure and cardiopulmonary bypass time was performed to account for time bias. RESULTS Perioperative acetaminophen exposure was associated with lower odds of stage 1 to 3 acute kidney injury (odds ratio [OR], 0.68; 95% CI, 0.56-0.83; P < .001) and decreased prolonged postoperative ventilation (OR, 0.53; 95% CI, 0.37-0.76; P < .001). A sensitivity analysis provided well-balanced (standard mean difference <0.10) groups of 401 pairs, in which acetaminophen was associated with a decreased incidence of postoperative AKI (OR, 0.7; 95% CI, 0.52-0.94; P = .016). CONCLUSIONS Exposure to acetaminophen on the day of surgery was associated with a decreased incidence of AKI in our patients undergoing cardiac surgery. These data serve as a measure of effect size to further explore the therapeutic potential of acetaminophen to reduce postoperative AKI after cardiac surgery and to elucidate the mechanisms involved.
Collapse
Affiliation(s)
- Andrew M Young
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Raymond J Strobel
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Evan P Rotar
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Amanda Kleiman
- Department of Anesthesiology, University of Virginia, Charlottesville, Va
| | - John S McNeil
- Department of Anesthesiology, University of Virginia, Charlottesville, Va
| | - Nicholas R Teman
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Robert B Hawkins
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, Mich
| | - Jacob Raphael
- Department of Anesthesiology, University of Virginia, Charlottesville, Va
| | - J Hunter Mehaffey
- Department of Cardiovascular and Thoracic Surgery, West Virginia University, Morgantown, WVa.
| |
Collapse
|
7
|
Lücht J, Seiler R, Herre AL, Brankova L, Fritsche-Guenther R, Kirwan J, Huscher D, Münzfeld H, Berger F, Photiadis J, Tong G, Schmitt KRL. Promising results of a clinical feasibility study: CIRBP as a potential biomarker in pediatric cardiac surgery. Front Cardiovasc Med 2024; 11:1247472. [PMID: 38361581 PMCID: PMC10867162 DOI: 10.3389/fcvm.2024.1247472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024] Open
Abstract
Objective Cold-inducible RNA binding Protein (CIRBP) has been shown to be a potent inflammatory mediator and could serve as a novel biomarker for inflammation. Systemic inflammatory response syndrome (SIRS) and capillary leak syndrome (CLS) are frequent complications after pediatric cardiac surgery increasing morbidity, therefore early diagnosis and therapy is crucial. As CIRBP serum levels have not been analyzed in a pediatric population, we conducted a clinical feasibility establishing a customized magnetic bead panel analyzing CIRBP in pediatric patients undergoing cardiac surgery. Methods A prospective hypothesis generating observational clinical study was conducted at the German Heart Center Berlin during a period of 9 months starting in May 2020 (DRKS00020885, https://drks.de/search/de/trial/DRKS00020885). Serum samples were obtained before the cardiac operation, upon arrival at the pediatric intensive care unit, 6 and 24 h after the operation in patients up to 18 years of age with congenital heart disease (CHD). Customized multiplex magnetic bead-based immunoassay panels were developed to analyze CIRBP, Interleukin-1β (IL-1β), Interleukin-6 (IL-6), Interleukin-8 (IL-8), Interleukin-10 (IL-10), Monocyte chemotactic protein 1 (MCP-1), Syndecan-1 (SDC-1), Thrombomodulin (TM), Vascular endothelial growth factor (VEGF-A), Angiopoietin-2 (Ang-2), and Fibroblast growth factor 23 (FGF-23) in 25 µl serum using the Luminex MagPix® system. Results 19 patients representing a broad range of CHD (10 male patients, median age 2 years, 9 female patients, median age 3 years) were included in the feasibility study. CIRBP was detectable in the whole patient cohort. Relative to individual baseline values, CIRBP concentrations increased 6 h after operation and returned to baseline levels over time. IL-6, IL-8, IL-10, and MCP-1 concentrations were significantly increased after operation and except for MCP-1 concentrations stayed upregulated over time. SDC-1, TM, Ang-2, as well as FGF-23 concentrations were also significantly increased, whereas VEGF-A concentration was significantly decreased after surgery. Discussion Using customized magnetic bead panels, we were able to detect CIRBP in a minimal serum volume (25 µl) in all enrolled patients. To our knowledge this is the first clinical study to assess CIRBP serum concentrations in a pediatric population.
Collapse
Affiliation(s)
- Jana Lücht
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Raphael Seiler
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Alexa Leona Herre
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Liliya Brankova
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Raphaela Fritsche-Guenther
- Metabolomics Platform, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Jennifer Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Dörte Huscher
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hanna Münzfeld
- Department of Radiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Felix Berger
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Joachim Photiadis
- Department of Congenital Heart Surgery and Pediatric Heart Surgery, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Giang Tong
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Katharina R. L. Schmitt
- Department of Congenital Heart Disease/Pediatric Cardiology, Deutsches Herzzentrum der Charité – Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| |
Collapse
|
8
|
Grange C, Lux F, Brichart T, David L, Couturier A, Leaf DE, Allaouchiche B, Tillement O. Iron as an emerging therapeutic target in critically ill patients. Crit Care 2023; 27:475. [PMID: 38049866 PMCID: PMC10694984 DOI: 10.1186/s13054-023-04759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/24/2023] [Indexed: 12/06/2023] Open
Abstract
The multiple roles of iron in the body have been known for decades, particularly its involvement in iron overload diseases such as hemochromatosis. More recently, compelling evidence has emerged regarding the critical role of non-transferrin bound iron (NTBI), also known as catalytic iron, in the care of critically ill patients in intensive care units (ICUs). These trace amounts of iron constitute a small percentage of the serum iron, yet they are heavily implicated in the exacerbation of diseases, primarily by catalyzing the formation of reactive oxygen species, which promote oxidative stress. Additionally, catalytic iron activates macrophages and facilitates the growth of pathogens. This review aims to shed light on this underappreciated phenomenon and explore the various common sources of NTBI in ICU patients, which lead to transient iron dysregulation during acute phases of disease. Iron serves as the linchpin of a vicious cycle in many ICU pathologies that are often multifactorial. The clinical evidence showing its detrimental impact on patient outcomes will be outlined in the major ICU pathologies. Finally, different therapeutic strategies will be reviewed, including the targeting of proteins involved in iron metabolism, conventional chelation therapy, and the combination of renal replacement therapy with chelation therapy.
Collapse
Affiliation(s)
- Coralie Grange
- MexBrain, 13 Avenue Albert Einstein, Villeurbanne, France
- Institut Lumière-Matière, UMR 5306, Université Claude Bernard Lyon1-CNRS, Villeurbanne Cedex, France
| | - François Lux
- Institut Lumière-Matière, UMR 5306, Université Claude Bernard Lyon1-CNRS, Villeurbanne Cedex, France.
- Institut Universitaire de France (IUF), 75231, Paris, France.
| | | | - Laurent David
- Institut National des Sciences Appliquées, CNRS UMR 5223, Ingénierie des Matériaux Polymères, Univ Claude Bernard Lyon 1, Université Jean Monnet, 15 bd Latarjet, 69622, Villeurbanne, France
| | - Aymeric Couturier
- MexBrain, 13 Avenue Albert Einstein, Villeurbanne, France
- Nephrology, American Hospital of Paris, Paris, France
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bernard Allaouchiche
- University of Lyon, University Lyon I Claude Bernard, APCSe VetAgro Sup UP, 2021. A10, Marcy L'Étoile, France
| | - Olivier Tillement
- Institut Lumière-Matière, UMR 5306, Université Claude Bernard Lyon1-CNRS, Villeurbanne Cedex, France
| |
Collapse
|
9
|
Cheruku SR, Raphael J, Neyra JA, Fox AA. Acute Kidney Injury after Cardiac Surgery: Prediction, Prevention, and Management. Anesthesiology 2023; 139:880-898. [PMID: 37812758 PMCID: PMC10841304 DOI: 10.1097/aln.0000000000004734] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Acute kidney injury (AKI) is a common complication in cardiac surgery patients, with a reported incidence of 20 to 30%. The development of AKI is associated with worse short- and long-term mortality, and longer hospital length of stay. The pathogenesis of cardiac surgery-associated AKI is poorly understood but likely involves an interplay between preoperative comorbidities and perioperative stressors. AKI is commonly diagnosed by using increases in serum creatinine or decreased urine output and staged using a standardized definition such as the Kidney Disease Improving Global Outcomes classification. Novel biomarkers under investigation may provide earlier detection and better prediction of AKI, enabling mitigating therapies early in the perioperative period. Recent clinical trials of cardiac surgery patients have demonstrated the benefit of goal-directed oxygen delivery, avoidance of hyperthermic perfusion and specific fluid and medication strategies. This review article highlights both advances and limitations regarding the prevention, prediction, and treatment of cardiac surgery-associated AKI.
Collapse
Affiliation(s)
- Sreekanth R Cheruku
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jacob Raphael
- Department of Anesthesiology and Perioperative Medicine, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Javier A Neyra
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amanda A Fox
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, Dallas, Texas; McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
10
|
Grange C, Aigle A, Ehrlich V, Salazar Ariza JF, Brichart T, Da Cruz-Boisson F, David L, Lux F, Tillement O. Design of a water-soluble chitosan-based polymer with antioxidant and chelating properties for labile iron extraction. Sci Rep 2023; 13:7920. [PMID: 37193699 DOI: 10.1038/s41598-023-34251-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/26/2023] [Indexed: 05/18/2023] Open
Abstract
Loosely bound iron, due to its contribution to oxidative stress and inflammation, has become an important therapeutic target for many diseases. A water-soluble chitosan-based polymer exhibiting both antioxidant and chelating properties due to the dual functionalization with DOTAGA and DFO has been developed to extract this iron therefore preventing its catalytic production of reactive oxygen species. This functionalized chitosan was shown to have stronger antioxidant properties compared to conventional chitosan, improved iron chelating properties compared to the clinical therapy, deferiprone, and provided promising results for its application and improved metal extraction within a conventional 4 h hemodialysis session with bovine plasma.
Collapse
Affiliation(s)
- Coralie Grange
- MexBrain, 13 avenue Albert Einstein, Villeurbanne, France
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France
| | - Axel Aigle
- MexBrain, 13 avenue Albert Einstein, Villeurbanne, France
| | - Victor Ehrlich
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France
| | - Juan Felipe Salazar Ariza
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France
- Ingénierie des Matériaux Polymères, CNRS UMR 5223, Univ Claude Bernard Lyon 1, Institut national des Sciences Appliquées, Université Jean Monnet, Univ Lyon, 15 bd Latarjet, 69622, Villeurbanne, France
- Institut Universitaire de France (IUF), 75231, Paris, France
| | | | - Fernande Da Cruz-Boisson
- Ingénierie des Matériaux Polymères, CNRS UMR 5223, Univ Claude Bernard Lyon 1, Institut national des Sciences Appliquées, Université Jean Monnet, Univ Lyon, 15 bd Latarjet, 69622, Villeurbanne, France
| | - Laurent David
- Ingénierie des Matériaux Polymères, CNRS UMR 5223, Univ Claude Bernard Lyon 1, Institut national des Sciences Appliquées, Université Jean Monnet, Univ Lyon, 15 bd Latarjet, 69622, Villeurbanne, France
| | - François Lux
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France.
- Institut Universitaire de France (IUF), 75231, Paris, France.
| | - Olivier Tillement
- Institut Lumière-Matière, UMR 5306, Université Lyon1-CNRS, Université de Lyon, Villeurbanne Cedex, France
| |
Collapse
|
11
|
Czempik PF, Wiórek A. Iron deficiency in sepsis patients managed with divided doses of iron dextran: a prospective cohort study. Sci Rep 2023; 13:5264. [PMID: 37002279 PMCID: PMC10066317 DOI: 10.1038/s41598-023-32002-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Iron deficiency (ID) impairs hemoglobin (Hb) synthesis and immune function, both crucial for sepsis patients. We assessed the impact of iron dextran on reticulocyte (Ret) Hb equivalent (Ret-He) and Ret subpopulations in iron-deficient sepsis patients. In this prospective clinical study we enrolled patients with sepsis or septic shock with procalcitonin concentration > 0.5 ng/mL, diagnosed with ID based on Ret-He. Study subjects received divided doses of iron dextran until normalization of Ret-He. The study population included 35 subjects. The median Ret-He increase after 2 doses of iron dextran was 3.0 (IQR 1.9-6.1) pg (p < 0.01) with median time to normalization 4 (IQR 3-5) days. Although no change in Ret percentage [Me 1.5 (IQR 1.1-2.1) vs. Me 1.4 (IQR 1.1-2.4) %, p = 0.39] and number [Me 0.05 (IQR 0.04-0.07) vs. Me 0.05 (IQR 0.03-0.06) 106/µL, p = 0.88] was noted, Ret subpopulations changed significantly (p for all < 0.01). Divided doses of iron dextran relatively quickly normalize Ret-He in iron-deficient sepsis patients. Changes in Ret subpopulations suggest increased erythropoietic activity. Further research is needed to explore the role of intravenous iron in this clinical setting.
Collapse
Affiliation(s)
- Piotr F Czempik
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, 40-752, Katowice, Poland.
- Transfusion Committee, University Clinical Center of Medical University of Silesia in Katowice, Katowice, Poland.
| | - Agnieszka Wiórek
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, 40-752, Katowice, Poland
| |
Collapse
|
12
|
Kosenko E, Tikhonova L, Alilova G, Montoliu C. Erythrocytes Functionality in SARS-CoV-2 Infection: Potential Link with Alzheimer's Disease. Int J Mol Sci 2023; 24:5739. [PMID: 36982809 PMCID: PMC10051442 DOI: 10.3390/ijms24065739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a rapidly spreading acute respiratory infection caused by SARS-CoV-2. The pathogenesis of the disease remains unclear. Recently, several hypotheses have emerged to explain the mechanism of interaction between SARS-CoV-2 and erythrocytes, and its negative effect on the oxygen-transport function that depends on erythrocyte metabolism, which is responsible for hemoglobin-oxygen affinity (Hb-O2 affinity). In clinical settings, the modulators of the Hb-O2 affinity are not currently measured to assess tissue oxygenation, thereby providing inadequate evaluation of erythrocyte dysfunction in the integrated oxygen-transport system. To discover more about hypoxemia/hypoxia in COVID-19 patients, this review highlights the need for further investigation of the relationship between biochemical aberrations in erythrocytes and oxygen-transport efficiency. Furthermore, patients with severe COVID-19 experience symptoms similar to Alzheimer's, suggesting that their brains have been altered in ways that increase the likelihood of Alzheimer's. Mindful of the partly assessed role of structural, metabolic abnormalities that underlie erythrocyte dysfunction in the pathophysiology of Alzheimer's disease (AD), we further summarize the available data showing that COVID-19 neurocognitive impairments most probably share similar patterns with known mechanisms of brain dysfunctions in AD. Identification of parameters responsible for erythrocyte function that vary under SARS-CoV-2 may contribute to the search for additional components of progressive and irreversible failure in the integrated oxygen-transport system leading to tissue hypoperfusion. This is particularly relevant for the older generation who experience age-related disorders of erythrocyte metabolism and are prone to AD, and provide an opportunity for new personalized therapies to control this deadly infection.
Collapse
Affiliation(s)
- Elena Kosenko
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Lyudmila Tikhonova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Gubidat Alilova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Carmina Montoliu
- Hospital Clinico Research Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain
- Pathology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
13
|
Chen S, Zhang C, Zhong Y, Tang B, Xie Q, Guo R, Qiao Z, Li C, Ge Y, Zhu J. Association between preoperative serum myoglobin and acute kidney injury after Stanford Type A aortic dissection surgery. Clin Chim Acta 2023; 541:117232. [PMID: 36690242 DOI: 10.1016/j.cca.2023.117232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/03/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication after Type A aortic dissection (TAAD) surgery, and it is associated with poor outcomes. The nephrotoxic effect of myoglobin was established, but its correlation with AKI following TAAD repair still lacks sufficient evidence. We clarified the correlation between preoperative serum myoglobin (pre-sMyo) concentrations and AKI after TAAD surgery. METHOD A retrospective analysis was performed on the perioperative data of 382 patients treated with TAAD surgery at Beijing Anzhen Hospital. AKI was defined and classified according to the criteria established by the Kidney Disease: Improving Global Outcomes Acute Kidney Injury Work Group. We attempted to determine the correlation between pre-sMyo concentrations and postoperative AKI. RESULTS The incidences of Stage 1, 2, and 3 AKI were 37.3 % (57/153), 23.5 % (36/153), and 39.2 % (60/153), respectively. The pre-sMyo concentrations of the AKI group were significantly increased than the non-AKI group [43.1 (21.4, 107.5) vs 26.4 (18.0, 37.2), P < 0.001]. Pre-sMyo concentrations have a linear correlation with preoperative renal function-related indicators. The multivariable logistic regression analysis showed that Ln (pre-sMyo) was an independent risk factor for AKI. When the pre-sMyo concentration was at the fourth quartile [109.3 (64.8, 213.4) ng/ml], the risk of developing any-stage and severe AKI was significantly increased (OR = 4.333, 95 % CI: 2.364-7.943, P < 0.001; OR = 3.862, 95 %, CI: 2.011-7.419, P < 0.001). This difference persisted after adjustment (OR = 3.830, 95 % CI: 1.848-7.936, P < 0.001; OR = 2.330, 95 % CI: 1.045-5.199, P = 0.039). Furthermore, pre-sMyo concentrations were not affected by lower limb malperfusion, myocardial malperfusion, and cardiac tamponade. CONCLUSIONS Increased pre-sMyo concentrations correlated with postoperative AKI in TAAD, which may increase the risk of developing any-stage AKI and severe AKI after TAAD surgery.
Collapse
Affiliation(s)
- Suwei Chen
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenhan Zhang
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yongliang Zhong
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Bing Tang
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Qiang Xie
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Rutao Guo
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhiyu Qiao
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengnan Li
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yipeng Ge
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Junming Zhu
- Department of Cardiovascular Surgery, Beijing Aortic Disease Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Boettcher W, O'Brien B, Photiadis J, Habazettl H, Eggert-Doktor D. Acute Kidney Injury With a Miniaturized Extracorporeal Circuit for Neonatal Cardiopulmonary Bypass. J Cardiothorac Vasc Anesth 2022; 36:4045-4053. [PMID: 36008209 DOI: 10.1053/j.jvca.2022.06.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVES The objectives of this study were to evaluate the incidence and to identify risk factors for acute kidney injury (AKI) in neonates undergoing cardiopulmonary bypass (CPB) with a miniaturized bloodless primed extracorporeal circuit. DESIGN A retrospective cohort study. SETTING A single-center, tertiary academic hospital. PARTICIPANTS Data of 462 patients were analyzed. INTERVENTIONS With a retrospective analysis of neonates undergoing CPB with bloodless priming between May 2007 and August 2019, the incidence of AKI was determined according to the neonatal Kidney Disease: Improving Global Outcomes classification. Multivariate logistic regression analyses were performed to determine risk factors for AKI. MEASUREMENTS AND MAIN RESULTS The incidence of AKI was 41.1% (190 of 462); 30.3% (n = 140) had mild stage 1, 6.5% (n = 30) reached stage 2, and 4.3% (n = 20) reached stage 3. Multivariate logistic regression showed that degree of hypothermia (p = 0.05), duration of CPB (p = 0.03), and lower baseline serum creatinine (p < 0.001) were associated independently with AKI. In the authors' patient population, patients without transfusion of donor-derived erythrocytes had a lower incidence of AKI (p = 0.003). AKI stages 2 and 3 were associated with longer duration of mechanical ventilation (p = 0.008) and increased length of stay in the intensive care unit (p = 0.03). CONCLUSIONS With a miniaturized CPB circuit and bloodless priming, the AKI incidence was well within the range consistent with previously reported studies from other institutions.
Collapse
Affiliation(s)
- Wolfgang Boettcher
- Department of Congenital Heart Surgery and Pediatric Heart Surgery, German Heart Center Berlin, Berlin, Germany.
| | - Benjamin O'Brien
- Department of Cardiac Anesthesiology and Intensive Care Medicine, German Heart Center Berlin, Berlin, Germany; Department of Cardiac Anesthesiology and Intensive Care Medicine, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Joachim Photiadis
- Department of Congenital Heart Surgery and Pediatric Heart Surgery, German Heart Center Berlin, Berlin, Germany
| | - Helmut Habazettl
- Department of Cardiac Anesthesiology and Intensive Care Medicine, German Heart Center Berlin, Berlin, Germany; Institute of Physiology, Charité - Universitaetsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dirk Eggert-Doktor
- Department of Cardiac Anesthesiology and Intensive Care Medicine, German Heart Center Berlin, Berlin, Germany
| |
Collapse
|
15
|
Scott JP, Tanem JM, Tomita-Mitchell A, Hoffman GM, Niebler RA, Liang HL, Simpson PM, Stamm KD, North PE, Mitchell ME. Elevated nuclear and mitochondrial cell-free deoxyribonucleic acid measurements are associated with death after infant cardiac surgery. J Thorac Cardiovasc Surg 2022; 164:367-375. [PMID: 35144816 DOI: 10.1016/j.jtcvs.2021.10.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVES Mortality rates following pediatric cardiac surgery with cardiopulmonary bypass have declined over decades, but have plateaued in recent years. This is in part attributable to persistent issues with postoperative global inflammation and myocardial dysfunction, commonly manifested by systemic inflammatory response syndrome and low cardiac output syndrome, respectively. Quantified cell-free DNA (cfDNA), of nuclear or mitochondrial origin, has emerged as a biomarker for both inflammation and myocardial injury. Recent data suggest that nuclear cfDNA (ncfDNA) may quantify inflammation, whereas mitochondrial cfDNA (mcfDNA) may correlate with the degree of myocardial injury. We hypothesize that threshold levels of ncfDNA and mcfDNA can be established that are sensitive and specific for postoperative mortality mediated through independent pathways, and that association will be enhanced with combined analysis. METHODS Prospective observational study of infants younger than age 1 year undergoing planned surgery with cardiopulmonary bypass. The study received institutional review board approval. Samples were drawn before skin incision, immediately after completion of cardiopulmonary bypass, and subsequently at predetermined intervals postoperatively. Association of early postoperative ncfDNA and mcfDNA levels with mortality were assessed by logistic regression with cut-points chosen by receiving operating characteristic curve exploration. RESULTS Data were available in 59 patients. Median age and weight were 122 days (interquartile range, 63-154 days) and 4.9 kg (interquartile range, 3.9-6.2 kg). Median STAT category was 3 (interquartile range, 1-4). The primary outcome of death was met in 3 out of 59 (5%). Combined analysis of ncfDNA and mcfDNA levels at 12 hours after the initiation of cardiopulmonary bypass with death at a threshold of 50 ng/mL ncfDNA and 17 copies/μL mcfDNA yielded 100% sensitivity and negative predictive value. The specificity (91%) and positive predictive value (38%) increased through combined analysis compared with univariate analysis. Combined analysis exhibited high specificity (93%) and negative predictive value (78%) for prolonged (>30 postoperative days) hospitalization. CONCLUSIONS Combined analysis of early postoperative ncfDNA and mcfDNA can stratify risk of mortality and prolonged hospitalization following infant cardiac surgery. Evaluation of both ncfDNA and mcfDNA to identify states of generalized inflammation and myocardial injury may allow for targeted interventions and improved outcomes.
Collapse
Affiliation(s)
- John P Scott
- Division of Pediatric Anesthesiology, Department of Anesthesiology, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis; Division of Pediatric Critical Medicine, Department of Pediatrics, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis.
| | - Justinn M Tanem
- Division of Pediatric Anesthesiology, Department of Anesthesiology, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis; Division of Pediatric Critical Medicine, Department of Pediatrics, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - Aoy Tomita-Mitchell
- Division of Pediatric Cardiothoracic Surgery, Department of Cardiothoracic Surgery, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - George M Hoffman
- Division of Pediatric Anesthesiology, Department of Anesthesiology, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis; Division of Pediatric Critical Medicine, Department of Pediatrics, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - Robert A Niebler
- Division of Pediatric Critical Medicine, Department of Pediatrics, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - Huan Ling Liang
- Division of Pediatric Cardiothoracic Surgery, Department of Cardiothoracic Surgery, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - Pippa M Simpson
- Division of Quantitative Health Sciences, Department of Pediatrics, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - Karl D Stamm
- Division of Pediatric Cardiothoracic Surgery, Department of Cardiothoracic Surgery, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - Paula E North
- Division of Pediatric Pathology, Department of Pathology, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| | - Michael E Mitchell
- Division of Pediatric Cardiothoracic Surgery, Department of Cardiothoracic Surgery, Herma Heart Institute, Children's Wisconsin, Medical College of Wisconsin, Milwaukee, Wis
| |
Collapse
|
16
|
Van Coillie S, Van San E, Goetschalckx I, Wiernicki B, Mukhopadhyay B, Tonnus W, Choi SM, Roelandt R, Dumitrascu C, Lamberts L, Dams G, Weyts W, Huysentruyt J, Hassannia B, Ingold I, Lele S, Meyer E, Berg M, Seurinck R, Saeys Y, Vermeulen A, van Nuijs ALN, Conrad M, Linkermann A, Rajapurkar M, Vandenabeele P, Hoste E, Augustyns K, Vanden Berghe T. Targeting ferroptosis protects against experimental (multi)organ dysfunction and death. Nat Commun 2022; 13:1046. [PMID: 35210435 PMCID: PMC8873468 DOI: 10.1038/s41467-022-28718-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/07/2022] [Indexed: 12/26/2022] Open
Abstract
The most common cause of death in the intensive care unit (ICU) is the development of multiorgan dysfunction syndrome (MODS). Besides life-supporting treatments, no cure exists, and its mechanisms are still poorly understood. Catalytic iron is associated with ICU mortality and is known to cause free radical-mediated cellular toxicity. It is thought to induce excessive lipid peroxidation, the main characteristic of an iron-dependent type of cell death conceptualized as ferroptosis. Here we show that the severity of multiorgan dysfunction and the probability of death are indeed associated with plasma catalytic iron and lipid peroxidation. Transgenic approaches underscore the role of ferroptosis in iron-induced multiorgan dysfunction. Blocking lipid peroxidation with our highly soluble ferrostatin-analogue protects mice from injury and death in experimental non-septic multiorgan dysfunction, but not in sepsis-induced multiorgan dysfunction. The limitations of the experimental mice models to mimic the complexity of clinical MODS warrant further preclinical testing. In conclusion, our data suggest ferroptosis targeting as possible treatment option for a stratifiable subset of MODS patients.
Collapse
Affiliation(s)
- Samya Van Coillie
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Emily Van San
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ines Goetschalckx
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Bartosz Wiernicki
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Banibrata Mukhopadhyay
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Wulf Tonnus
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus, the Technische Universität Dresden, Dresden, Germany
| | - Sze Men Choi
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ria Roelandt
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Catalina Dumitrascu
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Ludwig Lamberts
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Geert Dams
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wannes Weyts
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
| | - Jelle Huysentruyt
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Behrouz Hassannia
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Irina Ingold
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Suhas Lele
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Evelyne Meyer
- Department of Pharmacology, Toxicology and Biochemistry, Ghent University, Merelbeke, Belgium
| | - Maya Berg
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Ruth Seurinck
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Yvan Saeys
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - An Vermeulen
- Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Alexander L N van Nuijs
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,National Research Medical University, Laboratory of Experimental Oncology, Moscow, Russia
| | - Andreas Linkermann
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus, the Technische Universität Dresden, Dresden, Germany.,Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Mohan Rajapurkar
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Methusalem program, Ghent University, Ghent, Belgium
| | - Eric Hoste
- Intensive Care Unit, Ghent University Hospital; Ghent University, Ghent, Belgium
| | - Koen Augustyns
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium. .,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
17
|
Cardiac Surgery Associated AKI Prevention Strategies and Medical Treatment for CSA-AKI. J Clin Med 2021; 10:jcm10225285. [PMID: 34830567 PMCID: PMC8618011 DOI: 10.3390/jcm10225285] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/29/2022] Open
Abstract
Acute kidney injury (AKI) is common after cardiac surgery. To date, there are no specific pharmacological therapies. In this review, we summarise the existing evidence for prevention and management of cardiac surgery-associated AKI and outline areas for future research. Preoperatively, angiotensin-converting enzyme inhibitors and angiotensin receptor blockers should be withheld and nephrotoxins should be avoided to reduce the risk. Intraoperative strategies include goal-directed therapy with individualised blood pressure management and administration of balanced fluids, the use of circuits with biocompatible coatings, application of minimally invasive extracorporeal circulation, and lung protective ventilation. Postoperative management should be in accordance with current KDIGO AKI recommendations.
Collapse
|
18
|
Feasibility study and direct extraction of endogenous free metallic cations combining hemodialysis and chelating polymer. Sci Rep 2021; 11:19948. [PMID: 34620952 PMCID: PMC8497614 DOI: 10.1038/s41598-021-99462-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/21/2021] [Indexed: 11/29/2022] Open
Abstract
In this article, we report the conception and the use of dialysis-based medical device for the extraction of metals. The medical device is obtained by addition in the dialysate of a functionalized chitosan that can chelate endogenous metals like iron or copper. This water-soluble functionalized chitosan is obtained after controlled reacetylation and grafting of DOTAGA. Due to the high mass of chitosan, the polymer cannot cross through the membrane and the metals are trapped in the dialysate during hemodialysis. Copper extraction has been evaluated in vitro using an hemodialysis protocol. Feasibility study has been performed on healthy sheep showing no acute toxicity througout the entire dialysis procedure and first insights of metallic extraction even on healthy animals.
Collapse
|
19
|
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
|
20
|
Abstract
Significance: Iron is an essential element required for growth and proper functioning of the body. However, an excess of labile ferrous iron increases the risk of oxidative stress-induced injury due to the high reactivity of the unpaired reactive electrons of both ferrous iron and oxygen. This high reactivity can be exemplified in the outside world by one of its consequences, rust formation. In cells, this redox-active iron is involved in the formation of lipid radicals. Recent Advances: Defect or insufficient membrane-protective mechanisms can result in iron-catalyzed excessive lipid peroxidation and subsequent cell death, now conceptualized as ferroptosis. Growing reports propose the detrimental role of iron and ferroptosis in many experimental disease models such as ischemia-reperfusion, acute and chronic organ injuries. Critical Issues: This review first provides a snapshot of iron metabolism, followed by a brief introduction of the molecular mechanisms of ferroptosis, as an iron-dependent lipid peroxidation-driven mode of cell death. Upon describing how iron dysbiosis affects ferroptosis induction, we elaborate on the detrimental role of the iron-ferroptosis axis in several diseases. Future Directions: Despite compelling findings suggesting a role of ferroptosis in experimental animal models, the exact contribution of ferroptosis in human contexts still needs further investigation. Development of reliable ferroptosis biomarkers will be an important step in characterizing ferroptosis in human disease. This can provide therapeutic opportunities aiming at targeting ferroptosis in human diseases. Antioxid. Redox Signal. 35, 487-509.
Collapse
Affiliation(s)
- Behrouz Hassannia
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Samya Van Coillie
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
21
|
Balla J, Zarjou A. Heme Burden and Ensuing Mechanisms That Protect the Kidney: Insights from Bench and Bedside. Int J Mol Sci 2021; 22:8174. [PMID: 34360940 PMCID: PMC8347331 DOI: 10.3390/ijms22158174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
With iron at its core, the tetrapyrrole heme ring is a cardinal prosthetic group made up of many proteins that participate in a wide array of cellular functions and metabolism. Once released, due to its pro-oxidant properties, free heme in sufficient amounts can result in injurious effects to the kidney and other organs. Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. HO-1 induction is a beneficial response to tissue injury in diverse animal models of diseases, including those that affect the kidney. These protective attributes are mainly due to: (i) prompt degradation of heme leading to restraining potential hazardous effects of free heme, and (ii) generation of byproducts that along with induction of ferritin have proven beneficial in a number of pathological conditions. This review will focus on describing clinical aspects of some of the conditions with the unifying end-result of increased heme burden and will discuss the molecular mechanisms that ensue to protect the kidneys.
Collapse
Affiliation(s)
- József Balla
- ELKH-UD Vascular Biology and Myocardial Pathophysiology Research Group, Division of Nephrology, Department of Medicine, Faculty of Medicine, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary;
| | - Abolfazl Zarjou
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, 618 Zeigler Research Building, 703 South 19th Street, Birmingham, AL 35294, USA
| |
Collapse
|
22
|
Ludes PO, de Roquetaillade C, Chousterman BG, Pottecher J, Mebazaa A. Role of Damage-Associated Molecular Patterns in Septic Acute Kidney Injury, From Injury to Recovery. Front Immunol 2021; 12:606622. [PMID: 33732235 PMCID: PMC7957065 DOI: 10.3389/fimmu.2021.606622] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) are a group of immunostimulatory molecules, which take part in inflammatory response after tissue injury. Kidney-specific DAMPs include Tamm-Horsfall glycoprotein, crystals, and uromodulin, released by tubular damage for example. Non-kidney-specific DAMPs include intracellular particles such as nucleus [histones, high-mobility group box 1 protein (HMGB1)] and cytosol parts. DAMPs trigger innate immunity by activating the NRLP3 inflammasome, G-protein coupled class receptors or the Toll-like receptor. Tubular necrosis leads to acute kidney injury (AKI) in either septic, ischemic or toxic conditions. Tubular necrosis releases DAMPs such as histones and HMGB1 and increases vascular permeability, which perpetuates shock and hypoperfusion via Toll Like Receptors. In acute tubular necrosis, intracellular abundance of NADPH may explain a chain reaction where necrosis spreads from cell to cell. The nature AKI in intensive care units does not have preclinical models that meet a variation of blood perfusion or a variation of glomerular filtration within hours before catecholamine infusion. However, the dampening of several DAMPs in AKI could provide organ protection. Research should be focused on the numerous pathophysiological pathways to identify the relative contribution to renal dysfunction. The therapeutic perspectives could be strategies to suppress side effect of DAMPs and to promote renal function regeneration.
Collapse
Affiliation(s)
- Pierre-Olivier Ludes
- Department of Anesthesiology and Intensive Care, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France.,EA 3072, Mitochondrie Stress Oxydant et Protection Musculaire, Faculté de Médecine, FRU 6702, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Charles de Roquetaillade
- Department of Anesthesiology and Critical Care, Hôpital Lariboisière, DMU Parabol, APHP.Nord, Paris, France.,Inserm U942 MASCOT, Université de Paris, Paris, France
| | - Benjamin Glenn Chousterman
- Department of Anesthesiology and Critical Care, Hôpital Lariboisière, DMU Parabol, APHP.Nord, Paris, France.,Inserm U942 MASCOT, Université de Paris, Paris, France
| | - Julien Pottecher
- Department of Anesthesiology and Intensive Care, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France.,EA 3072, Mitochondrie Stress Oxydant et Protection Musculaire, Faculté de Médecine, FRU 6702, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Alexandre Mebazaa
- Department of Anesthesiology and Critical Care, Hôpital Lariboisière, DMU Parabol, APHP.Nord, Paris, France.,Inserm U942 MASCOT, Université de Paris, Paris, France
| |
Collapse
|
23
|
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
|
24
|
Gupta S, Coca SG, Chan L, Melamed ML, Brenner SK, Hayek SS, Sutherland A, Puri S, Srivastava A, Leonberg-Yoo A, Shehata AM, Flythe JE, Rashidi A, Schenck EJ, Goyal N, Hedayati SS, Dy R, Bansal A, Athavale A, Nguyen HB, Vijayan A, Charytan DM, Schulze CE, Joo MJ, Friedman AN, Zhang J, Sosa MA, Judd E, Velez JCQ, Mallappallil M, Redfern RE, Bansal AD, Neyra JA, Liu KD, Renaghan AD, Christov M, Molnar MZ, Sharma S, Kamal O, Boateng JO, Short SA, Admon AJ, Sise ME, Wang W, Parikh CR, Leaf DE. AKI Treated with Renal Replacement Therapy in Critically Ill Patients with COVID-19. J Am Soc Nephrol 2021; 32:161-176. [PMID: 33067383 PMCID: PMC7894677 DOI: 10.1681/asn.2020060897] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 08/27/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND AKI is a common sequela of coronavirus disease 2019 (COVID-19). However, few studies have focused on AKI treated with RRT (AKI-RRT). METHODS We conducted a multicenter cohort study of 3099 critically ill adults with COVID-19 admitted to intensive care units (ICUs) at 67 hospitals across the United States. We used multivariable logistic regression to identify patient-and hospital-level risk factors for AKI-RRT and to examine risk factors for 28-day mortality among such patients. RESULTS A total of 637 of 3099 patients (20.6%) developed AKI-RRT within 14 days of ICU admission, 350 of whom (54.9%) died within 28 days of ICU admission. Patient-level risk factors for AKI-RRT included CKD, men, non-White race, hypertension, diabetes mellitus, higher body mass index, higher d-dimer, and greater severity of hypoxemia on ICU admission. Predictors of 28-day mortality in patients with AKI-RRT were older age, severe oliguria, and admission to a hospital with fewer ICU beds or one with greater regional density of COVID-19. At the end of a median follow-up of 17 days (range, 1-123 days), 403 of the 637 patients (63.3%) with AKI-RRT had died, 216 (33.9%) were discharged, and 18 (2.8%) remained hospitalized. Of the 216 patients discharged, 73 (33.8%) remained RRT dependent at discharge, and 39 (18.1%) remained RRT dependent 60 days after ICU admission. CONCLUSIONS AKI-RRT is common among critically ill patients with COVID-19 and is associated with a hospital mortality rate of >60%. Among those who survive to discharge, one in three still depends on RRT at discharge, and one in six remains RRT dependent 60 days after ICU admission.
Collapse
Affiliation(s)
- Shruti Gupta
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Steven G. Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lili Chan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michal L. Melamed
- Department of Medicine, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York
| | - Samantha K. Brenner
- Department of Internal Medicine, Hackensack Meridian School of Medicine, Seton Hall, Nutley, New Jersey
- Department of Internal Medicine, Heart and Vascular Hospital, Hackensack Meridian Health Hackensack University Medical Center, Hackensack, New Jersey
| | - Salim S. Hayek
- Division of Cardiology, University of Michigan Medical Center, Ann Arbor, Michigan
| | - Anne Sutherland
- Division of Pulmonary and Critical Care Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Sonika Puri
- Division of Nephrology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Anand Srivastava
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amanda Leonberg-Yoo
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexandre M. Shehata
- Department of Medicine, Hackensack Meridian Health Mountainside Medical Center, Glen Ridge, New Jersey
| | - Jennifer E. Flythe
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina Kidney Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
- Cecil G. Sheps Center for Health Services Research, University of North Carolina, Chapel Hill, North Carolina
| | - Arash Rashidi
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Edward J. Schenck
- Divison of Pulmonary and Critical Care Medicine, Department of Medicine Weill Cornell Medicine, New York, New York
| | - Nitender Goyal
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - S. Susan Hedayati
- Division of Nephrology, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rajany Dy
- Division of Pulmonary and Critical Care Medicine, University Medical Center, University of Nevada, Las Vegas, Nevada
| | - Anip Bansal
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - H. Bryant Nguyen
- Division of Pulmonary, Critical Care, Hyperbaric, Allergy, and Sleep Medicine, Loma Linda University Health, Loma Linda, California
| | - Anitha Vijayan
- Division of Nephrology, Washington University, St. Louis, Missouri
| | - David M. Charytan
- Division of Nephrology, New York University Grossman School of Medicine, New York, New York
| | - Carl E. Schulze
- Division of Nephrology, Department of Medicine, University of California, Los Angeles, California
| | - Min J. Joo
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois, Chicago, Illinois
| | - Allon N. Friedman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jingjing Zhang
- Division of Nephrology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Marie Anne Sosa
- Division of Nephrology, Department of Medicine, University of Miami Miller School of Medicine and Jackson Memorial Hospital, Miami, Florida
| | - Eric Judd
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Juan Carlos Q. Velez
- Department of Nephrology, Ochsner Health System, New Orleans, Louisiana
- Ochsner Clinical School, The University of Queensland, Brisbane, Queensland, Australia
| | - Mary Mallappallil
- Division of Nephrology, Kings County Hospital Center, New York City Health and Hospital Corporation, Brooklyn, New York
| | - Roberta E. Redfern
- Research Department, ProMedica Research, ProMedica Toledo Hospital, Toledo, Ohio
| | - Amar D. Bansal
- Renal and Electrolyte Division, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Javier A. Neyra
- Division of Nephrology, Department of Internal Medicine, Bone and Mineral Metabolism, University of Kentucky, Lexington, Kentucky
| | - Kathleen D. Liu
- Division of Nephrology and Critical Care Medicine, University of California, San Francisco, California
| | - Amanda D. Renaghan
- Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| | - Marta Christov
- Department of Medicine-Nephrology, Westchester Medical Center, New York Medical College, New York, New York
| | - Miklos Z. Molnar
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shreyak Sharma
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Omer Kamal
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Jeffery Owusu Boateng
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| | - Samuel A.P. Short
- University of Vermont Larner College of Medicine, Burlington, Vermont
| | - Andrew J. Admon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Meghan E. Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Wei Wang
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Chirag R. Parikh
- Division of Nephrology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - David E. Leaf
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | |
Collapse
|
25
|
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
|
26
|
Annamalai C, Ganesh RN, Viswanathan P. Ferrotoxicity and its amelioration by endogenous vitamin D in experimental acute kidney injury. Exp Biol Med (Maywood) 2020; 245:1474-1489. [PMID: 32741217 PMCID: PMC7553091 DOI: 10.1177/1535370220946271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
This work provides in-depth insights on catalytic iron-induced cytotoxicity and the resultant triggering of endogenous vitamin D synthesis in experimental acute kidney injury. Our results reveal significantly elevated levels of catalytic iron culminating in oxidant-mediated renal injury and a concomitant increase in 1,25-dihdyroxyvitamin D3 levels. Also, changes in other iron-related proteins including transferrin, ferritin, and hepcidin were observed both in the serum as well as in their mRNA expression. We consider all these findings vital since no connection between catalytic iron and vitamin D has been established so far. Furthermore, we believe that this work provides new and interesting results, with catalytic iron emerging as an important target in ameliorating renal cellular injury, possibly by timely administration of vitamin D. It also needs to be seen if these observations made in rats could be translated to humans by means of robust clinical trials.
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, India
| | - Rajesh N Ganesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry 605 006, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, India
- Pragasam Viswanathan.
| |
Collapse
|
27
|
Borawski B, Malyszko J. Iron, ferroptosis, and new insights for prevention in acute kidney injury. Adv Med Sci 2020; 65:361-370. [PMID: 32592957 DOI: 10.1016/j.advms.2020.06.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/06/2020] [Accepted: 06/14/2020] [Indexed: 01/01/2023]
Abstract
Acute kidney injury (AKI) is a very common condition with high morbidity and mortality, which can be seen in 5-7% of all hospitalized patients and in up to 57% of all intensive care unit admissions. Despite recent advances in clinical care, the prevalence of AKI has been shown to increase with virtually no change in mortality. AKI is a complex syndrome occurring in a variety of clinical settings. Early detection is crucial to prevent irreversible loss of renal function. The pathogenesis of AKI is highly multifactorial and complex, including vasoconstriction, reactive oxygen species formation, cell death, abnormal immune modulators and growth factors. Emerging evidence from both human and animal studies suggests that dysregulation of iron metabolism may play a potentially important role in AKI. Therefore, targeting the iron homeostasis may provide a new therapeutic intervention for AKI. New therapeutic strategies including iron chelation therapy, targeting iron metabolism related proteins and direct inhibitors of ferroptosis are imperative to improve the outcomes of patients. Taking into consideration the complexity of AKI, one intervention may not be enough for therapeutic success. Future preclinical studies in animal disease models followed by well-designed clinical trials should be conducted to extend findings from animal AKI models to humans.
Collapse
|
28
|
Annamalai C, Ganesh RN, Viswanathan P. Ferrotoxicity and its amelioration by endogenous vitamin D in experimental acute kidney injury. Exp Biol Med (Maywood) 2020. [DOI: https://doi.org/10.1177/1535370220946271] [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
Acute kidney injury causes significant morbidity and mortality. This experimental animal study investigated the simultaneous impact of iron and vitamin D on acute kidney injury induced by iohexol, an iodinated, non-ionic monomeric radiocontrast agent in Wistar rats. Out of 36 healthy male Wistar rats, saline was injected into six control rats (group 1) and iohexol into the remaining 30 experimental rats (groups 2 to 6 comprising six rats each). Biochemical, renal histological changes, and gene expression of iron-regulating proteins and 1 α-hydroxylase were analyzed. Urinary neutrophil gelatinase-associated lipocalin (NGAL), serum creatinine, urine protein, serum and urine catalytic iron, 25-hydroxyvitamin D3, 1,25-dihydroxyvitamin D3, and tissue lipid peroxidation were assayed. Rats injected with iohexol showed elevated urinary NGAL (11.94 ± 6.79 ng/mL), serum creatinine (2.92 ± 0.91 mg/dL), and urinary protein levels (11.03 ± 9.68 mg/mg creatinine) together with histological evidence of tubular injury and iron accumulation. Gene expression of iron-regulating proteins and 1 α-hydroxylase was altered. Serum and urine catalytic iron levels were elevated (0.57 ± 0.17; 48.95 ± 29.13 µmol/L) compared to controls (0.49 ± 0.04; 20.7 ± 2.62 µmol/L, P < 0.001). Urine catalytic iron positively correlated with tissue peroxidation (r = 0.469, CI 0.122 to 0.667, P = 0.004) and urinary NGAL (r = 0.788, CI 0.620 to 0.887, P < 0.001). 25-hydroxyvitamin D3 (61.58 ± 9.60 ng/mL) and 1,25-dihydroxyvitamin D3 (50.44 ± 19.76 pg/mL) levels increased simultaneously. In a multivariate linear regression analysis, serum iron, urine catalytic iron, and tissue lipid peroxidation independently and positively predicted urinary NGAL, an acute kidney injury biomarker. This study highlights the nephrotoxic potential of catalytic iron besides demonstrating a concurrent induction of vitamin D endogenously for possible renoprotection in acute kidney injury.Impact statementThis work provides in-depth insights on catalytic iron-induced cytotoxicity and the resultant triggering of endogenous vitamin D synthesis in experimental acute kidney injury. Our results reveal significantly elevated levels of catalytic iron culminating in oxidant-mediated renal injury and a concomitant increase in 1,25-dihdyroxyvitamin D3 levels. Also, changes in other iron-related proteins including transferrin, ferritin, and hepcidin were observed both in the serum as well as in their mRNA expression. We consider all these findings vital since no connection between catalytic iron and vitamin D has been established so far. Furthermore, we believe that this work provides new and interesting results, with catalytic iron emerging as an important target in ameliorating renal cellular injury, possibly by timely administration of vitamin D. It also needs to be seen if these observations made in rats could be translated to humans by means of robust clinical trials.
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, India
| | - Rajesh N Ganesh
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry 605 006, India
| | - Pragasam Viswanathan
- Renal Research Lab, Centre for Biomedical Research, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, India
| |
Collapse
|
29
|
Shu J, Hu Y, Yu X, Chen J, Xu W, Pan J. Elevated serum iron level is a predictor of prognosis in ICU patients with acute kidney injury. BMC Nephrol 2020; 21:303. [PMID: 32711469 PMCID: PMC7382811 DOI: 10.1186/s12882-020-01965-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/17/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Accumulation of iron is associated with oxidative stress, inflammation, and regulated cell death processes that contribute to the development of acute kidney injury (AKI). We aimed to investigate the association between serum iron levels and prognosis in intensive care unit (ICU) patients with AKI. METHODS A total of 483 patients with AKI defined as per the Kidney Disease: Improving Global Guidelines were included in this retrospective study. The data was extracted from the single-centre Medical Information Mart for Intensive Care III database. AKI patients with serum iron parameters measured upon ICU admission were included and divided into two groups (low group and high group). The prognostic value of serum iron was analysed using univariate and multivariate Cox regression analysis. RESULTS The optimal cut-off value for serum iron was calculated to be 60 μg/dl. Univariable Cox regression analysis showed that serum iron levels were significantly correlated with prognosis of AKI patients. After adjusting for possible confounding variables, serum iron levels higher than 60 μg/dl were associated with increases in 28-day (hazard [HR] 1.832; P < 0.001) and 90-day (HR 1.741; P < 0.001) mortality, as per multivariable Cox regression analysis. CONCLUSIONS High serum iron levels were associated with increased short- and long-term mortality in ICU patients with AKI. Serum iron levels measured upon admission may be used for predicting prognosis in AKI patients.
Collapse
Affiliation(s)
- Jie Shu
- Department of Intensive Care Unit, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yufeng Hu
- Department of Intensive Care Unit, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Xueshu Yu
- Department of Intensive Care Unit, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Jiaxiu Chen
- Department of Intensive Care Unit, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Wenwei Xu
- Department of Intensive Care Unit, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Jingye Pan
- Department of Intensive Care Unit, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
30
|
Zhou P, Deng B, Wu M, Ding F, Wang L. Interleukin-27 Ameliorates Renal Ischemia-Reperfusion Injury through Signal Transducers and Activators of Transcription 3 Signaling Pathway. Kidney Blood Press Res 2019; 44:1453-1464. [PMID: 31722339 DOI: 10.1159/000503923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 10/04/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a clinical syndrome characterized by significant morbidity and a high death rate. Interleukin (IL)-27 is a newly described member of the IL-6/IL-12 heterodimeric cytokine family and displays anti-inflammatory and antiapoptotic properties. OBJECTIVES To determine the effect and mechanism of IL-27 in AKI. METHOD We used a mouse model of renal ischemia/reperfusion (I/R) injury to investigate whether IL-27 has a therapeutic potential for the treatment of AKI. For the IL-27 administration group, IL-27 protein was injected 1 h before ischemia. Human proximal tubular epithelial cells were exposed to ischemia for 2 h and followed by 2 h of reperfusion (I2h+R2h treatment) used as an in vitro model to investigate the effect of IL-27. RESULTS Two IL-27 subunits, Epstein-Barr virus gene 3 and p28, were upregulated in kidneys 24 h after I/R. Renal expression of IL-27 receptor subunits (gp130 and WSX-1) was also increased. Treatment with IL-27 reduced structural/functional damages, ameliorated renal inflammation, inhibited the cleaved caspase-3 expression, upregulated antiapoptotic protein Bcl-2 and downregulated proapoptotic protein Bax in the kidneys of mice subjected to I/R. Meanwhile, the level of IL-27 receptor on renal tubular epithelial cells was increased after I2h+R2h treatment, and IL-27 administration suppressed I2h+R2h-induced epithelial cell apoptosis. Furthermore, IL-27 treatment led to activation of signal transducer and activator of transcription 3 (STAT3) both in vivo and in vitro, and IL-27-mediated protection against I2h+R2h injury was abolished by STAT3 inhibition. CONCLUSIONS IL-27 protects against renal I/R injury by activating STAT3, suggesting that IL-27 may represent a novel strategy for the treatment of AKI.
Collapse
Affiliation(s)
- Peihui Zhou
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Deng
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Feng Ding
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,
| |
Collapse
|
31
|
Sharma S, Leaf DE. Iron Chelation as a Potential Therapeutic Strategy for AKI Prevention. J Am Soc Nephrol 2019; 30:2060-2071. [PMID: 31554656 DOI: 10.1681/asn.2019060595] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AKI remains a major public health concern. Despite years of investigation, no intervention has been demonstrated to reliably prevent AKI in humans. Thus, development of novel therapeutic targets is urgently needed. An important role of iron in the pathophysiology of AKI has been recognized for over three decades. When present in excess and in nonphysiologic labile forms, iron is toxic to the kidneys and multiple other organs, whereas iron chelation is protective across a broad spectrum of insults. In humans, small studies have investigated iron chelation as a novel therapeutic strategy for prevention of AKI and extrarenal acute organ injury, and have demonstrated encouraging initial results. In this review, we examine the existing data on iron chelation for AKI prevention in both animal models and human studies. We discuss practical considerations for future clinical trials of AKI prevention using iron chelators, including selection of the ideal clinical setting, patient population, iron chelating agent, and dosing regimen. Finally, we compare the key differences among the currently available iron chelators, including pharmacokinetics, routes of administration, and adverse effects.
Collapse
Affiliation(s)
- Shreyak Sharma
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
32
|
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
|
33
|
Jacob KA, Leaf DE. Prevention of Cardiac Surgery-Associated Acute Kidney Injury: A Review of Current Strategies. Anesthesiol Clin 2019; 37:729-749. [PMID: 31677688 PMCID: PMC7644277 DOI: 10.1016/j.anclin.2019.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Acute kidney injury is a common and often severe postoperative complication after cardiac surgery, and is associated with poor short-term and long-term outcomes. Numerous randomized controlled trials have been conducted to investigate various strategies for prevention of cardiac surgery-associated acute kidney injury. Unfortunately, most trials that have been conducted to date have been negative. However, encouraging results have been demonstrated with preoperative administration of corticosteroids, leukocyte filtration, and administration of inhaled nitric oxide intraoperatively, and implementation of a Kidney Disease: Improving Global Outcomes bundle of care approach postoperatively. These findings require validation in large, multicenter trials.
Collapse
Affiliation(s)
- Kirolos A Jacob
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Mail Stop E03.511, PO Box 85500, Utrecht 3508 GA, the Netherlands.
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, 75 Francis Street, Medial Research Building Room MR416B, Boston, MA 02115, USA
| |
Collapse
|
34
|
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: 109] [Impact Index Per Article: 18.2] [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
|
35
|
Abstract
Iron is required for key aspects of cellular physiology including mitochondrial function and DNA synthesis and repair. However, free iron is an aberration because of its ability to donate electrons, reduce oxygen, and generate reactive oxygen species. Iron-mediated cell injury or ferroptosis is a central player in the pathogenesis of acute kidney injury. There are several homeostatic proteins and pathways that maintain critical balance in iron homeostasis to allow iron's biologic functions yet avoid ferroptosis. Hepcidin serves as the master regulator of iron homeostasis through its ability to regulate ferroportin-mediated iron export and intracellular H-ferritin levels. Hepcidin is a protective molecule in acute kidney injury. Drugs targeting hepcidin, H-ferritin, and ferroptosis pathways hold great promise to prevent or treat kidney injury. In this review we discuss iron homeostasis under physiological and pathologic conditions and highlight its importance in acute kidney injury.
Collapse
|
36
|
Biomarkers of Acute Kidney Injury after Cardiac Surgery: A Narrative Review. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7298635. [PMID: 31346523 PMCID: PMC6620851 DOI: 10.1155/2019/7298635] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022]
Abstract
Cardiac surgery-associated acute kidney injury (CSA-AKI) is a major and serious complication in patients undergoing cardiac surgery and is independently associated with perioperative mortality and mortality. Therapeutic intervention aiming at reversing kidney dysfunction seems disappointing across multiple settings. Consequently, attention has shifted from treatment to prevention and early detection. The Kidney Disease: Improving Global Outcomes (KDIGO) guidelines have unified diagnostic standards mainly based on the serum creatinine (Scr) level or urine output, but neither marker is kidney specific. Efforts have been made to identify novel biomarkers with high sensitivity and specificity. The diagnostic capabilities of neutrophil gelatinase-associated lipocalin (NGAL) and G1 cell cycle arrest biomarker as biomarkers have been confirmed in a large number of clinical trials. The utility of biomarkers of cardiac function and inflammation has been validated in clinical studies. Aiming to offer valuable information for further research, we summarize the progress in defining current markers relevant to CSA-AKI in the last three years.
Collapse
|
37
|
Balla J, Balla G, Zarjou A. Ferritin in Kidney and Vascular Related Diseases: Novel Roles for an Old Player. Pharmaceuticals (Basel) 2019; 12:E96. [PMID: 31234273 PMCID: PMC6630272 DOI: 10.3390/ph12020096] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/14/2019] [Accepted: 06/19/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is at the forefront of a number of pivotal biological processes due to its ability to readily accept and donate electrons. However, this property may also catalyze the generation of free radicals with ensuing cellular and tissue toxicity. Accordingly, throughout evolution numerous pathways and proteins have evolved to minimize the potential hazardous effects of iron cations and yet allow for readily available iron cations in a wide variety of fundamental metabolic processes. One of the extensively studied proteins in the context of systemic and cellular iron metabolisms is ferritin. While clinicians utilize serum ferritin to monitor body iron stores and inflammation, it is important to note that the vast majority of ferritin is located intracellularly. Intracellular ferritin is made of two different subunits (heavy and light chain) and plays an imperative role as a safe iron depot. In the past couple of decades our understanding of ferritin biology has remarkably improved. Additionally, a significant body of evidence has emerged describing the significance of the kidney in iron trafficking and homeostasis. Here, we briefly discuss some of the most important findings that relate to the role of iron and ferritin heavy chain in the context of kidney-related diseases and, in particular, vascular calcification, which is a frequent complication of chronic kidney disease.
Collapse
Affiliation(s)
- József Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary.
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - György Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary.
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary.
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
38
|
Vilander LM, Vaara ST, Donner KM, Lakkisto P, Kaunisto MA, Pettilä V. Heme oxygenase-1 repeat polymorphism in septic acute kidney injury. PLoS One 2019; 14:e0217291. [PMID: 31120979 PMCID: PMC6532969 DOI: 10.1371/journal.pone.0217291] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/08/2019] [Indexed: 01/22/2023] Open
Abstract
Acute kidney injury (AKI) is a syndrome that frequently affects the critically ill. Recently, an increased number of dinucleotide repeats in the HMOX1 gene were reported to associate with development of AKI in cardiac surgery. We aimed to test the replicability of this finding in a Finnish cohort of critically ill septic patients. This multicenter study was part of the national FINNAKI study. We genotyped 300 patients with severe AKI (KDIGO 2 or 3) and 353 controls without AKI (KDIGO 0) for the guanine–thymine (GTn) repeat in the promoter region of the HMOX1 gene. The allele calling was based on the number of repeats, the cut off being 27 repeats in the S–L (short to long) classification, and 27 and 34 repeats for the S–M–L2 (short to medium to very long) classification. The plasma concentrations of heme oxygenase-1 (HO-1) enzyme were measured on admission. The allele distribution in our patients was similar to that published previously, with peaks at 23 and 30 repeats. The S-allele increases AKI risk. An adjusted OR was 1.30 for each S-allele in an additive genetic model (95% CI 1.01–1.66; p = 0.041). Alleles with a repeat number greater than 34 were significantly associated with lower HO-1 concentration (p<0.001). In septic patients, we report an association between a short repeat in HMOX1 and AKI risk.
Collapse
Affiliation(s)
- Laura M. Vilander
- Division of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- * E-mail:
| | - Suvi T. Vaara
- Division of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kati M. Donner
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Päivi Lakkisto
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Mari A. Kaunisto
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Ville Pettilä
- Division of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | |
Collapse
|
39
|
Chawla LS, Beers-Mulroy B, Tidmarsh GF. Therapeutic Opportunities for Hepcidin in Acute Care Medicine. Crit Care Clin 2019; 35:357-374. [DOI: 10.1016/j.ccc.2018.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
40
|
Leaf DE, Rajapurkar M, Lele SS, Mukhopadhyay B, Boerger EAS, Mc Causland FR, Eisenga MF, Singh K, Babitt JL, Kellum JA, Palevsky PM, Christov M, Waikar SS. Iron, Hepcidin, and Death in Human AKI. J Am Soc Nephrol 2019; 30:493-504. [PMID: 30737269 PMCID: PMC6405140 DOI: 10.1681/asn.2018100979] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/30/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Iron is a key mediator of AKI in animal models, but data on circulating iron parameters in human AKI are limited. METHODS We examined results from the ARF Trial Network study to assess the association of plasma catalytic iron, total iron, transferrin, ferritin, free hemoglobin, and hepcidin with 60-day mortality. Participants included critically ill patients with AKI requiring RRT who were enrolled in the study. RESULTS Of the 807 study participants, 409 (51%) died by day 60. In both unadjusted and multivariable adjusted models, higher plasma concentrations of catalytic iron were associated with a significantly greater risk of death, as were lower concentrations of hepcidin. After adjusting for other factors, patients with catalytic iron levels in the highest quintile versus the lowest quintile had a 4.06-fold increased risk of death, and patients with hepcidin levels in the lowest quintile versus the highest quintile of hepcidin had a 3.87-fold increased risk of death. These findings were consistent across multiple subgroups. Other iron markers were also associated with death, but the magnitude of the association was greatest for catalytic iron and hepcidin. Higher plasma concentrations of catalytic iron and lower concentrations of hepcidin are each independently associated with mortality in critically ill patients with AKI requiring RRT. CONCLUSIONS These findings suggest that plasma concentrations of catalytic iron and hepcidin may be useful prognostic markers in patients with AKI. Studies are needed to determine whether strategies to reduce catalytic iron or increase hepcidin might be beneficial in this patient population.
Collapse
Affiliation(s)
- David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts;
| | | | | | | | - Emily A S Boerger
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Michele F Eisenga
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Departments of
| | - Karandeep Singh
- Learning Health Sciences and
- Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jodie L Babitt
- Nephrology Division, Program in Membrane Biology, Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts
| | - John A Kellum
- Center for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Paul M Palevsky
- Renal Section, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
| | - Marta Christov
- Department of Medicine, New York Medical College, Valhalla, New York
| | - Sushrut S Waikar
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
41
|
Sarhan M, Land WG, Tonnus W, Hugo CP, Linkermann A. Origin and Consequences of Necroinflammation. Physiol Rev 2018; 98:727-780. [PMID: 29465288 DOI: 10.1152/physrev.00041.2016] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
When cells undergo necrotic cell death in either physiological or pathophysiological settings in vivo, they release highly immunogenic intracellular molecules and organelles into the interstitium and thereby represent the strongest known trigger of the immune system. With our increasing understanding of necrosis as a regulated and genetically determined process (RN, regulated necrosis), necrosis and necroinflammation can be pharmacologically prevented. This review discusses our current knowledge about signaling pathways of necrotic cell death as the origin of necroinflammation. Multiple pathways of RN such as necroptosis, ferroptosis, and pyroptosis have been evolutionary conserved most likely because of their differences in immunogenicity. As the consequence of necrosis, however, all necrotic cells release damage associated molecular patterns (DAMPs) that have been extensively investigated over the last two decades. Analysis of necroinflammation allows characterizing specific signatures for each particular pathway of cell death. While all RN-pathways share the release of DAMPs in general, most of them actively regulate the immune system by the additional expression and/or maturation of either pro- or anti-inflammatory cytokines/chemokines. In addition, DAMPs have been demonstrated to modulate the process of regeneration. For the purpose of better understanding of necroinflammation, we introduce a novel classification of DAMPs in this review to help detect the relative contribution of each RN-pathway to certain physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Walter G Land
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Wulf Tonnus
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Christian P Hugo
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Andreas Linkermann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
42
|
Paving the way for precision medicine v2.0 in intensive care by profiling necroinflammation in biofluids. Cell Death Differ 2018; 26:83-98. [PMID: 30201975 PMCID: PMC6294775 DOI: 10.1038/s41418-018-0196-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/16/2018] [Accepted: 08/10/2018] [Indexed: 12/21/2022] Open
Abstract
Current clinical diagnosis is typically based on a combination of approaches including clinical examination of the patient, clinical experience, physiologic and/or genetic parameters, high-tech diagnostic medical imaging, and an extended list of laboratory values mostly determined in biofluids such as blood and urine. One could consider this as precision medicine v1.0. However, recent advances in technology and better understanding of molecular mechanisms underlying disease will allow us to better characterize patients in the future. These improvements will enable us to distinguish patients who have similar clinical presentations but different cellular and molecular responses. Treatments will be able to be chosen more “precisely”, resulting in more appropriate therapy, precision medicine v2.0. In this review, we will reflect on the potential added value of recent advances in technology and a better molecular understanding of necrosis and inflammation for improving diagnosis and treatment of critically ill patients. We give a brief overview on the mutual interplay between necrosis and inflammation, which are two crucial detrimental factors in organ and/or systemic dysfunction. One of the challenges for the future will thus be the cellular and molecular profiling of necroinflammation in biofluids. The huge amount of data generated by profiling biomolecules and single cells through, for example, different omic-approaches is needed for data mining methods to allow patient-clustering and identify novel biomarkers. The real-time monitoring of biomarkers will allow continuous (re)evaluation of treatment strategies using machine learning models. Ultimately, we may be able to offer precision therapies specifically designed to target the molecular set-up of an individual patient, as has begun to be done in cancer therapeutics. Critical care mostly implies life-threatening situations involving systemic infection, inflammation and necrosis. Biofluids are an easily accessible source of liquid biopsies that can be used to monitor the evolution of the patient’s critical illness. The cellular and molecular profiling of necrosis and inflammation in biofluids using cutting-edge technologies such as realtime immunodiagnostics, next-generation sequencing and mass spectrometry will pave the way for precision medicine v2.0 in critical care. This is needed for data mining approaches to allow patientclustering, identify novel biomarkers and develop novel intervention strategies controlling necrosis and inflammation. The real-time monitoring of biomarkers will allow continued (re)evaluation of treatment strategies using machine learning models. ![]()
Collapse
|
43
|
Choi N, Whitlock R, Klassen J, Zappitelli M, Arora RC, Rigatto C, Ho J. Early intraoperative iron-binding proteins are associated with acute kidney injury after cardiac surgery. J Thorac Cardiovasc Surg 2018; 157:287-297.e2. [PMID: 30195593 DOI: 10.1016/j.jtcvs.2018.06.091] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/23/2018] [Accepted: 06/28/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Iron regulation is an important modifier of renal ischemia-reperfusion injury, but the role of iron-binding proteins during cardiopulmonary bypass remains unclear. The goal was to characterize iron-binding proteins throughout ischemia-reperfusion injury to determine their association with acute kidney injury development. METHODS A prospective observational cohort of adult patients who underwent cardiac surgery (n = 301) was obtained, and acute kidney injury was defined by Kidney Disease Improving Global Outcomes. Serum ferritin, transferrin saturation, and urine hepcidin-25 were measured. RESULTS Intraoperative serum ferritin was lower at the start of cardiopulmonary bypass (P = .005) and 1-hour cardiopulmonary bypass (P = .001) in patients with acute kidney injury versus patients without acute kidney injury. Lower serum ferritin and higher transferrin saturation at 1-hour cardiopulmonary bypass were independent predictors of acute kidney injury (serum ferritin odds ratio, 0.66; 95% confidence interval [CI], 0.48-0.91; transferrin saturation odds ratio, 1.26; 95% CI, 1.02-1.55) and improved model discrimination (area under the curve [AUC], 0.76; 95% CI, 0.67-0.85) compared with clinical prediction alone (AUC, 0.72; 95% CI, 0.62-0.81; ΔAUC and net reclassification index, P = .01). Lower ferritin, higher transferrin saturation at 1-hour cardiopulmonary bypass, and lower urine hepcidin-25 at postoperative day 1 were also independent predictors for acute kidney injury development, and this model demonstrated an AUC of 0.80 (0.72-0.87), which was superior to clinical prediction (ΔAUC P = .002, integrated discrimination improvement and net reclassification index P = .003). CONCLUSIONS Our findings suggest that lower levels of intraoperative iron-binding proteins may reflect an impaired capacity to rapidly handle catalytic iron released during cardiopulmonary bypass, leading to kidney injury. These data highlight the importance of iron homeostasis in human ischemia-reperfusion injury and suggest it is a potentially modifiable risk during cardiac surgery. Intraoperative detection of incipient acute kidney injury may be feasible and could be used as an enrichment strategy for clinical trials.
Collapse
Affiliation(s)
- Nora Choi
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Reid Whitlock
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jessica Klassen
- Section of Nephrology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael Zappitelli
- Division of Nephrology, Department of Pediatrics, Toronto Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Rakesh C Arora
- Department of Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Claudio Rigatto
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Nephrology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Chronic Disease Innovation Centre, Seven Oaks Hospital, Winnipeg, Manitoba, Canada
| | - Julie Ho
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Nephrology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
44
|
Swaminathan S. Iron Homeostasis Pathways as Therapeutic Targets in Acute Kidney Injury. Nephron Clin Pract 2018; 140:156-159. [PMID: 29982259 DOI: 10.1159/000490808] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/11/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Iron is critical for fundamental biologic functions such as cell division and mitochondrial electron transport. However, by the virtue of its ability to donate electrons, iron is probably the most effective oxidant in biologic systems. SUMMARY To avoid damage from iron-mediated oxidative injury or ferroptosis, multiple defense mechanisms exist including iron binding proteins and robust glutathione-dependent intracellular pathways. Hepcidin, through its ability to sequester iron within macrophages and induce H-ferritin, serves as an endogenous protective molecule against ferroptosis. Key Messages: Recent studies have demonstrated the protective role of hepcidin in both ischemic reperfusion injury and heme-mediated models of acute kidney injury (AKI). Ferroptosis-inhibiting drugs and hepcidin offer exciting novel prospects to treat AKI.
Collapse
|
45
|
Karkouti K, Yip P, Chan C, Chawla L, Rao V. Pre-operative anaemia, intra-operative hepcidin concentration and acute kidney injury after cardiac surgery: a retrospective observational study. Anaesthesia 2018. [PMID: 29529338 DOI: 10.1111/anae.14274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acute kidney after cardiac surgery is more common in anaemic patients, whereas haemolysis during cardiopulmonary bypass may lead to iron-induced renal injury. Hepcidin promotes iron sequestration by macrophages: hepcidin concentration is reduced by anaemia and increased by inflammation. We analysed the associations in 525 patients between pre-operative anaemia (haemoglobin < 130 g.l-1 in men and < 120 g.l-1 in women), intra-operative hepcidin concentration and acute kidney injury (dialysis or > 26.4 μmol.l-1 or > 50% creatinine increase during the first two days after cardiac surgery. Rates of pre-operative anaemia and postoperative kidney injury were 109/525 (21%) and 36/525 (7%), respectively. The median (IQR [range]) intra-operative hepcidin concentration was 20 (10-33 [0-125]) μg.l-1 and was lower in anaemic patients than those who were not: 15 (4-28 [0-125]) μg.l-1 vs. 21 (12-33 [0-125]) μg.l-1 , respectively, p = 0.002. Four variables were independently associated with postoperative kidney injury, for which the beta-coefficients (SE) were: minutes on cardiopulmonary bypass, 0.016 (0.004), p < 0.001; intra-operative hepcidin concentration, 0.032 (0.008), p < 0.001; pre-operative anaemia, 1.97 (0.56), p < 0.001; and Cleveland clinic risk score, 0.88 (0.35), p = 0.005. Contrary to generally increased rates of kidney injury in patients with higher hepcidin concentrations, rates of kidney injury in anaemic patients were lower in patients with higher hepcidin concentrations, beta-coefficient (SE) -0.037 (0.01), p = 0.007. In cardiac surgical patients the rate of postoperative acute kidney injury predicted by the Cleveland risk score might be adjusted for pre-operative anaemia and intra-operative cardiopulmonary bypass time and hepcidin concentration. Pre-operative correction of anaemia, reduction in intra-operative bypass time and modification of iron homeostasis and hepcidin concentration might reduce acute kidney injury.
Collapse
Affiliation(s)
- K Karkouti
- Department of Anesthesia and Pain Management, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| | - P Yip
- Department of Clinical Biochemistry, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| | - C Chan
- Division of Nephrology, Department of Medicine, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| | - L Chawla
- Department of Anesthesiology and Critical Care Medicine, The George Washington University, Washington, DC, USA.,La Jolla Pharmaceutical Company, San Diego, CA, USA
| | - V Rao
- Division of Cardiac Surgery, Department of Surgery, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| |
Collapse
|
46
|
Abstract
Necrosis is a hallmark of several widespread diseases or their direct complications. In the past decade, we learned that necrosis can be a regulated process that is potentially druggable. RIPK3- and MLKL-mediated necroptosis represents by far the best studied pathway of regulated necrosis. During necroptosis, the release of damage-associated molecular patterns (DAMPs) drives a phenomenon referred to as necroinflammation, a common consequence of necrosis. However, most studies of regulated necrosis investigated cell lines in vitro in a cell autonomous manner, which represents a non-physiological situation. Conclusions based on such work might not necessarily be transferrable to disease states in which synchronized, non-cell autonomous effects occur. Here, we summarize the current knowledge of the pathophysiological relevance of necroptosis in vivo, and in light of this understanding, we reassess the morphological classification of necrosis that is generally used by pathologists. Along these lines, we discuss the paucity of data implicating necroptosis in human disease. Finally, the in vivo relevance of non-necroptotic forms of necrosis, such as ferroptosis, is addressed.
Collapse
Affiliation(s)
- Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
47
|
Sarhan M, von Mässenhausen A, Hugo C, Oberbauer R, Linkermann A. Immunological consequences of kidney cell death. Cell Death Dis 2018; 9:114. [PMID: 29371597 PMCID: PMC5833784 DOI: 10.1038/s41419-017-0057-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Death of renal cells is central to the pathophysiology of acute tubular necrosis, autoimmunity, necrotizing glomerulonephritis, cystic kidney disease, urosepsis, delayed graft function and transplant rejection. By means of regulated necrosis, immunogenic damage-associated molecular patterns (DAMPs) and highly reactive organelles such as lysosomes, peroxisomes and mitochondria are released from the dying cells, thereby causing an overwhelming immunologic response. The rupture of the plasma membrane exhibits the "point of no return" for the immunogenicity of regulated cell death, explaining why apoptosis, a highly organized cell death subroutine with long-lasting plasma membrane integrity, elicits hardly any immune response. Ferroptosis, an iron-dependent necrotic type cell death, results in the release of DAMPs and large amounts of lipid peroxides. In contrast, anti-inflammatory cytokines are actively released from cells that die by necroptosis, limiting the DAMP-induced immune response to a surrounding microenvironment, whereas at the same time, inflammasome-associated caspases drive maturation of intracellularly expressed interleukin-1β (IL-1β). In a distinct setting, additionally interleukin-18 (IL-18) is expressed during pyroptosis, initiated by gasdermin-mediated plasma membrane rupture. As all of these pathways are druggable, we provide an overview of regulated necrosis in kidney diseases with a focus on immunogenicity and potential therapeutic interventions.
Collapse
Affiliation(s)
- Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Anne von Mässenhausen
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Christian Hugo
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Rainer Oberbauer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
48
|
Regulated Cell Death. DAMAGE-ASSOCIATED MOLECULAR PATTERNS IN HUMAN DISEASES 2018. [PMCID: PMC7123501 DOI: 10.1007/978-3-319-78655-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In this chapter, the various subroutines of regulated cell death are neatly described by highlighting apoptosis and subforms of regulated necrosis such as necroptosis, ferroptosis, pyroptosis, and NETosis. Typically, all forms of regulated necrosis are defined by finite rupture of the plasma cell membrane. Apoptosis is characterized by an enzymatic machinery that consists of caspases which cause the morphologic features of this type of cell death. Mechanistically, apoptosis can be instigated by two major cellular signalling pathways: an intrinsic pathway that is initiated inside cells by mitochondrial release of pro-apoptotic factors or an extrinsic pathway that is initiated at the cell surface by various death receptors. In necroptosis, the biochemical processes are distinct from those found in apoptosis; in particular, there is no caspase activation. As such, necroptosis is a kinase-mediated cell death that relies on “receptor-interacting protein kinase 3” which mediates phosphorylation of the pseudokinase “mixed lineage kinase domain-like protein.” While ferroptosis is an iron-dependent, oxidative form of regulated necrosis that is biochemically characterized by accumulation of ROS from iron metabolism, oxidase activity, and lipid peroxidation products, pyroptosis is defined as a form of cell death (predominantly of phagocytes) that develops during inflammasome activation and is executed by caspase-mediated cleavage of the pore-forming protein gasdermin D. Finally, NETosis refers to a regulated death of neutrophils that is characterized by the release of chromatin-derived weblike structures released into the extracellular space. The chapter ends up with a discussion on the characteristic feature of regulated necrosis: the passive release of large amounts of constitutive DAMPs as a consequence of final plasma membrane rupture as well as the active secretion of inducible DAMPs earlier during the dying process. Notably, per cell death subroutine, the active secretion of inducible DAMPs varies, thereby determining different immunogenicity of dying cells.
Collapse
|
49
|
What's new in trace elements? Intensive Care Med 2017; 44:643-645. [PMID: 29022061 DOI: 10.1007/s00134-017-4955-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 09/30/2017] [Indexed: 10/18/2022]
|
50
|
Feng C, Naik BI, Xin W, Ma JZ, Scalzo DC, Thammishetti S, Thiele RH, Zuo Z, Raphael J. Haptoglobin 2-2 Phenotype Is Associated With Increased Acute Kidney Injury After Elective Cardiac Surgery in Patients With Diabetes Mellitus. J Am Heart Assoc 2017; 6:e006565. [PMID: 28982674 PMCID: PMC5721862 DOI: 10.1161/jaha.117.006565] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/15/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Recent studies reported an association between the 2-2 phenotype of haptoglobin (Hp 2-2) and increased cardiorenal morbidity in nonsurgical diabetic patients. Our goal was to determine whether the Hp 2-2 phenotype was associated with acute kidney injury (AKI) after elective cardiac surgery in patients with diabetes mellitus. METHODS AND RESULTS We prospectively enrolled 99 diabetic patients requiring elective cardiac surgery with cardiopulmonary bypass. Haptoglobin phenotypes were determined by gel electrophoresis. Cell-free hemoglobin, haptoglobin, and total serum bilirubin were quantified as hemolysis markers. The primary outcome was postoperative AKI, as defined by the Acute Kidney Injury Network classification. The incidence of AKI was significantly higher in Hp 2-2 patients compared with patients without this phenotype (non-Hp-2-2; 55.6% versus 27%, P<0.01). The need for renal replacement therapy was also significantly higher in the Hp 2-2 group (5 patients versus 1 patient, P=0.02). Thirty-day mortality (3 versus 0 patients, P=0.04) and 1-year mortality (5 versus 0 patients, P<0.01) were also significantly higher in patients with the Hp 2-2 phenotype. In multivariable analysis, Hp 2-2 was an independent predictor of postoperative AKI (P=0.01; odds ratio: 4.17; 95% confidence interval, 1.35-12.48). CONCLUSIONS Hp 2-2 phenotype is an independent predictor of postoperative AKI and is associated with decreased short and long-term survival after cardiac surgery in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Chenzhuo Feng
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
- Department of Basic Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Bhiken I Naik
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
| | - Wenjun Xin
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Jennie Z Ma
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - David C Scalzo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
| | | | - Robert H Thiele
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
| | - Jacob Raphael
- Department of Anesthesiology, University of Virginia, Charlottesville, VA
| |
Collapse
|