51
|
Hongisto M, Lassus J, Tarvasmäki T, Sans-Roselló J, Tolppanen H, Kataja A, Jäntti T, Sabell T, Banaszewski M, Silva-Cardoso J, Parissis J, Jurkko R, Spinar J, Castrén M, Mebazaa A, Masip J, Harjola VP. Soluble urokinase-type plasminogen activator receptor improves early risk stratification in cardiogenic shock. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:731–738. [PMID: 35949144 PMCID: PMC9629697 DOI: 10.1093/ehjacc/zuac096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/14/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
Abstract
AIMS Soluble urokinase-type plasminogen activator receptor (suPAR) is a biomarker reflecting the level of immune activation. It has been shown to have prognostic value in acute coronary syndrome and heart failure as well as in critical illness. Considering the complex pathophysiology of cardiogenic shock (CS), we hypothesized suPAR might have prognostic properties in CS as well. The aim of this study was to assess the kinetics and prognostic utility of suPAR in CS. METHODS AND RESULTS SuPAR levels were determined in serial plasma samples (0-96 h) from 161 CS patients in the prospective, observational, multicentre CardShock study. Kinetics of suPAR, its association with 90-day mortality, and additional value in risk-stratification were investigated. The median suPAR-level at baseline was 4.4 [interquartile range (IQR) 3.2-6.6)] ng/mL. SuPAR levels above median were associated with underlying comorbidities, biomarkers reflecting renal and cardiac dysfunction, and higher 90-day mortality (49% vs. 31%; P = 0.02). Serial measurements showed that survivors had significantly lower suPAR levels at all time points compared with nonsurvivors. For risk stratification, suPAR at 12 h (suPAR12h) with a cut-off of 4.4 ng/mL was strongly associated with mortality independently of established risk factors in CS: OR 5.6 (95% CI 2.0-15.5); P = 0.001) for death by 90 days. Adding suPAR12h > 4.4 ng/mL to the CardShock risk score improved discrimination identifying high-risk patients originally categorized in the intermediate-risk category. CONCLUSION SuPAR associates with mortality and improves risk stratification independently of other previously known risk factors in CS patients.
Collapse
Affiliation(s)
- Mari Hongisto
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| | - Johan Lassus
- Cardiology, University of Helsinki and Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Tuukka Tarvasmäki
- Cardiology, University of Helsinki and Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Jordi Sans-Roselló
- Cardiology Department, Hospital de la Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, Biomedical Research Institute IIB-Sant Pau, CIBER-CV, Spain
| | - Heli Tolppanen
- Cardiology, University of Helsinki and Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Anu Kataja
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| | - Toni Jäntti
- Cardiology, University of Helsinki and Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Tuija Sabell
- Cardiology, University of Helsinki and Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | | | - Jose Silva-Cardoso
- CINTESIS—Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, São João University Medical Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | - John Parissis
- ER and Heart Failure Unit, Attikon University Hospital, National and Kapodestrian University of Athens, Athens, Greece
| | - Raija Jurkko
- Cardiology, University of Helsinki and Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Jindrich Spinar
- St. Ann university hospital and Medical faculty Masaryk University, Brno, Czech Republic
| | - Maaret Castrén
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| | | | - Josep Masip
- Research Direction, Consorci Sanitari Integral, University of Barcelona, Barcelona, Spain
| | - Veli-Pekka Harjola
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
52
|
Chen Y, Zhang C, Du Y, Yang X, Liu M, Yang W, Lei G, Wang G. Exosomal transfer of microRNA-590-3p between renal tubular epithelial cells after renal ischemia-reperfusion injury regulates autophagy by targeting TRAF6. Chin Med J (Engl) 2022; 135:2467-2477. [PMID: 36449688 PMCID: PMC9945297 DOI: 10.1097/cm9.0000000000002377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication in patients, especially elderly patients, who undergo cardiac surgery with cardiopulmonary bypass. Studies have indicated a protective role of autophagy in AKI. However, the mechanisms underlying the regulatory effect of autophagy in AKI among patients undergoing cardiac surgeries are poorly understood. In this study, we aimed to test the hypothesis that exosomal microRNAs (miRNAs) regulate autophagy in tubular epithelial cells after AKI. METHODS Plasma exosomal RNA was extracted from young and elderly AKI patients undergoing cardiac surgery, and the miRNAs expression during the perioperative period were analyzed using next-generation sequencing. The screened miRNAs and their target genes were subjected to gene oncology function and Kyoto Encyclopedia of Genes and Genome enrichment analyses. Renal tubular epithelial cell line (HK-2 cells) was cultured and hypoxia/reoxygenation (H/R) model was established, which is an in vitro renal ischemia/reperfusion (I/R) model. We used Western blot analysis, cell viability assay, transfection, luciferase assay to investigate the mechanisms underlying the observed increases in the levels of renal I/R injury-mediated exosomal miRNAs and their roles in regulating HK-2 cells autophagy. RESULTS miR-590-3p was highly enriched in the plasma exosomes of young AKI patients after cardiac surgery. Increased levels of miR-590-3p led to the increases in the expression of autophagy marker proteins, including Beclin-1 and microtubule associated protein 1 light chain 3 beta (LC3II), and prolonged the autophagic response in HK-2 cells after H/R treatment. These effects were achieved mainly via increases in the exosomal miR-590-3p levels, and the tumor necrosis factor receptor-associated factor 6 protein was shown to play a key role in I/R injury-mediated autophagy induction. CONCLUSION Exosomes released from HK-2 cells after renal I/R injury regulate autophagy by transferring miR-590-3p in a paracrine manner, which suggests that increasing the miR-590-3p levels in HK-2 cell-derived exosomes may increase autophagy and protect against kidney injury after renal I/R injury.
Collapse
Affiliation(s)
- Yimeng Chen
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Congya Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Yingjie Du
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiying Yang
- Weifang Medical University, School of Anesthesiology, Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, Weifang, Shandong 261053, China
| | - Min Liu
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Wenjing Yang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Guiyu Lei
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
53
|
Luo S, Vasbinder A, Du‐Fay‐de‐Lavallaz JM, Gomez JMD, Suboc T, Anderson E, Tekumulla A, Shadid H, Berlin H, Pan M, Azam TU, Khaleel I, Padalia K, Meloche C, O'Hayer P, Catalan T, Blakely P, Launius C, Amadi K, Pop‐Busui R, Loosen SH, Chalkias A, Tacke F, Giamarellos‐Bourboulis EJ, Altintas I, Eugen‐Olsen J, Williams KA, Volgman AS, Reiser J, Hayek SS. Soluble Urokinase Plasminogen Activator Receptor and Venous Thromboembolism in COVID-19. J Am Heart Assoc 2022; 11:e025198. [PMID: 35924778 PMCID: PMC9683642 DOI: 10.1161/jaha.122.025198] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Background Venous thromboembolism (VTE) contributes significantly to COVID-19 morbidity and mortality. The urokinase receptor system is involved in the regulation of coagulation. Levels of soluble urokinase plasminogen activator receptor (suPAR) reflect hyperinflammation and are strongly predictive of outcomes in COVID-19. Whether suPAR levels identify patients with COVID-19 at risk for VTE is unclear. Methods and Results We leveraged a multinational observational study of patients hospitalized for COVID-19 with suPAR and D-dimer levels measured on admission. In 1960 patients (mean age, 58 years; 57% men; 20% Black race), we assessed the association between suPAR and incident VTE (defined as pulmonary embolism or deep vein thrombosis) using logistic regression and Fine-Gray modeling, accounting for the competing risk of death. VTE occurred in 163 (8%) patients and was associated with higher suPAR and D-dimer levels. There was a positive association between suPAR and D-dimer (β=7.34; P=0.002). Adjusted for clinical covariables, including D-dimer, the odds of VTE were 168% higher comparing the third with first suPAR tertiles (adjusted odds ratio, 2.68 [95% CI, 1.51-4.75]; P<0.001). Findings were consistent when stratified by D-dimer levels and in survival analysis accounting for death as a competing risk. On the basis of predicted probabilities from random forest, a decision tree found the combined D-dimer <1 mg/L and suPAR <11 ng/mL cutoffs, identifying 41% of patients with only 3.6% VTE probability. Conclusions Higher suPAR was associated with incident VTE independently of D-dimer in patients hospitalized for COVID-19. Combining suPAR and D-dimer identified patients at low VTE risk. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT04818866.
Collapse
Affiliation(s)
- Shengyuan Luo
- Department of MedicineRush University Medical CenterChicagoIL
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | | | | | - Tisha Suboc
- Department of MedicineRush University Medical CenterChicagoIL
| | - Elizabeth Anderson
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Annika Tekumulla
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Husam Shadid
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Hanna Berlin
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Michael Pan
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Tariq U. Azam
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Ibrahim Khaleel
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Kishan Padalia
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Chelsea Meloche
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Patrick O'Hayer
- Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Tonimarie Catalan
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Christopher Launius
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Kingsley‐Michael Amadi
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Rodica Pop‐Busui
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| | - Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Medical FacultyUniversity Hospital DüsseldorfDüsseldorfGermany
| | - Athanasios Chalkias
- Department of Anesthesiology, Faculty of MedicineUniversity of ThessalyLarisaGreece
- Outcomes Research ConsortiumClevelandOH
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Charité Mitte and Campus Virchow‐KlinikumCharité University Medicine BerlinBerlinGermany
| | | | - Izzet Altintas
- Department of Clinical ResearchCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Jesper Eugen‐Olsen
- Department of Clinical ResearchCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Kim A. Williams
- Department of Internal MedicineUniversity of Louisville School of MedicineLouisvilleKY
| | | | - Jochen Reiser
- Department of MedicineRush University Medical CenterChicagoIL
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal MedicineUniversity of MichiganAnn ArborMI
| |
Collapse
|
54
|
Liao TH, Wu HC, Liao MT, Hu WC, Tsai KW, Lin CC, Lu KC. The Perspective of Vitamin D on suPAR-Related AKI in COVID-19. Int J Mol Sci 2022; 23:10725. [PMID: 36142634 PMCID: PMC9500944 DOI: 10.3390/ijms231810725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed the lives of millions of people around the world. Severe vitamin D deficiency can increase the risk of death in people with COVID-19. There is growing evidence that acute kidney injury (AKI) is common in COVID-19 patients and is associated with poorer clinical outcomes. The kidney effects of SARS-CoV-2 are directly mediated by angiotensin 2-converting enzyme (ACE2) receptors. AKI is also caused by indirect causes such as the hypercoagulable state and microvascular thrombosis. The increased release of soluble urokinase-type plasminogen activator receptor (suPAR) from immature myeloid cells reduces plasminogen activation by the competitive inhibition of urokinase-type plasminogen activator, which results in low plasmin levels and a fibrinolytic state in COVID-19. Frequent hypercoagulability in critically ill patients with COVID-19 may exacerbate the severity of thrombosis. Versican expression in proximal tubular cells leads to the proliferation of interstitial fibroblasts through the C3a and suPAR pathways. Vitamin D attenuates the local expression of podocyte uPAR and decreases elevated circulating suPAR levels caused by systemic inflammation. This decrease preserves the function and structure of the glomerular barrier, thereby maintaining renal function. The attenuated hyperinflammatory state reduces complement activation, resulting in lower serum C3a levels. Vitamin D can also protect against COVID-19 by modulating innate and adaptive immunity, increasing ACE2 expression, and inhibiting the renin-angiotensin-aldosterone system. We hypothesized that by reducing suPAR levels, appropriate vitamin D supplementation could prevent the progression and reduce the severity of AKI in COVID-19 patients, although the data available require further elucidation.
Collapse
Affiliation(s)
- Tzu-Hsien Liao
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Hsien-Chang Wu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology and Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
| | - Ching-Chieh Lin
- Department of Chest Medicine, Taoyuan Armed Forces General Hospital Hsinchu Branch, Hsinchu City 300, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
55
|
Lausecker F, Koehler S, Fresquet M, Naylor RW, Tian P, Wanner N, Braun F, Butt L, Huber TB, Lennon R. Integrating basic science with translational research: the 13th International Podocyte Conference 2021. Kidney Int 2022; 102:708-719. [PMID: 35964799 PMCID: PMC9386279 DOI: 10.1016/j.kint.2022.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/30/2022]
Abstract
The 13th International Podocyte Conference was held in Manchester, UK, and online from July 28 to 30, 2021. Originally planned for 2020, this biannual meeting was postponed by a year because of the coronavirus disease 2019 (COVID-19) pandemic and proceeded as an innovative hybrid meeting. In addition to in-person attendance, online registration was offered, and this attracted 490 conference registrations in total. As a Podocyte Conference first, a day for early-career researchers was introduced. This premeeting included talks from graduate students and postdoctoral researchers. It gave early career researchers the opportunity to ask a panel, comprising academic leaders and journal editors, about career pathways and the future for podocyte research. The main meeting over 3 days included a keynote talk and 4 focused sessions each day incorporating invited talks, followed by selected abstract presentations, and an open panel discussion. The conference concluded with a Patient Day, which brought together patients, clinicians, researchers, and industry representatives. The Patient Day was an interactive and diverse day. As well as updates on improving diagnosis and potential new therapies, the Patient Day included a PodoArt competition, exercise and cooking classes with practical nutrition advice, and inspirational stories from patients and family members. This review summarizes the exciting science presented during the 13th International Podocyte Conference and demonstrates the resilience of researchers during a global pandemic.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Sybille Koehler
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Richard W Naylor
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Pinyuan Tian
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Nicola Wanner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Braun
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linus Butt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
56
|
Curran CS, Kopp JB. RAGE pathway activation and function in chronic kidney disease and COVID-19. Front Med (Lausanne) 2022; 9:970423. [PMID: 36017003 PMCID: PMC9395689 DOI: 10.3389/fmed.2022.970423] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/21/2022] [Indexed: 12/23/2022] Open
Abstract
The multi-ligand receptor for advanced glycation end-products (RAGE) and its ligands are contributing factors in autoimmunity, cancers, and infectious disease. RAGE activation is increased in chronic kidney disease (CKD) and coronavirus disease 2019 (COVID-19). CKD may increase the risk of COVID-19 severity and may also develop in the form of long COVID. RAGE is expressed in essentially all kidney cell types. Increased production of RAGE isoforms and RAGE ligands during CKD and COVID-19 promotes RAGE activity. The downstream effects include cellular dysfunction, tissue injury, fibrosis, and inflammation, which in turn contribute to a decline in kidney function, hypertension, thrombotic disorders, and cognitive impairment. In this review, we discuss the forms and mechanisms of RAGE and RAGE ligands in the kidney and COVID-19. Because various small molecules antagonize RAGE activity in animal models, targeting RAGE, its co-receptors, or its ligands may offer novel therapeutic approaches to slowing or halting progressive kidney disease, for which current therapies are often inadequate.
Collapse
Affiliation(s)
- Colleen S. Curran
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jeffrey B. Kopp
- Kidney Disease Section, NIDDK (National Institute of Diabetes and Digestive and Kidney Diseases), National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
57
|
Cottam D, Azzopardi G, Forni LG. Biomarkers for early detection and predicting outcomes in acute kidney injury. Br J Hosp Med (Lond) 2022; 83:1-11. [DOI: 10.12968/hmed.2022.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The current diagnosis of acute kidney injury relies on the measurement of serum creatinine levels and urine output. However, both measures are subject to considerable limitations; for example, change in serum creatinine levels ideally requires a knowledge of baseline function that is often not available. Furthermore, creatinine levels are influenced by many factors including diet, drug therapy, muscle mass, gender and ethnicity, which may lead to underestimation of the extent of renal dysfunction. Similarly, urine output lacks both specificity and sensitivity as a marker of acute kidney injury given that oliguria may be an appropriate physiological response to a multitude of stressors and that output may be maintained until significant renal damage has already occurred. Given the well-documented consequences of acute kidney injury and the considerable burden associated with its development, much attention has focused on early identification of patients at high risk to try and improve outcomes. Many studies have focused on the identification of candidate molecules that may enable the early detection of individuals at risk of developing acute kidney injury, including constitutive proteins associated with kidney damage, as well as molecules upregulated in response to injury, non-renal products that may be filtered, reabsorbed or secreted by the kidney, and markers of renal stress. Such biomarkers may also aid stratification for adverse events, such as the need for kidney replacement therapy or progression to chronic kidney disease and end-stage kidney disease. This article discusses some of these novel biomarkers and assesses the role they may have in the understanding, management, diagnosis and prognostication of acute kidney injury.
Collapse
Affiliation(s)
- Daniel Cottam
- Intensive Care Unit, Royal Surrey Hospital Foundation Trust, Guildford, UK
| | - Giada Azzopardi
- South West Thames Renal and Transplantation Unit, Epsom and St Helier University Hospitals, Carshalton, UK
| | - Lui G Forni
- Intensive Care Unit, Royal Surrey Hospital Foundation Trust, Guildford, UK
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
58
|
Soluble urokinase Plasminogen Activator Receptor (suPAR) levels are predictive of COVID-19 severity: An Italian experience. Clin Immunol 2022; 242:109091. [PMID: 35944880 PMCID: PMC9356594 DOI: 10.1016/j.clim.2022.109091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 11/21/2022]
Abstract
Background The soluble urokinase Plasminogen Activator Receptor (suPAR) has been identified as a reliable marker of COVID-19 severity, helping in personalizing COVID-19 therapy. This study aims to evaluate the correlation between suPAR levels and COVID-19 severity, in relation to the traditional inflammatory markers. Methods Sera from 71 COVID-19 patients were tested for suPAR levels using Chorus suPAR assay (Diesse Diagnostica Senese SpA, Italy). suPAR levels were compared with other inflammatory markers: IL-1β, IL-6, TNF-α, circulating calprotectin, neutrophil and lymphocyte counts, and Neutrophil/Lymphocytes Ratio (NLR). Respiratory failure, expressed as P/F ratio, and mortality rate were used as indicators of disease severity. Results A positive correlation of suPAR levels with IL-6 (r = 0.479, p = 0.000), TNF-α (r = 0.348, p = 0.003), circulating calprotectin (r = 0.369, p = 0.002), neutrophil counts (r = 0.447, p = 0.001), NLR (r = 0.492, p = 0.001) has been shown. Stratifying COVID-19 population by suPAR concentration above and below 6 ng/mL, we observed higher levels of circulating calprotectin (10.1 μg/mL, SD 7.9 versus 6.4 μg/mL, SD 7.5, p < 0.001), higher levels of P/F ratio (207.5 IQR 188.3 vs 312.0 IQR 127.8, p = 0.013) and higher mortality rate. Median levels of suPAR were increased in all COVID-19 patients requiring additional respiratory support (Nasal Cannula, Venturi Mask, BPAP and CPAP) (6.5 IQR = 4.9) compared to the group at room air (4.6 IQR = 4.2). Conclusion suPAR levels correlate with disease severity and survival rate of COVID-19 patients, representing a promising prognostic biomarker for the risk assessment of the disease.
Collapse
|
59
|
Tong-Minh K, van der Does Y, van Rosmalen J, Ramakers C, Gommers D, van Gorp E, Rizopoulos D, Endeman H. Joint Modeling of Repeated Measurements of Different Biomarkers Predicts Mortality in COVID-19 Patients in the Intensive Care Unit. Biomark Insights 2022; 17:11772719221112370. [PMID: 35859926 PMCID: PMC9290097 DOI: 10.1177/11772719221112370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 06/21/2022] [Indexed: 01/28/2023] Open
Abstract
Introduction: Predicting disease severity is important for treatment decisions in patients with COVID-19 in the intensive care unit (ICU). Different biomarkers have been investigated in COVID-19 as predictor of mortality, including C-reactive protein (CRP), procalcitonin (PCT), interleukin-6 (IL-6), and soluble urokinase-type plasminogen activator receptor (suPAR). Using repeated measurements in a prediction model may result in a more accurate risk prediction than the use of single point measurements. The goal of this study is to investigate the predictive value of trends in repeated measurements of CRP, PCT, IL-6, and suPAR on mortality in patients admitted to the ICU with COVID-19. Methods: This was a retrospective single center cohort study. Patients were included if they tested positive for SARS-CoV-2 by PCR test and if IL-6, PCT, suPAR was measured during any of the ICU admission days. There were no exclusion criteria for this study. We used joint models to predict ICU-mortality. This analysis was done using the framework of joint models for longitudinal and survival data. The reported hazard ratios express the relative change in the risk of death resulting from a doubling or 20% increase of the biomarker’s value in a day compared to no change in the same period. Results: A total of 107 patients were included, of which 26 died during ICU admission. Adjusted for sex and age, a doubling in the next day in either levels of PCT, IL-6, and suPAR were significantly predictive of in-hospital mortality with HRs of 1.523 (1.012-6.540), 75.25 (1.116-6247), and 24.45 (1.696-1057) respectively. With a 20% increase in biomarker value in a subsequent day, the HR of PCT, IL-6, and suPAR were 1.117 (1.03-1.639), 3.116 (1.029-9.963), and 2.319 (1.149-6.243) respectively. Conclusion: Joint models for the analysis of repeated measurements of PCT, suPAR, and IL-6 are a useful method for predicting mortality in COVID-19 patients in the ICU. Patients with an increasing trend of biomarker levels in consecutive days are at increased risk for mortality.
Collapse
Affiliation(s)
- Kirby Tong-Minh
- Department of Emergency Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Yuri van der Does
- Department of Emergency Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joost van Rosmalen
- Department of Biostatistics, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Christian Ramakers
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Diederik Gommers
- Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Eric van Gorp
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus University Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Henrik Endeman
- Department of Intensive Care, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
60
|
Wu LP, Pang K, Li B, Le Y, Tang YZ. Predictive Value of Glycosylated Hemoglobin for Post-operative Acute Kidney Injury in Non-cardiac Surgery Patients. Front Med (Lausanne) 2022; 9:886210. [PMID: 35899215 PMCID: PMC9309303 DOI: 10.3389/fmed.2022.886210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/22/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Recent studies have indicated that patients (both with and without diabetes) with elevated hemoglobin A1c (HbA1c) have a higher rate of acute kidney injury (AKI) following cardiac surgery. However, whether HbA1c could help to predict post-operative AKI in patients after non-cardiac surgery is less clear. This study aims to explore the predictive value of pre-operative HbA1c for post-operative AKI in non-cardiac surgery. Methods We reviewed the medical records of patients (≥ 18 years old) who underwent non-cardiac surgery between 2011 and 2020. Patient-related variables, including demographic and laboratory and procedure-related information, were collected, and univariable and multivariable logistic regression analyses were performed to determine the association of HbA1c with AKI. The area under the receiver operating curve (AUC), net reclassification improvement index (NRI), and integrated discriminant improvement index (IDI) were used to evaluate the predictive ability of the model, and decision curve analysis was used to evaluate the clinical utility of the HbA1c-added predictive model. Results A total of 3.3% of patients (94 of 2,785) developed AKI within 1 week after surgery. Pre-operative HbA1c was an independent predictor of AKI after adjustment for some clinical variables (OR comparing top to bottom quintiles 5.02, 95% CI, 1.90 to 13.24, P < 0.001 for trend; OR per percentage point increment in HbA1c 1.20, 95% CI, 1.07 to 1.33). Compared to the model with only clinical variables, the incorporation of HbA1c increased the model fit, modestly improved the discrimination (change in area under the curve from 0.7387 to 0.7543) and reclassification (continuous net reclassification improvement 0.2767, 95% CI, 0.0715 to 0.4818, improved integrated discrimination 0.0048, 95% CI, -5e-04 to 0.0101) of AKI and non-AKI cases, NRI for non-AKI improvement 0.3222, 95% CI, 0.2864 to 0.3580 and achieved a higher net benefit in decision curve analysis. Conclusion Elevated pre-operative HbA1c was independently associated with post-operative AKI risk and provided predictive value in patients after non-cardiac surgery. HbA1c improved the predictive power of a logistic regression model based on traditional clinical risk factors for AKI. Further prospective studies are needed to demonstrate the results and clinical application.
Collapse
Affiliation(s)
- Lan-Ping Wu
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Pang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Bo Li
- Surgery Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Le
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yuan Le,
| | - Yong-Zhong Tang
- Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, China
- Yong-Zhong Tang,
| |
Collapse
|
61
|
Roehm B, McAdams M, Hedayati SS. Novel Biomarkers of Kidney Disease in Advanced Heart Failure: Beyond GFR and Proteinuria. Curr Heart Fail Rep 2022; 19:223-235. [PMID: 35624386 DOI: 10.1007/s11897-022-00557-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Kidney disease is a common finding in patients with heart failure and can significantly impact treatment decisions and outcomes. Abnormal kidney function is currently determined in clinical practice using filtration markers in the blood to estimate glomerular filtration rate, but the manifestations of kidney disease in the setting of heart failure are much more complex than this. In this manuscript, we review novel biomarkers that may provide a more well-rounded assessment of kidney disease in patients with heart failure. RECENT FINDINGS Galectin-3, ST2, FGF-23, suPAR, miRNA, GDF-15, and NAG may be prognostic of kidney disease progression. L-FABP and suPAR may help predict acute kidney injury (AKI). ST2 and NAG may be helpful in diuretic resistance. Several biomarkers may be useful in determining prognosis of long-term kidney disease progression, prediction of AKI, and development of diuretic resistance. Further research into the mechanisms of kidney disease in heart failure utilizing many of these biomarkers may lead to the identification of therapeutic targets.
Collapse
Affiliation(s)
- Bethany Roehm
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 6201 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| | - Meredith McAdams
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 6201 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - S Susan Hedayati
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, 6201 Harry Hines Boulevard, Dallas, TX, 75390, USA
| |
Collapse
|
62
|
Mukherjee K, Gu C, Collins A, Mettlen M, Samelko B, Altintas MM, Sudhini YR, Wang X, Bouley R, Brown D, Pedro BP, Bane SL, Gupta V, Brinkkoetter PT, Hagmann H, Reiser J, Sever S. Simultaneous stabilization of actin cytoskeleton in multiple nephron-specific cells protects the kidney from diverse injury. Nat Commun 2022; 13:2422. [PMID: 35504916 PMCID: PMC9065033 DOI: 10.1038/s41467-022-30101-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/04/2022] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney diseases and acute kidney injury are mechanistically distinct kidney diseases. While chronic kidney diseases are associated with podocyte injury, acute kidney injury affects renal tubular epithelial cells. Despite these differences, a cardinal feature of both acute and chronic kidney diseases is dysregulated actin cytoskeleton. We have shown that pharmacological activation of GTPase dynamin ameliorates podocyte injury in murine models of chronic kidney diseases by promoting actin polymerization. Here we establish dynamin's role in modulating stiffness and polarity of renal tubular epithelial cells by crosslinking actin filaments into branched networks. Activation of dynamin's crosslinking capability by a small molecule agonist stabilizes the actomyosin cortex of the apical membrane against injury, which in turn preserves renal function in various murine models of acute kidney injury. Notably, a dynamin agonist simultaneously attenuates podocyte and tubular injury in the genetic murine model of Alport syndrome. Our study provides evidence for the feasibility and highlights the benefits of novel holistic nephron-protective therapies.
Collapse
Affiliation(s)
- Kamalika Mukherjee
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Changkyu Gu
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Agnieszka Collins
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Beata Samelko
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | | | - Xuexiang Wang
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Richard Bouley
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Dennis Brown
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Bradley P Pedro
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Susan L Bane
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, NY, USA
| | - Vineet Gupta
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Paul T Brinkkoetter
- Department of Internal Medicine-Center for Molecular Medicine Cologne, University of Cologne and Faculty of Medicine-University Hospital Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Henning Hagmann
- Department of Internal Medicine-Center for Molecular Medicine Cologne, University of Cologne and Faculty of Medicine-University Hospital Cologne, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne (Sybacol), Cologne, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
| | - Sanja Sever
- Department of Medicine, Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
63
|
Özdirik B, Maibier M, Scherf M, Nicklaus JM, Frohme J, Puengel T, Meyer zum Büschenfelde D, Tacke F, Mueller T, Sigal M. Soluble Urokinase Plasminogen Activator Receptor Levels Are Associated with Severity of Fibrosis in Patients with Primary Sclerosing Cholangitis. J Clin Med 2022; 11:jcm11092479. [PMID: 35566603 PMCID: PMC9105770 DOI: 10.3390/jcm11092479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/23/2022] [Accepted: 04/26/2022] [Indexed: 12/07/2022] Open
Abstract
The soluble urokinase-type plasminogen activator receptor (suPAR) has evolved as a useful biomarker for different entities of chronic liver disease. However, its role in patients with primary sclerosing cholangitis (PSC) is obscure. We analyzed plasma levels of suPAR in 84 patients with PSC and compared them to 68 patients with inflammatory bowel disease (IBD) without PSC and to 40 healthy controls. Results are correlated with clinical records. suPAR concentrations were elevated in patients with PSC compared to patients with IBD only and to healthy controls (p < 0.001). Elevated suPAR levels were associated with the presence of liver cirrhosis (p < 0.001) and signs of portal hypertension (p < 0.001). suPAR revealed a high accuracy for the discrimination of the presence of liver cirrhosis comparable to previously validated noninvasive fibrosis markers (area under the curve (AUC) 0.802 (95%CI: 0.702−0.902)). Further, we demonstrated that suPAR levels may indicate the presence of acute cholangitis episodes (p < 0.001). Finally, despite the high proportion of PSC patients with IBD, presence of IBD and its disease activity did not influence circulating suPAR levels. suPAR represents a previously unrecognized biomarker for diagnosis and liver cirrhosis detection in patients with PSC. However, it does not appear to be confounded by intestinal inflammation in the context of IBD.
Collapse
Affiliation(s)
- Burcin Özdirik
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
- Berlin Institute of Health, 10178 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-656-004; Fax: +49-30-450-553-902
| | - Martin Maibier
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
| | - Maria Scherf
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
| | - Jule Marie Nicklaus
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
| | - Josephine Frohme
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
| | - Tobias Puengel
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Dirk Meyer zum Büschenfelde
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany;
- Labor Berlin—Charité Vivantes GmbH, 13353 Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
| | - Tobias Mueller
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
| | - Michael Sigal
- Department of Hepatology and Gastroenterology, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (M.M.); (M.S.); (J.M.N.); (J.F.); (T.P.); (F.T.); (T.M.); (M.S.)
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| |
Collapse
|
64
|
Rotbain Curovic V, Houlind MB, Hansen TW, Eugen-Olsen J, Laursen JC, Eickhoff MK, Persson F, Rossing P. Acute and Long-Term Treatment With Dapagliflozin and Association With Serum Soluble Urokinase Plasminogen Activator Receptor. Front Pharmacol 2022; 13:799915. [PMID: 35571091 PMCID: PMC9091812 DOI: 10.3389/fphar.2022.799915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/09/2022] [Indexed: 01/10/2023] Open
Abstract
Background: Elevated soluble urokinase plasminogen activator receptor (suPAR) is highly associated with increased risk of diabetic complications. Dapagliflozin is a drug inhibiting the sodium-glucose co-transporter 2 in the kidney to decrease blood glucose, while also decreasing risk of kidney disease, heart failure, and death. Therefore, we have investigated suPAR as a monitor for treatment effect with dapagliflozin in diabetes. Methods: suPAR was measured in two double-blinded randomized clinical cross-over trials. The first trial investigated the effect of a single dose dapagliflozin 50 mg or placebo 12 h after intake, in individuals with type 1 diabetes and albuminuria. The second trial investigated the effect of a daily dose dapagliflozin 10 mg or placebo for 12 weeks, in individuals with type 2 diabetes and albuminuria. suPAR was measured in serum samples taken, in the acute trial, after treatment with dapagliflozin and placebo, and in the long-term trial, before and after treatment with dapagliflozin and placebo. Effect of dapagliflozin on suPAR levels were assessed using paired t-test. Results: 15 participants completed the acute trial and 35 completed the long-term trial. Mean difference in suPAR between dapagliflozin and placebo in the acute trial after 12 h was 0.70 ng/ml (95% CI: 0.66; 1.33, p = 0.49). In the long-term trial the mean difference was 0.06 ng/ml (95% CI -0.15; 0.27, p = 0.57). Conclusion: Based on our findings we conclude that suPAR is not a feasible marker to monitor the effect of treatment with dapagliflozin. Thus, a further search of suitable markers must continue.
Collapse
Affiliation(s)
| | - Morten B. Houlind
- Department of Clinical Research, Hvidovre Hospital, Hvidovre, Denmark
| | | | | | | | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
65
|
Long-Term Clinical Impact of Contrast-Associated Acute Kidney Injury Following PCI: An ADAPT-DES Substudy. JACC Cardiovasc Interv 2022; 15:753-766. [PMID: 35305904 DOI: 10.1016/j.jcin.2021.11.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES This study sought to determine correlates and consequences of contrast-associated acute kidney injury (CA-AKI) on clinical outcomes in patients with or without pre-existing chronic kidney disease (CKD). BACKGROUND The incidence and impact of CA-AKI on clinical outcomes during contemporary percutaneous coronary intervention (PCI) are not fully defined. METHODS The ADAPT-DES (Assessment of Dual AntiPlatelet Therapy With Drug Eluting Stents) study was a prospective, multicenter registry of 8,582 patients treated with ≥1 drug-eluting stent(s). CA-AKI was defined as a post-PCI increase in serum creatinine of >0.5 mg/dL or a relative increase of ≥25% compared with pre-PCI. CKD was defined as estimated glomerular filtration rate <60 mL/min/1.73 m2. The primary endpoint was the 2-year rate of net adverse clinical events (NACE): All-cause mortality, myocardial infarction (MI), definite or probable stent thrombosis, or major bleeding. RESULTS Of 7287 (85%) patients with evaluable data, 476 (6.5%) developed CA-AKI. In a multivariable model, older age, female sex, Caucasian race, congestive heart failure, diabetes, hypertension, CKD, presentation with ST-segment elevation MI, Killip class II to IV, radial access, intra-aortic balloon pump use, hypotension, and number of stents were independent predictors of CA-AKI. The 2-year NACE rate was higher in patients with CA-AKI (adjusted HR: 1.88; 95% CI: 1.42-2.49), as was each component of NACE (all-cause mortality, HR: 1.77; 95% CI: 1.22-2.55; MI, HR: 1.67; 95% CI: 1.18-2.36; definite/probable stent thrombosis, HR: 1.71; 95% CI: 1.10-2.65; and major bleeding, HR: 1.38; 95% CI: 1.06-1.80). Compared with the CA-AKI-/CKD- group, the CA-AKI+/CKD- (HR: 1.83; 95% CI: 1.33-2.52), CA-AKI-/CKD+ (HR: 1.56; 95% CI: 1.15-2.13), CA-AKI+/CKD+ (HR: 3.29; 95% CI: 1.92-5.67), and maintenance dialysis (HR: 2.67; 95% CI: 1.65-4.31) groups were at higher risk of NACE. CONCLUSIONS CA-AKI was relatively common after contemporary PCI and was associated with increased 2-year rates of NACE. Patients with pre-existing CKD were at particularly high risk for NACE after CA-AKI.
Collapse
|
66
|
Yu S, Sui Y, Wang J, Li Y, Li H, Cao Y, Chen L, Jiang L, Yuan C, Huang M. Crystal structure and cellular functions of uPAR dimer. Nat Commun 2022; 13:1665. [PMID: 35351875 PMCID: PMC8964761 DOI: 10.1038/s41467-022-29344-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/25/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractReceptor dimerization of urokinase-type plasminogen activator receptor (uPAR) was previously identified at protein level and on cell surface. Recently, a dimeric form of mouse uPAR isoform 2 was proposed to induce kidney disease. Here, we report the crystal structure of human uPAR dimer at 2.96 Å. The structure reveals enormous conformational changes of the dimer compared to the monomeric structure: D1 of uPAR opens up into a large expanded ring that captures a β-hairpin loop of a neighboring uPAR to form an expanded β-sheet, leading to an elongated, highly intertwined dimeric uPAR. Based on the structure, we identify E49P as a mutation promoting dimer formation. The mutation increases receptor binding to the amino terminal fragment of its primary ligand uPA, induces the receptor to distribute to the basal membrane, promotes cell proliferation, and alters cell morphology via β1 integrin signaling. These results reveal the structural basis for uPAR dimerization, its effect on cellular functions, and provide a basis to further study this multifunctional receptor.
Collapse
|
67
|
Skalec T, Adamik B, Kobylinska K, Gozdzik W. Soluble Urokinase-Type Plasminogen Activator Receptor Levels as a Predictor of Kidney Replacement Therapy in Septic Patients with Acute Kidney Injury: An Observational Study. J Clin Med 2022; 11:1717. [PMID: 35330042 PMCID: PMC8954771 DOI: 10.3390/jcm11061717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
The soluble urokinase-type plasminogen activator receptor (suPAR) is involved in the pathogenesis of acute kidney injury (AKI). Our goal was to establish the optimal suPAR cut-off point for predicting the need for kidney replacement therapy (KRT) use in sepsis patients and to analyze survival rates based on the suPAR level, AKI diagnosis, and the requirement for KRT. In total, 51 septic patients were included (82% septic shock; 96% mechanically ventilated, 35% KRT). Patients were stratified according to the AKI diagnosis and the need for KRT into three groups: AKI(+)/KRT(+), AKI(+)/KRT(−), and AKI(−)/KRT(−). A control group (N = 20) without sepsis and kidney failure was included. Sepsis patients had higher levels of the suPAR than control (13.01 vs. 4.05 ng/mL, p < 0.001). On ICU admission, the suPAR level was significantly higher in the AKI(+)/KRT(+) group than in the AKI(+)/KRT(−) and AKI(−)/KRT(−) groups (18.5 vs. 10.6 and 9.5 ng/mL, respectively; p = 0.001). The optimal suPAR cut-off point for predicting the need for KRT was established at 10.422 ng/mL (area under the curve 0.801, sensitivity 0.889, specificity 0.636). Moreover, patients AKI(+)/KRT(+) had the lowest probability of survival compared to patients AKI(+)/KRT(−) and AKI(−)/KRT(−) (p = 0.0003). The results indicate that the suPAR measurements may constitute an important element in the diagnosis of a patient with sepsis.
Collapse
Affiliation(s)
- Tomasz Skalec
- Clinical Department of Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Borowska St. 213, 50-556 Wroclaw, Poland; (T.S.); (W.G.)
| | - Barbara Adamik
- Clinical Department of Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Borowska St. 213, 50-556 Wroclaw, Poland; (T.S.); (W.G.)
| | - Katarzyna Kobylinska
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland;
| | - Waldemar Gozdzik
- Clinical Department of Anaesthesiology and Intensive Therapy, Wroclaw Medical University, Borowska St. 213, 50-556 Wroclaw, Poland; (T.S.); (W.G.)
| |
Collapse
|
68
|
Vasbinder A, Anderson E, Shadid H, Berlin H, Pan M, Azam TU, Khaleel I, Padalia K, Meloche C, O'Hayer P, Michaud E, Catalan T, Feroze R, Blakely P, Launius C, Huang Y, Zhao L, Ang L, Mikhael M, Mizokami-Stout K, Pennathur S, Kretzler M, Loosen SH, Chalkias A, Tacke F, Giamarellos-Bourboulis EJ, Reiser J, Eugen-Olsen J, Feldman EL, Pop-Busui R, Hayek SS. Inflammation, Hyperglycemia, and Adverse Outcomes in Individuals With Diabetes Mellitus Hospitalized for COVID-19. Diabetes Care 2022; 45:692-700. [PMID: 35045184 PMCID: PMC8918261 DOI: 10.2337/dc21-2102] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/19/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Diabetes mellitus (DM) is a major risk factor for severe coronavirus disease 2019 (COVID-19) for reasons that are unclear. RESEARCH DESIGN AND METHODS We leveraged the International Study of Inflammation in COVID-19 (ISIC), a multicenter observational study of 2,044 patients hospitalized with COVID-19, to characterize the impact of DM on in-hospital outcomes and assess the contribution of inflammation and hyperglycemia to the risk attributed to DM. We measured biomarkers of inflammation collected at hospital admission and collected glucose levels and insulin data throughout hospitalization. The primary outcome was the composite of in-hospital death, need for mechanical ventilation, and need for renal replacement therapy. RESULTS Among participants (mean age 60 years, 58.2% males), those with DM (n = 686, 33.5%) had a significantly higher cumulative incidence of the primary outcome (37.8% vs. 28.6%) and higher levels of inflammatory biomarkers than those without DM. Among biomarkers, DM was only associated with higher soluble urokinase plasminogen activator receptor (suPAR) levels in multivariable analysis. Adjusting for suPAR levels abrogated the association between DM and the primary outcome (adjusted odds ratio 1.23 [95% CI 0.78, 1.37]). In mediation analysis, we estimated the proportion of the effect of DM on the primary outcome mediated by suPAR at 84.2%. Hyperglycemia and higher insulin doses were independent predictors of the primary outcome, with effect sizes unaffected by adjusting for suPAR levels. CONCLUSIONS Our findings suggest that the association between DM and outcomes in COVID-19 is largely mediated by hyperinflammation as assessed by suPAR levels, while the impact of hyperglycemia is independent of inflammation.
Collapse
Affiliation(s)
- Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Elizabeth Anderson
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Husam Shadid
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Hanna Berlin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Michael Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Tariq U Azam
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Ibrahim Khaleel
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Kishan Padalia
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Chelsea Meloche
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Patrick O'Hayer
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Erinleigh Michaud
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Tonimarie Catalan
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Rafey Feroze
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Christopher Launius
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Yiyuan Huang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Lynn Ang
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Monica Mikhael
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Kara Mizokami-Stout
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Sven H Loosen
- Medical Faculty, Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Athanasios Chalkias
- Faculty of Medicine, Department of Anesthesiology, University of Thessaly, Larisa, Greece.,Outcomes Research Consortium, Cleveland, OH
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Charité Mitte and Campus Virchow-Klinikum, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI
| | - Rodica Pop-Busui
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
69
|
Rowaiye OO, Kusztal M, Zabinska M, Bartoszek D, Myszka M, Kościelska-Kasprzak K, Banasik M, Mazanowska O, Klinger M, Krajewska M. Anti-ETAR and suPAR as markers of disease activity in renal ANCA-associated vasculitis. Adv Med Sci 2022; 67:23-28. [PMID: 34781174 DOI: 10.1016/j.advms.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 08/26/2021] [Accepted: 11/07/2021] [Indexed: 11/01/2022]
Abstract
PURPOSE In anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), there is a lack of reliable biomarkers of disease activity. The aim of the study was to evaluate soluble urokinase plasminogen activator receptor (suPAR) and anti-endothelin-1 type A receptor (anti-ETAR) antibodies levels in active phase and remission of AAV. PATIENTS AND METHODS We enrolled 60 patients (median age 63.0 years) with renal AAV into this study. Plasma suPAR, urine suPAR (expressed as urine suPAR/creatinine ratio) and serum anti-ETAR antibodies were assayed by ELISA. Disease activity was assessed using Birmingham Vasculitis Activity Score (BVAS) and patients were divided into 2 subgroups based on their BVAS scores, namely: active AAV subgroup (BVAS≥1) and remission subgroup (BVAS = 0). Median follow-up was 12 months. RESULTS Patients with active AAV had higher levels of all candidate biomarkers in comparison to those in remission (p < 0.05). C-statistics for plasma suPAR, urine suPAR/creatinine ratio and serum anti-ETAR were 0.807, 0.713 and 0.783, respectively. In multivariable analysis, no clear associations were found between serum anti-ETAR and BVAS, while both plasma suPAR and serum anti-ETAR were independently influenced by estimated glomerular filtration rate (eGFR). CONCLUSIONS Plasma suPAR better discriminated between active AAV and remission in comparison to urine suPAR/creatinine ratio and serum anti-ETAR antibodies.
Collapse
|
70
|
Walter JE, Amrein MLF, Schäfer I, Zimmermann T, Lopez-Ayala P, Boeddinghaus J, Twerenbold R, Puelacher C, Nestelberger T, Wussler D, Honegger U, Badertscher P, Eugen-Olsen J, Koechlin L, Fahrni G, Jeger R, Kaiser C, Zellweger M, Mueller C. Soluble urokinase plasminogen activator receptor and functionally relevant coronary artery disease: a prospective cohort study. Biomarkers 2022; 27:278-285. [PMID: 35112976 DOI: 10.1080/1354750x.2022.2038269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Soluble urokinase plasminogen activator receptor (suPAR) is an emerging biomarker associated with anatomical CAD burden and cardiovascular outcomes including myocardial infarction (MI) and death. We aimed at validating previous findings of the prognostic value of suPAR and evaluated its diagnostic potential for functional relevant CAD (fCAD). METHODS Consecutive patients with suspected fCAD were enrolled. Adjudication of fCAD was performed blinded to suPAR concentrations by myocardial perfusion single photon emission tomography (MPI-SPECT) and coronary angiography. Prognostic outcome measures included all-cause, cardiovascular death, and incident MI during 2-year follow-up. RESULTS Among consecutive 968 patients, SuPAR concentrations were higher in patients with fCAD compared to those without (3.45ng/mL versus 3.20ng/mL, p = 0.007), without acceptable diagnostic accuracy (area under the curve [AUC]: 0.56, 95%CI 0.52-0.60). SuPAR correlated with high-sensitivity cardiac-troponin (hs-cTn) T (Spearman's rho (ρ) 0.393, p < 0.001), NT-proBNP (ρ = 0.327, p < 0.001), age (ρ = 0.364, p < 0.001) and very weakly with coronary atherosclerosis (ρ = 0.123, p < 0.001). Prognostic discrimination of suPAR was moderate for cardiovascular death (AUC =0.72, 95%CI 0.62-0.81) and all-cause death (AUC =0.72, 95%CI 0.65-0.79) at 2-years. SuPAR remained a significant predictor for all-cause death in the full model (HR =1.96, p = 0.001). CONCLUSIONS SuPAR was an independent predictor of all-cause death, without diagnostic utility for fCAD.
Collapse
Affiliation(s)
- Joan Elias Walter
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Department of Radiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Melissa Lee Fen Amrein
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Ibrahim Schäfer
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Tobias Zimmermann
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Pedro Lopez-Ayala
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Jasper Boeddinghaus
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Raphael Twerenbold
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,University Heart & Vascular Center Hamburg, Germany
| | - Christian Puelacher
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Department of Radiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Thomas Nestelberger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Vancouver General Hospital, University of British Columbia, Canada
| | - Desiree Wussler
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Universitäts-Herzzentrum Bad Krozingen, Germany
| | - Ursina Honegger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Patrick Badertscher
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Copenhagen University Hospital Hvidovre, Denmark
| | - Luca Koechlin
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Department of Cardiac Surgery, University Hospital Basel, University of Basel, Switzerland
| | - Gregor Fahrni
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Raban Jeger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Christoph Kaiser
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Michael Zellweger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Christian Mueller
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| |
Collapse
|
71
|
Morath C, Hayek SS, Döhler B, Nusshag C, Sommerer C, Zeier M, Reiser J, Süsal C. Soluble Urokinase Receptor and Mortality in Kidney Transplant Recipients. Transpl Int 2022; 35:10071. [PMID: 35185364 PMCID: PMC8842271 DOI: 10.3389/ti.2021.10071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/30/2021] [Indexed: 01/20/2023]
Abstract
Main problem: Soluble urokinase plasminogen activator receptor (suPAR) is an immunological risk factor for kidney disease and a prognostic marker for cardiovascular events. Methods: We measured serum suPAR levels in a total of 1,023 kidney transplant recipients either before (cohort 1, n = 474) or at year 1 after transplantation (cohort 2, n = 549). The association of suPAR levels and all-cause and cardiovascular mortality was evaluated by multivariable Cox regression analysis. Results: The highest suPAR tertile compared to the two lower tertiles had a significantly higher risk of all-cause mortality in both cohorts separately (cohort 1: hazard ratio (HR) 1.92, 95% confidence interval (CI) 1.20–3.08, p = 0.007; cohort 2: HR = 2.78, 95% CI 1.51–5.13, p = 0.001) and combined (n = 1,023, combined HR = 2.14, 95% CI 1.48–3.08, p < 0.001). The association remained significant in the subgroup of patients with normal kidney function (cohort 2: HR = 5.40, 95% CI 1.42–20.5, p = 0.013). The increased mortality risk in patients with high suPAR levels was attributable mainly to an increased rate of cardiovascular death (n = 1,023, HR = 4.24, 95% CI 1.81–9.96, p < 0.001). Conclusion: A high suPAR level prior to and at 1 year after kidney transplantation was associated with an increased risk of patient death independent of kidney function, predominantly from cardiovascular cause.
Collapse
Affiliation(s)
- Christian Morath
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
- *Correspondence: Christian Morath,
| | - Salim S. Hayek
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Bernd Döhler
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Claudia Sommerer
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jochen Reiser
- Department of Medicine, Rush Medical College, Rush University, Chicago, IL, United States
| | - Caner Süsal
- Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
- Transplant Immunology Research Center of Excellence, Koç Üniversitesi, Istanbul, Turkey
| |
Collapse
|
72
|
Iversen E, Kallemose T, Hornum M, Bengaard AK, Nehlin JO, Rasmussen LJH, Sandholdt H, Tavenier J, Feldt-Rasmussen B, Andersen O, Eugen-Olsen J, Houlind MB. OUP accepted manuscript. Clin Kidney J 2022; 15:1534-1541. [PMID: 35892012 PMCID: PMC9308102 DOI: 10.1093/ckj/sfac048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Indexed: 11/12/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Mads Hornum
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Kathrine Bengaard
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Capital Region Pharmacy, Herlev, Denmark
| | - Jan Olof Nehlin
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Haakon Sandholdt
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Juliette Tavenier
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Bo Feldt-Rasmussen
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
73
|
Wang L, Zhao YT. Development and Validation of a Prediction Model for Acute Kidney Injury Among Patients With Acute Decompensated Heart Failure. Front Cardiovasc Med 2021; 8:719307. [PMID: 34869626 PMCID: PMC8634389 DOI: 10.3389/fcvm.2021.719307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Acute kidney injury is an adverse event that carries significant morbidity among patients with acute decompensated heart failure (ADHF). We planned to develop a parsimonious model that is simple enough to use in clinical practice to predict the risk of acute kidney injury (AKI) occurrence. Methods: Six hundred and fifty patients with ADHF were enrolled in this study. Data for each patient were collected from medical records. We took three different approaches of variable selection to derive four multivariable logistic regression model. We selected six candidate predictors that led to a relatively stable outcome in different models to derive the final prediction model. The prediction model was verified through the use of the C-Statistics and calibration curve. Results: Acute kidney injury occurred in 42.8% of the patients. Advanced age, diabetes, previous renal dysfunction, high baseline creatinine, high B-type natriuretic peptide, and hypoalbuminemia were the strongest predictors for AKI. The prediction model showed moderate discrimination C-Statistics: 0.766 (95% CI, 0.729-0.803) and good identical calibration. Conclusion: In this study, we developed a prediction model and nomogram to estimate the risk of AKI among patients with ADHF. It may help clinical physicians detect AKI and manage it promptly.
Collapse
Affiliation(s)
- Lei Wang
- Department of Cardiology, Aerospace Center Hospital, Beijing, China
| | - Yun-Tao Zhao
- Department of Cardiology, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
74
|
Rasmussen LJH, Petersen JEV, Eugen-Olsen J. Soluble Urokinase Plasminogen Activator Receptor (suPAR) as a Biomarker of Systemic Chronic Inflammation. Front Immunol 2021; 12:780641. [PMID: 34925360 PMCID: PMC8674945 DOI: 10.3389/fimmu.2021.780641] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/09/2021] [Indexed: 01/08/2023] Open
Abstract
Systemic chronic inflammation (SCI) is persistent, health-damaging, low-grade inflammation that plays a major role in immunosenescence and in development and progression of many diseases. But currently, there are no recognized standard biomarkers to assess SCI levels alone, and SCI is typically measured by combining biomarkers of acute inflammation and infection, e.g., CRP, IL-6, and TNFα. In this review, we highlight 10 properties and characteristics that are shared by the blood protein soluble urokinase plasminogen activator receptor (suPAR) and SCI, supporting the argument that suPAR is a biomarker of SCI: (1) Expression and release of suPAR is upregulated by immune activation; (2) uPAR and suPAR exert pro-inflammatory functions; (3) suPAR is associated with the amount of circulating immune cells; (4) Blood suPAR levels correlate with the levels of established inflammatory biomarkers; (5) suPAR is minimally affected by acute changes and short-term influences, in contrast to many currently used markers of systemic inflammation; (6) Like SCI, suPAR is non-specifically associated with multiple diseases; (7) suPAR and SCI both predict morbidity and mortality; (8) suPAR and SCI share the same risk factors; (9) suPAR is associated with risk factors and outcomes of inflammation above and beyond other inflammatory biomarkers; (10) The suPAR level can be reduced by anti-inflammatory interventions and treatment of disease. Assessing SCI has the potential to inform risk for morbidity and mortality. Blood suPAR is a newer biomarker which may, in fact, be a biomarker of SCI since it is stably associated with inflammation and immune activation; shares the same risk factors as many age-related diseases; is both elevated by and predicts age-related diseases. There is strong evidence that suPAR is a prognostic marker of adverse events, morbidity, and mortality. It is associated with immune activity and prognosis across diverse conditions, including kidney disease, cardiovascular disease, cancer, diabetes, and inflammatory disorders. Thus, we think it likely represents a common underlying disease-process shared by many diseases; that is, SCI. We review the supporting literature and propose a research agenda that can help test the hypothesis that suPAR indexes SCI, with the potential of becoming the new gold standard for measuring SCI.
Collapse
Affiliation(s)
- Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| | - Jens Emil Vang Petersen
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| |
Collapse
|
75
|
Lupușoru G, Ailincăi I, Sorohan BM, Andronesi A, Achim C, Micu G, Caragheorgheopol A, Manda D, Lupușoru M, Ismail G. Serum soluble urokinase plasminogen activator receptor as a potential biomarker of renal impairment severity in diabetic nephropathy. Diabetes Res Clin Pract 2021; 182:109116. [PMID: 34728182 DOI: 10.1016/j.diabres.2021.109116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 12/28/2022]
Abstract
AIMS To investigate serum soluble form of urokinase-type plasminogen activator receptor (suPAR) in patients with diabetic kidney disease (DKD) and biopsy-proven diabetic nephropathy (DN), its correlation with histological parameters and its capacity as a biomarker for renal impairment severity. METHODS We conducted a cross-sectional study on 75 patients with diabetes mellitus (DM) and DKD, among whom 28 had biopsy-proven DN. RESULTS Among the 75 patients, 9 (12%) had type 1 and 66 (88%) type 2 DM. The median value of the serum suPAR level was 2857.2 pg/mL (1916.4-3700) in the entire cohort and 2472.1 pg/mL (1782.6-3745.8) in the biopsy-proven DN subgroup, respectively. suPAR was significantly correlated with diabetes duration, diabetic retinopathy, anti-proteinuric treatment, albuminuria, kidney function, DN class, interstitial fibrosis and tubular atrophy (IFTA) score and with interstitial inflammation score. suPAR had a good accuracy for the association with chronic kidney disease (CKD) stages G3b-5, macroalbuminuria, DN class IV, IFTA score 3 and interstitial inflammation score 2. CONCLUSIONS Serum suPAR was increased in DN patients and was associated with DM duration, diabetic retinopathy, renoprotective treatment, kidney function, proteinuria, DN class, IFTA and interstitial inflammation scores. Also, suPAR had a good capacity as a biomarker for advanced renal impairment and severe histological lesions of DN.
Collapse
Affiliation(s)
- Gabriela Lupușoru
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Ioana Ailincăi
- Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Bogdan Marian Sorohan
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania.
| | - Andreea Andronesi
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Camelia Achim
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Georgia Micu
- Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Andra Caragheorgheopol
- Research Department, "C.I.Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Dana Manda
- Research Department, "C.I.Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Mircea Lupușoru
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Gener Ismail
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
76
|
Liu W, Zhang A, He H, Zhao X, Tao F, Sun Y. Inflammatory burden in adolescents with prolonged parent-child separation. Brain Behav Immun 2021; 98:257-262. [PMID: 34454061 DOI: 10.1016/j.bbi.2021.08.227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/15/2021] [Accepted: 08/21/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Prolonged parent-child separation is associated with a broad array of poor developmental outcomes. A potential pathway may be through changes in inflammatory processes. However, relatively little is known about the relationship between parent-child separation pattern (timing and duration) and inflammatory burden. The aim of this study was to investigate whether parent-child separation since birth is associated with inflammatory burden in adolescents. METHODS A total of 574 adolescents (mean age 12.07 years, SD: 0.62) were enrolled from rural areas of Chizhou, Anhui Province, China. Parent-child separation was reported mainly by primary caregivers, and other adverse childhood adversities (ACEs) were derived from adolescents semi-structured interview or questionnaire. Blood samples were collected from venepuncture for C-reactive protein (CRP) as well as soluble urokinase plasminogen activator receptor (suPAR). RESULTS Nearly 40% (232/574) participants experienced parent-child separation, among which more than 1 of 4 persistently separated from both parents since birth. Both CRP and suPAR levels were significantly higher among adolescents persistently separated from both parents, compared with those who did not separate from both parents (CRP: 1.75 vs. 1.36 mg/L, P < 0.001; suPAR: 2.85 ng/mL vs. 2.55 ng/mL, P < 0.001). After adjusted for demographic covariates, body mass index, ACEs as well as parental characteristics, persistent parent-child separation was associated with elevated suPAR (B = 0.30; 95% CI, 0.12-0.48) and CRP (B = 1.34; 95% CI, 1.02-1.75). No similar associations were observed between inflammatory burden with current or early childhood parent-child separation groups. Adolescents who exposed to persistent parent-child separation were more likely to have elevated suPAR levels even if they did not have elevated CRP levels (aOR, 4.26, 95% CI, 1.23-14.80; P = 0.022). CONCLUSION Elevated inflammatory burden observed in persistent parent-child separation adolescents highlights the need to support children and adolescents undergoing separation from both parents in order to minimize the long-term impact on disease vulnerability.
Collapse
Affiliation(s)
- Wanxu Liu
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China
| | - Anhui Zhang
- Department of Child Health Care, Wuhu Maternal and Child Health (MCH) Center, Wuhu, Anhui Province, China
| | - Haiyan He
- Department of Child Health Care, Wuhu Maternal and Child Health (MCH) Center, Wuhu, Anhui Province, China
| | - Xudong Zhao
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China
| | - Fangbiao Tao
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University) Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health&Aristogenics, Hefei, Anhui Province, China
| | - Ying Sun
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University) Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health&Aristogenics, Hefei, Anhui Province, China.
| |
Collapse
|
77
|
Ix JH, Shlipak MG. The Promise of Tubule Biomarkers in Kidney Disease: A Review. Am J Kidney Dis 2021; 78:719-727. [PMID: 34051308 PMCID: PMC8545710 DOI: 10.1053/j.ajkd.2021.03.026] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/10/2021] [Indexed: 12/23/2022]
Abstract
For over 70 years, serum creatinine has remained the primary index for detection and monitoring of kidney disease. Tubulointerstitial damage and fibrosis are highly prognostic for subsequent kidney failure in biopsy studies, yet this pathology is invisible to the clinician in the absence of a biopsy. Recent discovery of biomarkers that reflect distinct aspects of kidney tubule disease have led to investigations of whether these markers can provide additional information on risk of chronic kidney disease (CKD) progression and associated adverse clinical end points, above and beyond estimated glomerular filtration rate and albuminuria. These biomarkers can be loosely grouped into those that mark tubule cell injury (eg, kidney injury molecule 1, monocyte chemoattractant protein 1) and those that mark tubule cell dysfunction (eg, α1-microglobulin, uromodulin). These kidney tubule biomarkers provide new opportunities to monitor response to therapeutics used to treat CKD patients. In this review, we describe results from some unique contributions in this area and discuss the current challenges and requirements in the field to bring these markers to clinical practice. We advocate for a broader assessment of kidney health that moves beyond a focus on the glomerulus, and we highlight how such tools can improve diagnostic accuracy and earlier assessment of therapeutic efficacy or harm in CKD patients.
Collapse
Affiliation(s)
- Joachim H Ix
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, San Diego, California; Nephrology Section, Veterans Affairs San Diego Healthcare System, La Jolla, California; Kidney Research Innovation Hub of San Diego, San Diego, California.
| | - Michael G Shlipak
- Kidney Health Research Collaborative, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, California; Division of General Internal Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, California
| |
Collapse
|
78
|
Qin Y, Qiao Y, Wang D, Yan G, Tang C, Ma G. The Predictive Value of Soluble Urokinase-Type Plasminogen Activator Receptor in Contrast-Induced Acute Kidney Injury in Patients Undergoing Percutaneous Coronary Intervention. Int J Gen Med 2021; 14:6497-6504. [PMID: 34675617 PMCID: PMC8504866 DOI: 10.2147/ijgm.s339075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/01/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Soluble urokinase-type plasminogen activator receptor (SuPAR) is a circulating protein and a novel identified promising biomarker for various renal diseases and kidney injury. However, it remains unknown on the predictive value of suPAR in contrast induced acute kidney injury (CI-AKI) in patients undergoing percutaneous coronary intervention (PCI). Methods A total of 399 patients undergoing PCI were enrolled in the research from June 2020 to June 2021 in Zhongda Hospital. Patients were divided into CI-AKI and non-CI-AKI groups according to the preoperative and postoperative serum creatinine levels. Plasma suPAR level was detected through enzyme-linked immunosorbent assay on admission. Demographic data, hematological parameters, coronary angiography data and medications were recorded and compared between CI-AKI and non-CI-AKI groups. Logistic regression analysis and receiver operator characteristic (ROC) curve analysis were performed for identifying the independent risk factors of CI-AKI and assessment of the predictive value of suPAR for CI-AKI. Results CI-AKI occurred in 65 (16.3%) patients undergoing PCI. The level of suPAR in CI-AKI group was significantly higher than that in the non-CI-AKI group. Multivariate logistic regression indicated diabetes, lower LVEF, lower hydration rate, lower baseline eGFR, higher plasma suPAR (OR = 2.875, 95% CI = 2.038–3.672, P < 0.001) and volume of contrast media were all independent risk factors for CI-AKI after adjustment of the confounding factors. ROC analysis illustrated that the optimal area under the curve was 0.765, indicating plasma suPAR was a splendid predictor for CI-AKI. The corresponding cut-off value was 3.305 ng/mL, and the sensitivity and specificity were 63.1% and 82.3%, respectively. Conclusion Increased suPAR level is independently associated with elevated risk of suffering CI-AKI, and suPAR is a strong predictor for CI-AKI in patients undergoing PCI. SuPAR might act as a novel biomarker for CI-AKI in clinical practice.
Collapse
Affiliation(s)
- Yuhan Qin
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
79
|
Bourassa KJ, Rasmussen LJH, Danese A, Eugen-Olsen J, Harrington H, Houts R, Poulton R, Ramrakha S, Sugden K, Williams B, Moffitt TE, Caspi A. Linking stressful life events and chronic inflammation using suPAR (soluble urokinase plasminogen activator receptor). Brain Behav Immun 2021; 97:79-88. [PMID: 34224821 PMCID: PMC8453112 DOI: 10.1016/j.bbi.2021.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/27/2022] Open
Abstract
Stressful life events have been linked to declining health, and inflammation has been proposed as a physiological mechanism that might explain this association. Using 828 participants from the Dunedin Longitudinal Study, we tested whether people who experienced more stressful life events during adulthood would show elevated systemic inflammation when followed up in midlife, at age 45. We studied three inflammatory biomarkers: C-reactive protein (CRP), interleukin-6 (IL-6), and a newer biomarker, soluble urokinase plasminogen activator receptor (suPAR), which is thought to index systemic chronic inflammation. Stressful life events were not associated with CRP or IL-6. However, people who experienced more stressful life events from age 38 to 44 had elevated suPAR at age 45, and had significantly greater increases in suPAR from baseline to follow-up across the same period. When examining stressful life events across the lifespan, both adverse childhood experiences (ACEs) and adult stressful life events were independently associated with suPAR at age 45. ACEs moderated the association of adult stressful life events and suPAR at age 45-children with more ACEs showed higher suPAR levels after experiencing stressful life events as adults. The results suggest systemic chronic inflammation is one physiological mechanism that could link stressful life events and health, and support the use of suPAR as a useful biomarker for such research.
Collapse
Affiliation(s)
- Kyle J. Bourassa
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC,Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Line J. H. Rasmussen
- Department of Psychology & Neuroscience, Duke University, Durham, NC,Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Andrea Danese
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, King’s College London, London, United Kingdom,Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom,National and Specialist Child and Adolescent Mental Health Services Trauma, Anxiety, and Depression Clinic, South London and Maudsley National Health Service Foundation Trust, London, United Kingdom
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | | | - Renate Houts
- Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Richie Poulton
- Department of Psychology, University of Otago, Otago, New Zealand
| | - Sandhya Ramrakha
- Department of Psychology, University of Otago, Otago, New Zealand
| | - Karen Sugden
- Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Ben Williams
- Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Terrie E. Moffitt
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC,Department of Psychology & Neuroscience, Duke University, Durham, NC,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC,Center for Genomic and Computational Biology, Duke University, Durham, NC
| | - Avshalom Caspi
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC,Department of Psychology & Neuroscience, Duke University, Durham, NC,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC,Center for Genomic and Computational Biology, Duke University, Durham, NC
| |
Collapse
|
80
|
Liu M, Chen S, Zhang A, Zheng Q, Fu J. PLAUR as a Potential Biomarker Associated with Immune Infiltration in Bladder Urothelial Carcinoma. J Inflamm Res 2021; 14:4629-4641. [PMID: 34552345 PMCID: PMC8450190 DOI: 10.2147/jir.s326559] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022] Open
Abstract
Background Bladder urothelial carcinoma (BLCA) is one of the most lethal and aggressive malignancies of genitourinary system that affects human health. The urokinase plasminogen activator receptor (PLAUR) plays essential roles in tumorigenesis and immune modulation, and its aberrant expression is closely correlated with cancer progression. However, whether PLAUR has the potential to be one promising biomarker or immunotherapy target for BLCA is unknown. Methodology Various online databases were applied to assess the expression profile and prognostic value of PLAUR, as well as its correlation with immune infiltration in BLCA, including Oncomine, PrognoScan, TCGA, cBioPortal, TIMER, TISIDB, UALCAN, and MethSurv. The expression of PLAUR in BLCA was confirmed with ELISA assay for serum samples and immunohistochemistry for tissue samples. Results The results showed that the expression of PLAUR was elevated in BLCA, which was further confirmed by ELISA and immunohistochemistry. Patients with higher PLAUR level were predicted to have lower overall survival and disease specific survival rates, which were not impacted by the genetic alterations of PLAUR. In addition, the expression of PLAUR was positively associated with immune infiltration, and also the expression levels of gene markers of various immune cells. The negative correlation between PLAUR expression and PLAUR methylation level was observed, among which PLAUR expression was positively correlated with the abundance of 28 kinds of tumor-infiltrating lymphocytes, while PLAUR methylation level was negatively correlated with the abundance of 11 types of tumor-infiltrating lymphocytes. Moreover, the methylation level of PLAUR was closely correlated with patients’ clinicopathological features, and hypomethylation of PLAUR was associated with better outcomes of BLCA patients. Conclusion These findings suggested that PLAUR had the potential to serve as a valuable detection and prognostic biomarker or immunotherapeutic target for BLCA.
Collapse
Affiliation(s)
- Mulin Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, People's Republic of China
| | - Siyi Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Aihui Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Qin Zheng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning Province, 116044, People's Republic of China
| | - Juan Fu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011, People's Republic of China
| |
Collapse
|
81
|
Qiu ZL, Yan BQ, Zhao R, Xu DW, Shen K, Deng XQ, Lu SQ. Combination of hepcidin with neutrophil gelatinase-associated lipocalin for prediction of the development of sepsis-induced acute kidney injury. Clin Chim Acta 2021; 523:38-44. [PMID: 34480953 DOI: 10.1016/j.cca.2021.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/15/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND AIMS The early prediction of the development of acute kidney injury (AKI) in critically ill patients with sepsis would facilitate early effective intervention. Recently, interest has focused on the biomarkers for AKI-linked iron metabolism. This study aimed to assess the early predictive values of hepcidin, neutrophil gelatinase-associated lipocalin (NGAL), and their combination for secondary AKI in patients with sepsis. MATERIALS AND METHODS A prospective cohort study was performed in septic patients. Serum and urine hepcidin, and urine NGAL were analyzed at admission. The primary outcome measure was occurrence of sepsis-induced AKI based on 2011 Kidney Disease: Improving Global Outcomes (KDIGO) criteria during the first week of ICU stay. RESULTS Of the 90 patients analyzed finally in the study, 44 (48.9%) patients developed AKI. Patients with AKI occurrence were more likely than those without AKI to have higher serum hepcidin and urine NGAL levels at admission (P < 0.01). Higher concentrations of these biomarkers were each independent predictor of the development of AKI in critically septic patients within the first week of their ICU stay. Serum hepcidin and urine NGAL (AUROC 0.787, 95% CI 0.688 to 0.8660 and AUROC 0.729, 95% CI 0.625 to 0.818, respectively) were comparable predictive indicators of AKI occurrence (P = 0.43 for DeLong's test). Combining both biomarkers increased the AUROC to 0.828(95% CI 0.733 to 0.899), and this performance was statistically significantly better than urine NGAL alone (P = 0.03 for DeLong's test). CONCLUSION Serum hepcidin measured at admission predicts the development of AKI similarly to urine NGAL. However, serum hepcidin adds significant accuracy to this prediction in combination with urine NGAL alone and has a good predictive value in patients with sepsis. Larger studies are needed to validate and explain these findings.
Collapse
Affiliation(s)
- Ze-Liang Qiu
- Department of Emergency, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Critical Care Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Bi-Qing Yan
- Intensive Care Unit, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Rui Zhao
- Department of Critical Care Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Dong-Wei Xu
- Department of Critical Care Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Kan Shen
- Department of Critical Care Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Xing-Qi Deng
- Department of Critical Care Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 201318, China
| | - Shi-Qi Lu
- Department of Emergency, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
82
|
Reisinger AC, Niedrist T, Posch F, Hatzl S, Hackl G, Prattes J, Schilcher G, Meißl AM, Raggam RB, Herrmann M, Eller P. Soluble urokinase plasminogen activator receptor (suPAR) predicts critical illness and kidney failure in patients admitted to the intensive care unit. Sci Rep 2021; 11:17476. [PMID: 34471146 PMCID: PMC8410930 DOI: 10.1038/s41598-021-96352-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is an inflammatory biomarker and risk factor for kidney diseases, with a potential prognostic value in critically ill patients. In this monocentric prospective study, we measured plasma suPAR levels immediately after ICU admission in unselected 237 consecutive patients using a turbidimetric assay. Primary objective was the prognostic value for ICU- and 28-day mortality. Secondary objectives were association with sequential organ failure assessment (SOFA) score, coagulation and inflammation markers, AKI-3 and differences in prespecified subgroups. Median suPAR levels were 8.0 ng/mL [25th-75th percentile 4.3-14.4], with lower levels in ICU survivors than non-survivors (6.7 vs. 11.6 ng/mL, p < 0.001). SuPAR levels were higher in COVID-19, kidney disease, moderate-to-severe liver disease, and sepsis. ICU mortality increased by an odds ratio (OR) of 4.7 in patients with the highest compared to lowest quartile suPAR. Kaplan-Meier overall survival estimates at 3 months were 63% and 49%, in patients with suPAR below/above 12 ng/mL (log-rank p = 0.027). Due to an observed interaction between SOFA score and suPAR, we performed a random forest method identifying cutoffs. ICU mortality was 53%, 17% and 2% in patients with a SOFA score > 7, SOFA ≤ 7 & suPAR > 8 ng/mL, and SOFA score ≤ 7 & suPAR ≤ 8 ng/mL, respectively. suPAR was a significant predictor for AKI-3 occurrence (OR per doubling 1.89, 95% CI: 1.20-2.98; p = 0.006). suPAR levels at ICU admission may offer additional value for risk stratification especially in ICU patients with moderate organ dysfunction as reflected by a SOFA score ≤ 7.
Collapse
Affiliation(s)
- Alexander C. Reisinger
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Tobias Niedrist
- grid.11598.340000 0000 8988 2476Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Florian Posch
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Stefan Hatzl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria ,grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Gerald Hackl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Juergen Prattes
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Section of Infectious Diseases and Tropical Medicine, Medical University of Graz, Graz, Austria
| | - Gernot Schilcher
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Anna-Maria Meißl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Reinhard B. Raggam
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Angiology, Medical University of Graz, Graz, Austria
| | - Markus Herrmann
- grid.11598.340000 0000 8988 2476Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| |
Collapse
|
83
|
Commentary: Quality metrics are important, but we must also become stewards of health care value. J Thorac Cardiovasc Surg 2021; 162:889-891. [DOI: 10.1016/j.jtcvs.2020.02.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 11/18/2022]
|
84
|
Abstract
Serum creatinine and level of proteinuria, as biomarkers of chronic kidney disease (CKD) progression, inadequately explain the variability of glomerular filtration rate decline, and are late markers of glomerular filtration rate decline. Recent studies have identified plasma and urine biomarkers at higher levels in children with CKD and also associate independently with CKD progression, even after adjustment for serum creatinine and proteinuria. These novel biomarkers represent diverse biologic pathways of tubular injury, tubular dysfunction, inflammation, and tubular health, and can be used as a liquid biopsy to better characterize CKD in children. In this review, we highlight the biomarker findings from the Chronic Kidney Disease in Children cohort, a large longitudinal study of children with CKD, and compare results with those from other pediatric CKD cohorts. The biomarkers in focus in this review include plasma kidney injury molecule-1, monocyte chemoattractant protein-1, fibroblast growth factor-23, tumor necrosis factor receptor-1, tumor necrosis factor receptor-2, soluble urokinase plasminogen activator receptor, and chitinase-3-like protein 1, as well as urine epidermal growth factor, α-1 microglobulin, kidney injury molecule-1, monocyte chemoattractant protein-1, and chitinase-3-like protein 1. Blood and urine biomarkers improve our ability to prognosticate CKD progression and may improve our understanding of CKD pathophysiology. Further research is required to establish how these biomarkers can be used in the clinical setting to improve the clinical management of CKD.
Collapse
Affiliation(s)
- Ibrahim Sandokji
- Section of Nephrology, Clinical and Translational Research Accelerator, Department of Pediatrics, Yale University School of Medicine, New Haven, CT; Department of Pediatrics, Taibah University College of Medicine, Medina, Saudi Arabia
| | - Jason H Greenberg
- Section of Nephrology, Clinical and Translational Research Accelerator, Department of Pediatrics, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
85
|
Walls AB, Bengaard AK, Iversen E, Nguyen CN, Kallemose T, Juul-Larsen HG, Jawad BN, Hornum M, Andersen O, Eugen-Olsen J, Houlind MB. Utility of suPAR and NGAL for AKI Risk Stratification and Early Optimization of Renal Risk Medications among Older Patients in the Emergency Department. Pharmaceuticals (Basel) 2021; 14:843. [PMID: 34577543 PMCID: PMC8471084 DOI: 10.3390/ph14090843] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 12/29/2022] Open
Abstract
Diagnosis of acute kidney injury (AKI) based on plasma creatinine often lags behind actual changes in renal function. Here, we investigated early detection of AKI using the plasma soluble urokinase plasminogen activator receptor (suPAR) and neutrophil gelatinase-sssociated lipocalin (NGAL) and observed the impact of early detection on prescribing recommendations for renally-eliminated medications. This study is a secondary analysis of data from the DISABLMENT cohort on acutely admitted older (≥65 years) medical patients (n = 339). Presence of AKI according to kidney disease: improving global outcomes (KDIGO) criteria was identified from inclusion to 48 h after inclusion. Discriminatory power of suPAR and NGAL was determined by receiver-operating characteristic (ROC). Selected medications that are contraindicated in AKI were identified in Renbase®. A total of 33 (9.7%) patients developed AKI. Discriminatory power for suPAR and NGAL was 0.69 and 0.78, respectively, at a cutoff of 4.26 ng/mL and 139.5 ng/mL, respectively. The interaction of suPAR and NGAL yielded a discriminatory power of 0.80, which was significantly higher than for suPAR alone (p = 0.0059). Among patients with AKI, 22 (60.6%) used at least one medication that should be avoided in AKI. Overall, suPAR and NGAL levels were independently associated with incident AKI and their combination yielded excellent discriminatory power for risk determination of AKI.
Collapse
Affiliation(s)
- Anne Byriel Walls
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (A.B.W.); (A.K.B.); (C.N.N.)
- The Capital Region Pharmacy, 2730 Herlev, Denmark
| | - Anne Kathrine Bengaard
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (A.B.W.); (A.K.B.); (C.N.N.)
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
| | - Camilla Ngoc Nguyen
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (A.B.W.); (A.K.B.); (C.N.N.)
- The Capital Region Pharmacy, 2730 Herlev, Denmark
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
| | - Helle Gybel Juul-Larsen
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
| | - Baker Nawfal Jawad
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark;
- Emergency Department, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark
| | - Mads Hornum
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark;
- Department of Nephrology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark;
- Emergency Department, Copenhagen University Hospital—Amager and Hvidovre, 2650 Hvidovre, Denmark
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
| | - Morten Baltzer Houlind
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (A.B.W.); (A.K.B.); (C.N.N.)
- The Capital Region Pharmacy, 2730 Herlev, Denmark
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark; (E.I.); (T.K.); (H.G.J.-L.); (B.N.J.); (O.A.); (J.E.-O.)
| |
Collapse
|
86
|
Altintas I, Eugen-Olsen J, Seppälä S, Tingleff J, Stauning MA, El Caidi NO, Elmajdoubi S, Gamst-Jensen H, Lindstrøm MB, Rasmussen LJH, Kristiansen KT, Rasmussen C, Nehlin JO, Kallemose T, Hyppölä H, Andersen O. suPAR Cut-Offs for Risk Stratification in Patients With Symptoms of COVID-19. Biomark Insights 2021; 16:11772719211034685. [PMID: 34421295 PMCID: PMC8371731 DOI: 10.1177/11772719211034685] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/02/2021] [Indexed: 01/14/2023] Open
Abstract
Objectives Elevated soluble urokinase Plasminogen Activator Receptor (suPAR) is a biomarker associated with adverse outcomes. We aimed to investigate the associations between plasma suPAR levels (testing the cut-offs ⩽4, 4-6, and ⩾6 ng/mL) with risk of 14-day mortality, and with the risk of mechanical ventilation in patients that tested positive for SARS-CoV-2. Methods Observational cohort study of patients presenting with symptoms of COVID-19 at Department of Emergency Medicine, Amager and Hvidovre Hospital, Denmark from March 19th, 2020 to April 3rd, 2020. Plasma suPAR was measured using suPARnostic technologies. Patients were followed for development of mechanical ventilation and mortality for 14 days. Validation of our findings were carried out in a similar sized COVID-19 patient cohort from Mikkeli Central Hospital, Finland. Results Among 386 patients with symptoms of COVID-19, the median (interquartile range) age was 64 years (46-77), 57% were women, median suPAR was 4.0 ng/mL (2.7-5.9). In total, 35 patients (9.1%) died during the 14 days follow-up. Patients with suPAR ⩽4 ng/mL (N = 196; 50.8%) had a low risk of mortality (N = 2; 1.0%; negative predictive value of 99.0%, specificity 55.3%, sensitivity 95.2%, positive predictive value 17.4%). Among patients with suPAR ⩾6 ng/mL (N = 92; 23.8%), 16 died (17.4%). About 99 patients (25.6%) tested positive for SARS CoV-2 and of those 12 (12.1%) developed need for mechanical ventilation. None of the SARS-CoV-2 positive patients with suPAR ⩽4 ng/mL (N = 28; 38.8%) needed mechanical ventilation or died. The Mikkeli Central Hospital validation cohort confirmed our findings concerning suPAR cut-offs for risk of development of mechanical ventilation and mortality. Conclusions Patients with symptoms of COVID-19 and suPAR ⩽4 or ⩾6 ng/mL had low or high risk, respectively, concerning the need for mechanical ventilation or mortality. We suggest cut-offs for identification of risk groups in patients presenting to the ED with symptoms of or confirmed COVID-19.
Collapse
Affiliation(s)
- Izzet Altintas
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark.,Department of Emergency Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Santeri Seppälä
- Emergency Department, Mikkeli Central Hospital, Mikkeli, Finland
| | - Jens Tingleff
- Department of Emergency Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Marius Ahm Stauning
- Department of Emergency Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Nora Olsen El Caidi
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Sanaá Elmajdoubi
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Hejdi Gamst-Jensen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark.,Department of Emergency Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Mette B Lindstrøm
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark.,Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Klaus Tjelle Kristiansen
- Department of Anesthesiology Intensive Care Unit, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Christian Rasmussen
- Department of Emergency Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Harri Hyppölä
- Emergency Department, Mikkeli Central Hospital, Mikkeli, Finland
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark.,Department of Emergency Medicine, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
87
|
Zhao L, Yu S, Wang L, Zhang X, Hou J, Li X. Blood suPAR, Th1 and Th17 cell may serve as potential biomarkers for elderly sepsis management. Scandinavian Journal of Clinical and Laboratory Investigation 2021; 81:488-493. [PMID: 34369213 DOI: 10.1080/00365513.2021.1952483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Although soluble urokinase plasminogen activator receptor (suPAR), T helper (Th)-1 and Th17 cells are separately reported to be dysregulated and correlate with disease severity in several infection-mediated diseases, fewer studies report their interaction and clinical value in sepsis management. Thus, this study aimed to investigate the correlation of blood suPAR with Th1 and Th17 cell proportion, as well as their diagnostic and prognostic value in elderly sepsis patients. Totally, 223 elderly sepsis patients were recruited. Serum suPAR was detected by enzyme-linked immunosorbent assay. Besides, Th1 and Th17 cell proportion from CD4+ T cells were determined by flow cytometry. For sepsis severity evaluation, Acute Physiology and Chronic Health Evaluation (APACHE) II score and the Sequential Organ Failure Assessment (SOFA) score were used. Moreover, survival profile within 28 days was documented. The mean value of suPAR, Th1 cell proportion and Th17 cell proportion was 27.5 ± 15.1 ng/mL, 15.3 ± 4.3% and 4.0 ± 2.3%, respectively. Furthermore, suPAR was positively correlated with Th1 cell proportion, Th17 cell proportion, IFN-γ, IL-17 and TNF-α. Meanwhile, suPAR was positively correlated with APACHE II score and SOFA score, so did Th17 cell proportion. Regarding their prognostic value, suPAR and Th17 cell proportion were superior to differ survivors from deaths than Th1 cell proportion. SuPAR positively correlates with Th1, Th17 cell proportion; and they correlate with increased disease severity and mortality risk in elderly sepsis patients.
Collapse
Affiliation(s)
- Lixin Zhao
- The Second Department of Critical Care Medicine, Tangshan Grongren Hospital, Tangshan, China
| | - Sifang Yu
- Department of Critical Care Medicine, Tangshan Central Hospital, Tangshan, China
| | - Lin Wang
- The Second Department of Critical Care Medicine, Tangshan Grongren Hospital, Tangshan, China
| | - Xiuping Zhang
- Department of General Surgery and Urology, Caofeidian District Hospital, Tangshan, China
| | - Junming Hou
- The Second Department of Critical Care Medicine, Tangshan Grongren Hospital, Tangshan, China
| | - Xuesong Li
- The Second Department of Critical Care Medicine, Tangshan Grongren Hospital, Tangshan, China
| |
Collapse
|
88
|
Zhu K, Reiser J. ALKBH1 reduces DNA N6-methyladenine to allow for vascular calcification in chronic kidney disease. J Clin Invest 2021; 131:e150966. [PMID: 34263739 DOI: 10.1172/jci150966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Vascular calcification is a common complication of chronic kidney disease (CKD), and one of the main risk factors for increased cardiovascular morbidity and mortality in patients with CKD. In this issue of the JCI, Ouyang and Su et al. report that Alkb homolog 1 (ALKBH1), a DNA demethylase, reduced DNA N6-methyladenine (6mA) in vascular smooth muscle cells (VSMCs) and leukocytes, thus leading to aortic arch calcification in the patients with CKD. During the progression of vascular calcification, increased ALKBH1 expression was linked to decreased 6mA levels, findings that the authors noted in both patients with CKD and CKD mouse models. The kidney and vascular disease risk factor soluble urokinase receptor (suPAR) was also elevated in the plasma. Notably, lower 6mA levels induced BMP2-mediated osteogenic reprogramming in the VSMCs. These findings present a function of ALKBH1 in vascular calcification and provide a framework for therapeutic strategies.
Collapse
|
89
|
Ouyang L, Su X, Li W, Tang L, Zhang M, Zhu Y, Xie C, Zhang P, Chen J, Huang H. ALKBH1-demethylated DNA N6-methyladenine modification triggers vascular calcification via osteogenic reprogramming in chronic kidney disease. J Clin Invest 2021; 131:146985. [PMID: 34003800 PMCID: PMC8279589 DOI: 10.1172/jci146985] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
Vascular calcification (VC) predicts cardiovascular morbidity and mortality in chronic kidney disease (CKD). To date, the underlying mechanisms remain unclear. We detected leukocyte DNA N6-methyladenine (6mA) levels in patients with CKD with or without aortic arch calcification. We used arteries from CKD mice infected with vascular smooth muscle cell-targeted (VSMC-targeted) adeno-associated virus encoding alkB homolog 1 (Alkbh1) gene or Alkbh1 shRNA to evaluate features of calcification. We identified that leukocyte 6mA levels were significantly reduced as the severity of VC increased in patients with CKD. Decreased 6mA demethylation resulted from the upregulation of ALKBH1. Here, ALKBH1 overexpression aggravated whereas its depletion blunted VC progression and osteogenic reprogramming in vivo and in vitro. Mechanistically, ALKBH1-demethylated DNA 6mA modification could facilitate the binding of octamer-binding transcription factor 4 (Oct4) to bone morphogenetic protein 2 (BMP2) promoter and activate BMP2 transcription. This resulted in osteogenic reprogramming of VSMCs and subsequent VC progression. Either BMP2 or Oct4 depletion alleviated the procalcifying effects of ALKBH1. This suggests that targeting ALKBH1 might be a therapeutic method to reduce the burden of VC in CKD.
Collapse
Affiliation(s)
- Liu Ouyang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaoyan Su
- Department of Nephropathy, Tungwah Hospital, Sun Yat-sen University, Dongguan, China
| | - Wenxin Li
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Liangqiu Tang
- Department of Cardiology, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Mengbi Zhang
- Department of Nephropathy, Tungwah Hospital, Sun Yat-sen University, Dongguan, China
| | - Yongjun Zhu
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Changming Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Puhua Zhang
- Department of Nephrology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jie Chen
- Department of Radiation Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
90
|
Rasmussen LJH, Caspi A, Ambler A, Danese A, Elliott M, Eugen-Olsen J, Hariri AR, Harrington H, Houts R, Poulton R, Ramrakha S, Sugden K, Williams B, Moffitt TE. Association Between Elevated suPAR, a New Biomarker of Inflammation, and Accelerated Aging. J Gerontol A Biol Sci Med Sci 2021; 76:318-327. [PMID: 32766674 PMCID: PMC7812430 DOI: 10.1093/gerona/glaa178] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background To understand and measure the association between chronic inflammation, aging, and age-related diseases, broadly applicable standard biomarkers of systemic chronic inflammation are needed. We tested whether elevated blood levels of the emerging chronic inflammation marker soluble urokinase plasminogen activator receptor (suPAR) were associated with accelerated aging, lower functional capacity, and cognitive decline. Methods We used data from the Dunedin Study, a population-representative 1972–1973 New Zealand birth cohort (n = 1037) that has observed participants to age 45 years. Plasma suPAR levels were analyzed at ages 38 and 45 years. We performed regression analyses adjusted for sex, smoking, C-reactive protein, and current health conditions. Results Of 997 still-living participants, 875 (88%) had plasma suPAR measured at age 45. Elevated suPAR was associated with accelerated pace of biological aging across multiple organ systems, older facial appearance, and with structural signs of older brain age. Moreover, participants with higher suPAR levels had greater decline in physical function and cognitive function from childhood to adulthood compared to those with lower suPAR levels. Finally, improvements in health habits between ages 38 and 45 (smoking cessation or increased physical activity) were associated with less steep increases in suPAR levels over those years. Conclusions Our findings provide initial support for the utility of suPAR in studying the role of chronic inflammation in accelerated aging and functional decline.
Collapse
Affiliation(s)
- Line Jee Hartmann Rasmussen
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina.,Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina.,Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK
| | - Antony Ambler
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Andrea Danese
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK.,National and Specialist Child and Adolescent Mental Health Services Trauma, Anxiety, and Depression Clinic, South London and Maudsley National Health Service Foundation Trust, London, UK
| | - Maxwell Elliott
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - HonaLee Harrington
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Renate Houts
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Richie Poulton
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Sandhya Ramrakha
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Benjamin Williams
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina.,Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK
| |
Collapse
|
91
|
Bengaard AK, Iversen E, Kallemose T, Juul-Larsen HG, Rasmussen LJH, Dalhoff KP, Andersen O, Eugen-Olsen J, Houlind MB. Using soluble urokinase plasminogen activator receptor to stratify patients for medication review in the emergency department. Br J Clin Pharmacol 2021; 88:1679-1690. [PMID: 34242432 DOI: 10.1111/bcp.14982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 11/30/2022] Open
Abstract
AIMS To investigate whether the association between levels of medication use (including polypharmacy and potentially inappropriate medications [PIMs]) and health outcomes such as readmission and mortality is dependent on baseline soluble urokinase plasminogen activator receptor (suPAR). METHODS This registry-based cohort study included medical patients admitted to the emergency department at Copenhagen University Hospital Hvidovre, Denmark. Patients were grouped according to their admission suPAR levels: low (0-3 ng/mL), intermediate (3-6 ng/mL), or high (>6 ng/mL). Hyper-polypharmacy was defined as ≥10 prescribed medications. PIMs were identified based on the EU(7)-PIM list, and data on admissions and mortality were obtained from national registries. Risk of 90-day readmission and mortality was assessed by Cox regression analysis adjusted for sex, age and Charlson comorbidity index. Results were reported as hazard ratios within 90 days of index discharge. RESULTS In total, 26 291 patients (median age 57.3 y; 52.7% female) were included. Risk of 90-day readmission and mortality increased significantly for patients with higher suPAR or higher number of medications. Among patients with low suPAR, patients with ≥10 prescribed medications had a hazard ratio of 2.41 (95% confidence interval = 2.09-2.78) for 90-day readmission and 8.46 (95% confidence interval = 2.53-28.28) for 90-day mortality compared to patients with 0 medications. Patients with high suPAR generally had high risk of readmission and mortality, and the impact of medication use was less pronounced in this group. Similar, but weaker, association patterns were observed between suPAR and PIMs. CONCLUSION The association between levels of medication use and health outcomes is dependent on baseline suPAR.
Collapse
Affiliation(s)
- Anne Kathrine Bengaard
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,The Capital Region Pharmacy, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Helle Gybel Juul-Larsen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Kim Peder Dalhoff
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Emergency Department, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,The Capital Region Pharmacy, Herlev, Denmark.,Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
92
|
Acute kidney injury in the critically ill: an updated review on pathophysiology and management. Intensive Care Med 2021; 47:835-850. [PMID: 34213593 PMCID: PMC8249842 DOI: 10.1007/s00134-021-06454-7] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/04/2021] [Indexed: 01/10/2023]
Abstract
Acute kidney injury (AKI) is now recognized as a heterogeneous syndrome that not only affects acute morbidity and mortality, but also a patient’s long-term prognosis. In this narrative review, an update on various aspects of AKI in critically ill patients will be provided. Focus will be on prediction and early detection of AKI (e.g., the role of biomarkers to identify high-risk patients and the use of machine learning to predict AKI), aspects of pathophysiology and progress in the recognition of different phenotypes of AKI, as well as an update on nephrotoxicity and organ cross-talk. In addition, prevention of AKI (focusing on fluid management, kidney perfusion pressure, and the choice of vasopressor) and supportive treatment of AKI is discussed. Finally, post-AKI risk of long-term sequelae including incident or progression of chronic kidney disease, cardiovascular events and mortality, will be addressed.
Collapse
|
93
|
Kim EY, Dryer SE. RAGE and αVβ3-integrin are essential for suPAR signaling in podocytes. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166186. [PMID: 34166766 DOI: 10.1016/j.bbadis.2021.166186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 12/25/2022]
Abstract
The soluble urokinase plasminogen activator receptor (suPAR) has been implicated in the pathogenesis of kidney diseases including primary and recurrent focal and segmental glomerulosclerosis (FSGS), diabetic nephropathy, and acute kidney injuries (AKI). Elevated serum suPAR concentration is a negative prognostic indicator in multiple critical clinical conditions. This study has examined the initial transduction steps used by suPAR in cultured mouse podocytes. We now report that the receptor for advanced glycation end-products (RAGE) co-immunoprecipitates with αV and β3 integrin subunits, which have been previously shown to initiate suPAR signal transduction at the podocyte cell surface. siRNA knock-down of RAGE attenuated Src phosphorylation evoked by either suPAR or by glycated albumin (AGE-BSA), a prototypical RAGE agonist. suPAR effects on Src phosphorylation were also blocked by the structurally dissimilar RAGE antagonists FPS-ZM1 and azeliragon, as well as by cilengitide, an inhibitor of outside-in signaling through αV-integrins. FPS-ZM1 also blocked Src phosphorylation evoked by AGE-BSA. FPS-ZM1 blocked increases in cell surface TRPC6 abundance, cytosolic reactive oxygen species (ROS) and activation of the small GTPase Rac1 evoked by either suPAR or AGE-BSA. In addition, FPS-ZM1 inhibited Src phosphorylation evoked by serum collected from a patient with recurrent FSGS during a relapse. The magnitude of this inhibition was indistinguishable from the effect produced by a neutralizing antibody against suPAR. These data suggest that orally bioavailable small molecule RAGE antagonists could represent a useful therapeutic strategy for a wide range of clinical conditions associated with elevated serum suPAR, including primary FSGS and AKI.
Collapse
Affiliation(s)
- Eun Young Kim
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA.
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA; Department of Biomedical Sciences, University of Houston College of Medicine, Houston, TX 77204, USA.
| |
Collapse
|
94
|
Eleven genomic loci affect plasma levels of chronic inflammation marker soluble urokinase-type plasminogen activator receptor. Commun Biol 2021; 4:655. [PMID: 34079037 PMCID: PMC8172928 DOI: 10.1038/s42003-021-02144-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
Soluble urokinase-type plasminogen activator receptor (suPAR) is a chronic inflammation marker associated with the development of a range of diseases, including cancer and cardiovascular disease. The genetics of suPAR remain unexplored but may shed light on the biology of the marker and its connection to outcomes. We report a heritability estimate of 60% for the variation in suPAR and performed a genome-wide association meta-analysis on suPAR levels measured in Iceland (N = 35,559) and in Denmark (N = 12,177). We identified 13 independently genome-wide significant sequence variants associated with suPAR across 11 distinct loci. Associated variants were found in and around genes encoding uPAR (PLAUR), its ligand uPA (PLAU), the kidney-disease-associated gene PLA2R1 as well as genes with relations to glycosylation, glycoprotein biosynthesis, and the immune response. These findings provide new insight into the causes of variation in suPAR plasma levels, which may clarify suPAR's potential role in associated diseases, as well as the underlying mechanisms that give suPAR its prognostic value as a unique marker of chronic inflammation.
Collapse
|
95
|
Clendenen N, Ahlgren B, Robitaille MJ, Christensen E, Morabito J, Grae L, Lyman M, Weitzel N. Year in Review 2020: Noteworthy Literature in Cardiothoracic Anesthesiology. Semin Cardiothorac Vasc Anesth 2021; 25:94-106. [PMID: 33938302 PMCID: PMC10088871 DOI: 10.1177/10892532211013614] [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] [Indexed: 12/15/2022]
Abstract
The year 2020 was marred by the emergence of a deadly pandemic that disrupted every aspect of life. Despite the disruption, notable research accomplishments in the practice of cardiothoracic anesthesiology occurred in 2020 with an emphasis on optimizing care, improving outcomes, and expanding what is possible for patients undergoing cardiac surgery. This year's edition of Noteworthy Literature Review will focus on specific themes in cardiac anesthesiology that include preoperative anemia, predictors of acute kidney injury following cardiac surgery, pain management modalities, anticoagulation strategies after transcatheter aortic valve replacement, mechanical circulatory support, and future directions in research.
Collapse
Affiliation(s)
| | - Bryan Ahlgren
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mark J Robitaille
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Joseph Morabito
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lyndsey Grae
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew Lyman
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nathaen Weitzel
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
96
|
Pre-operative N-terminal pro-B-type natriuretic peptide for prediction of acute kidney injury after noncardiac surgery: A retrospective cohort study. Eur J Anaesthesiol 2021; 38:591-599. [PMID: 33720062 DOI: 10.1097/eja.0000000000001495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) is associated with poor outcomes after noncardiac surgery. Whether pre-operative N-terminal pro-B-type natriuretic peptide (NT-proBNP) predicts AKI after noncardiac surgery is unclear. OBJECTIVE To investigate the predictive role of pre-operative NT-proBNP on postoperative AKI. DESIGN Retrospective cohort study. SETTING Nanfang Hospital, Southern Medical University, China. PATIENTS Adult patients who had a serum creatinine and NT-proBNP measurement within 30 pre-operative days and at least one serum creatinine measurement within 7 days after noncardiac surgery between February 2008 and May 2018 were identified. MAIN OUTCOME MEASURES The primary outcome was postoperative AKI, defined by the kidney disease: improving global outcomes creatinine criteria. RESULTS In all, 6.1% (444 of 7248) of patients developed AKI within 1 week after surgery. Pre-operative NT-proBNP was an independent predictor of AKI after adjustment for clinical variables (OR comparing top to bottom quintiles 2.29, 95% CI, 1.47 to 3.65, P < 0.001 for trend; OR per 1-unit increment in natural log transformed NT-proBNP 1.27, 95% CI, 1.16 to 1.39). Compared with clinical variables alone, the addition of NT-proBNP improved model fit, modestly improved the discrimination (change in area under the curve from 0.764 to 0.773, P = 0.005) and reclassification (continuous net reclassification improvement 0.210, 95% CI, 0.111 to 0.308, improved integrated discrimination 0.0044, 95% CI, 0.0016 to 0.0072) of AKI and non-AKI cases, and achieved higher net benefit in decision curve analysis. CONCLUSIONS Pre-operative NT-proBNP concentrations provided predictive information for AKI in a cohort of patients undergoing noncardiac surgery, independent of and incremental to conventional risk factors. Prospective studies are required to confirm this finding and examine its clinical impact. TRIAL REGISTRATION Chinese Clinical Trial Registry, ChiCTR1900024056. www.chictr.org.cn/showproj.aspx?proj=40385.
Collapse
|
97
|
Oulhaj A, Alsuwaidi AR, Suliman A, Gasmelseed H, Khan S, Alawi S, Hukan Y, George J, Alshamsi F, Sheikh F, Babiker ZOE, Prattes J, Sourij H. Admission levels of Soluble Urokinase Plasminogen Activator Receptor (suPAR) are Associated with the Development of Severe Complications in Hospitalised COVID-19 Patients: A Prospective Cohort Study. Int J Infect Dis 2021; 107:188-194. [PMID: 33862208 PMCID: PMC8056823 DOI: 10.1016/j.ijid.2021.04.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
Objective To examine the association between plasma levels of the soluble urokinase plasminogen activator receptor (suPAR) and the incidence of severe complications of COVID-19. Methods 403 RT-PCR-confirmed COVID-19 patients were recruited and prospectively followed-up at a major hospital in the United Arab Emirates. The primary endpoint was time from admission until the development of a composite outcome, including acute respiratory distress syndrome (ARDS), intensive care unit (ICU) admission, or death from any cause. Patients discharged alive were considered as competing events to the primary outcome. Competing risk regression was used to quantify the association between suPAR and the incidence of the primary outcome. Results 6.2% of patients experienced ARDS or ICU admission, but none died. Taking into account competing risk, the incidence of the primary outcome was 11.5% (95% confidence interval [CI], 6.7–16.3) in patients with suPAR levels >3.91 ng/mL compared to 2.9% (95% CI, 0.4–5.5) in those with suPAR ≤3.91 ng/mL. Also, an increase by 1 ng/mL in baseline suPAR resulted in a 58% rise in the hazard of developing the primary outcome (hazard ratio 1.6, 95% CI, 1.2–2.1, p = 0.003). Conclusion suPAR has an excellent prognostic utility in predicting severe complications in hospitalised COVID-19 patients.
Collapse
Affiliation(s)
- Abderrahim Oulhaj
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates; Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates.
| | - Ahmed R Alsuwaidi
- Department of Paediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Abubaker Suliman
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Shaima Khan
- Academic Affairs Department, Tawam Hospital, Al Ain, United Arab Emirates
| | - Shamma Alawi
- Academic Affairs Department, Tawam Hospital, Al Ain, United Arab Emirates
| | - Yaman Hukan
- Academic Affairs Department, Tawam Hospital, Al Ain, United Arab Emirates
| | - Junu George
- Department of Paediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Fayez Alshamsi
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Zahir Osman Eltahir Babiker
- Division of Infectious Diseases, Sheikh Shakhbout Medical City in Partnership with Mayo Clinic, Abu Dhabi, United Arab Emirates
| | - Juergen Prattes
- Department of Internal Medicine, Section of Infectious Diseases and Tropical Medicine, Medical University of Graz, Austria
| | - Harald Sourij
- Zayed Center for Health Sciences, United Arab Emirates University, United Arab Emirates; Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Austria
| |
Collapse
|
98
|
Duan YC, Shi L, Jin Z, Hu M, Huang H, Yan T, Zhang KR. Swimming Exercise Ameliorates Hypertension-Induced Kidney Dysfunction via Alleviating Renal Interstitial Fibrosis and Apoptosis. Kidney Blood Press Res 2021; 46:219-228. [PMID: 33849006 DOI: 10.1159/000514680] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/23/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Hypertensive nephropathy is one of the major causes of ESRD. Exercise has been considered a nonpathological therapy for hypertension and its complications, yet mechanisms remain unclear. We sought to investigate whether periodic swimming could ameliorate hypertension-induced kidney dysfunction and its underlying mechanisms. METHODS Four-week male spontaneously hypertensive rats (SHRs) were randomly divided into the hypertension group (SHR, n = 8) and exercise group (SE, n = 8, 60 min swimming/day, 6 days per week, for 8 weeks). Wistar-Kyoto rats (WKY, n = 8) were served as a sedentary normotensive group. Bodyweight and blood pressure (BP) were recorded weekly. After 8-week sedentary or swimming exercise, lipids profile, BUN, and Cr were measured. The renal interstitial fibrosis was examined by the histopathological analysis using Masson's trichrome staining and hematoxylin and eosin staining. The kidney cell apoptosis was tested by TUNEL staining. The expressions of critical proteins responsible for the TGF-β1/Smad signaling of fibrosis, that is, TGF-β1, Smad2/3, and Smad7, as well as apoptosis related proteins, Bax and Bcl-2 in kidney cortex tissues were measured. RESULTS The 8-week swimming exercise reduced BP and bodyweight, lowered concentrations of BUN, and serum Cr, compared with SHR. Exercise remarkably inhibited hypertension-induced tubular degeneration, cellular cluster, and tubular cell swelling as well as glomerular degeneration in the kidney cortical tissues, attenuated renal interstitial fibrosis, and renal cell apoptosis. Moreover, expressions of TGF-β1, Smad2/3, and Bax were higher in the SHR than the WKY, which were significantly suppressed by the exercise. In contrast, hypertension-reduced expressions of Smad7 and Bcl-2 were enhanced by the swimming exercise. Strong correlations were found between kidney function indices, blood lipids, and key protein expressions. CONCLUSION Our results demonstrate beneficial effects of the periodic swimming on ameliorating hypertension-induced kidney dysfunction highlighting the potential of swimming exercise as a nonpathological therapy for early prevention of hypertension-caused kidney diseases.
Collapse
Affiliation(s)
- Yong-Chang Duan
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Zheng Jin
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Meng Hu
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Hao Huang
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Tao Yan
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Kun-Ru Zhang
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
99
|
Rasmussen SR, Nielsen RV, Møgelvang R, Ostrowski SR, Ravn HB. Prognostic value of suPAR and hsCRP on acute kidney injury after cardiac surgery. BMC Nephrol 2021; 22:120. [PMID: 33827466 PMCID: PMC8025450 DOI: 10.1186/s12882-021-02322-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/24/2021] [Indexed: 11/16/2022] Open
Abstract
Background Acute kidney injury (AKI) represents a serious complication following cardiac surgery. Adverse outcome after cardiac surgery has been observed in the presence of elevated levels of soluble urokinase-type plasminogen activator receptor (suPAR) and high-sensitivity C-Reactive Protein (hsCRP). The aim of study was (i) to investigate the relationship between preoperative elevated levels of suPAR and hsCRP and postoperative AKI in unselected cardiac surgery patients and (ii) to assess whether the concentration of the biomarkers reflected severity of AKI. Methods In a retrospective observational study, biobank blood plasma samples (n = 924) from patients admitted for elective on-pump cardiac surgery were analysed for suPAR and hsCRP levels. The relation between suPAR and hsCRP-values and AKI (any stage), defined by the KDIGO (Kidney Disease: Improving Global Outcomes) criteria, was assessed using adjusted logistic regression. Further, the association between biomarkers and severity (KDIGO 1, KDIGO 2–3 and renal replacement therapy (RRT)) was assessed using adjusted logistic regression. Results Postoperative AKI (any stage) was observed in 327 patients (35.4 %). A doubling of preoperative suPAR corresponded to an adjusted odds ratio (OR) for postoperative AKI (any stage) of 1.62 (95 % CI 1.26–2.09, p < 0.001). Furthermore, a doubling of suPAR had an adjusted OR of 1.50 (95 % CI 1.16–1.93, p = 0.002), 2.44 (95 % CI 1.56–3.82, p < 0.001) and 1.92 (95 % CI 1.15–3.23, p = 0.002), for KDIGO 1, KDIGO 2–3 and need for RRT, respectively. No significant association was found between elevated levels of hsCRP and any degree of AKI. Conclusions Increasing levels of suPAR, but not hsCRP, were associated with development and severity of AKI following on-pump cardiac surgery.
Collapse
Affiliation(s)
- Sebastian Roed Rasmussen
- Department of Cardiothoracic Anaesthesiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Rikke Vibeke Nielsen
- Department of Cardiothoracic Anaesthesiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rasmus Møgelvang
- Department of Cardiothoracic Anaesthesiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Clinical Cardiovascular Research Unit, University of Southern Denmark, Svendborg, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hanne Berg Ravn
- Department of Cardiothoracic Anaesthesiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Anaesthesiology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
100
|
Jing L, Chen W, Guo L, Zhao L, Liang C, Chen J, Wang C. Acute kidney injury after lung transplantation: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:717. [PMID: 33987415 PMCID: PMC8106087 DOI: 10.21037/atm-20-7644] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Acute kidney injury (AKI) is a commonly recognized complication after lung transplantation (LT) and is related to increased mortality and morbidity. With the improvement of survival after LT and the increasing number of lung transplant recipients, the detrimental impact of current management on renal function has become increasingly apparent. Multifarious risk factors in the perioperative setting contribute to the development of AKI, including the preoperative status and complications of the recipient, complex perioperative problems especially hemodynamic fluctuation, and exposure to nephrotoxic agents, mainly calcineurin inhibitors (CNIs) and antimicrobial drugs. Identification and minimization of the effects of these risk factors can relieve AKI severity and incidence in high-risk patients. Close monitoring of urine output and serum creatinine (sCr) levels and of specific biomarkers may promote early recognition of AKI and rapid nephrology intervention to improve outcomes. This review summarizes advances in the epidemiology, diagnostic criteria, biological markers of AKI, and further recommends appropriate treatment strategies for the long-term management of AKI related manifestations in lung transplant recipients. Future work will need to focus on developing more accurate measures of renal function and identifying patients before the occurrence of early renal damage. Combining renal protection strategies with the use of new biomarkers to develop early kidney risk identification and protection protocols is a promising idea that requires further investigation.
Collapse
Affiliation(s)
- Lei Jing
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Lung Transplantation, Centre of Lung Transplantation, Centre of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| | - Wenhui Chen
- Department of Lung Transplantation, Centre of Lung Transplantation, Centre of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| | - Lijuan Guo
- Department of Lung Transplantation, Centre of Lung Transplantation, Centre of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| | - Li Zhao
- Department of Lung Transplantation, Centre of Lung Transplantation, Centre of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| | - Chaoyang Liang
- Department of Lung Transplantation, Centre of Lung Transplantation, Centre of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| | - Jingyu Chen
- Department of Lung Transplantation, Centre of Lung Transplantation, Centre of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| | - Chen Wang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Lung Transplantation, Centre of Lung Transplantation, Centre of Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China.,WHO Collaborating Centre for Tobacco Cessation and Respiratory Diseases Prevention, Beijing, China
| |
Collapse
|