1
|
Mathis MR, Mentz GB, Cao J, Balczewski EA, Janda AM, Likosky DS, Schonberger RB, Hawkins RB, Heung M, Ailawadi G, Ladhania R, Sjoding MW, Kheterpal S, Singh K. Hospital and Clinician Practice Variation in Cardiac Surgery and Postoperative Acute Kidney Injury. JAMA Netw Open 2025; 8:e258342. [PMID: 40314957 PMCID: PMC12048843 DOI: 10.1001/jamanetworkopen.2025.8342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 02/27/2025] [Indexed: 05/03/2025] Open
Abstract
Importance Approximately 30% of US patients develop acute kidney injury (AKI) after cardiac surgery, which is associated with increased morbidity, mortality, and health care costs. The variation in potentially modifiable hospital- and clinician-level operating room practices and their implications for AKI have not been rigorously evaluated. Objective To quantify variation in clinician- and hospital-level hemodynamic and resuscitative practices during cardiac surgery and identify their associations with AKI. Design, Setting, and Participants This cohort study analyzed integrated hospital, clinician, and patient data extracted from the Multicenter Perioperative Outcomes Group dataset and the Society of Thoracic Surgeons Adult Cardiac Surgical Database. Participants were adult patients (aged ≥18 years) who underwent cardiac surgical procedures between January 1, 2014, and February 1, 2022, at 8 geographically diverse US hospitals. Patients were followed up through March 2, 2022. Statistical analyses were performed from October 2024 to February 2025. Exposures Hospital- and clinician-level variations in operating room hemodynamic practices (inotrope infusion >60 minutes and vasopressor infusion >60 minutes) and resuscitative practices (homologous red blood cell [RBC] transfusion and total fluid volume administration). Main Outcomes and Measures The primary outcome was consensus guideline-defined AKI (any stage) within 7 days after cardiac surgery. Hospital- and clinician-level variations were quantified using intraclass correlation coefficients (ICCs). Associations of hospital- and clinician-level practices with AKI were analyzed using multilevel mixed-effects models, adjusting for patient-level characteristics. Results Among 23 389 patients (mean [SD] age, 63 [13] years; 16 122 males [68.9%]), 4779 (20.4%) developed AKI after cardiac surgery. AKI rates varied across hospitals (median [IQR], 21.7% [15.5%-27.2%]) and clinicians (18.1% [10.1%-23.7%]). Significant clinician- and hospital-level variation existed for inotrope infusion (ICC, 6.2% [95% CI, 4.2%-8.0%] vs 17.9% [95% CI, 3.3%-31.9%]), vasopressor infusion (ICC, 11.7% [95% CI, 8.3%-14.9%] vs 44.5% [95% CI, 11.7%-63.5%]), RBC transfusion (ICC, 1.7% [95% CI, 0.9%-2.6%] vs 4.5% [95% CI, 1.2%-9.4%]), and fluid volume administration (ICC, 2.1% [95% CI, 1.3%-2.7%] vs 23.8% [95% CI, 2.7%-39.9%]). In multilevel risk-adjusted models, the AKI rate was higher for patients at hospitals with higher inotrope infusion rates (adjusted odds ratio [AOR], 1.98; 95% CI, 1.18-3.33; P = .01) and lower among clinicians with higher RBC transfusion rates (AOR, 0.89; 95% CI, 0.79-0.99; P = .03). Other practice variations were not associated with AKI. Conclusions and Relevance This cohort study of adult patients found that hospital- and clinician-level variation in operating room practices was associated with AKI after cardiac surgery, suggesting possible targets for intervention.
Collapse
Affiliation(s)
- Michael R. Mathis
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor
- Department of Computational Bioinformatics, University of Michigan Medical School, Ann Arbor
| | - Graciela B. Mentz
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor
| | - Jie Cao
- Joan and Irwin Jacobs Center for Health Innovation, University of California San Diego
| | - Emily A. Balczewski
- Department of Computational Bioinformatics, University of Michigan Medical School, Ann Arbor
| | - Allison M. Janda
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor
| | - Donald S. Likosky
- Department of Cardiac Surgery, University of Michigan Medical School, Ann Arbor
| | | | - Robert B. Hawkins
- Department of Cardiac Surgery, University of Michigan Medical School, Ann Arbor
| | - Michael Heung
- Nephrology Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor
| | - Gorav Ailawadi
- Department of Cardiac Surgery, University of Michigan Medical School, Ann Arbor
| | - Rahul Ladhania
- Department of Health Management and Policy, University of Michigan, Ann Arbor
- Department of Biostatistics, University of Michigan, Ann Arbor
| | - Michael W. Sjoding
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor
| | - Sachin Kheterpal
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor
| | - Karandeep Singh
- Joan and Irwin Jacobs Center for Health Innovation, University of California San Diego
- Division of Biomedical Informatics, Department of Medicine, University of California, San Diego
| |
Collapse
|
2
|
Erixon C, Thelaus L, Johannesson E, Nilsson J, Teurneau-Hermansson K, Linder A, Ragnarsson S, Sterner N, Zindovic I, Dardashti A. The predictive value of postoperative soluble urokinase plasminogen activator receptor concentration for postoperative complications following valvular surgery. Scand J Clin Lab Invest 2025; 85:160-167. [PMID: 40088459 DOI: 10.1080/00365513.2025.2479042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is an inflammatory marker that has been shown to predict poorer outcomes in cardiovascular disease and after cardiac surgery. The relationship between suPAR concentrations and postoperative complications after valvular surgery, however, remains unclear. This study aims to evaluate the predictive value of suPAR concentrations for infection, acute kidney injury (AKI) and prolonged mechanical ventilation after valvular surgery. This prospective, observational, single-centre study included 414 patients who underwent valvular cardiac surgery at Skåne University Hospital between 1 February 2020 and 22 September 2021. Early postoperative suPAR levels were measured, and multivariable logistic regression was used to identify significant risk factors for postoperative infection, AKI and prolonged mechanical ventilation. Left ventricular ejection fraction (LVEF) 30-50% (OR 3.57 [1.29-9.86], p = 0.014) and suPAR concentration (OR 1.41 [1.56-1.71], p <0.001) were found to be predictive risk factors for developing postoperative infection. Additionally, suPAR concentration (OR 1.23 [1.05-1.43], p = 0.008), cardiopulmonary bypass (CPB) time (OR 1.01 [1.00-1.02], p = 0.004) and age (OR 1.04 [1.01-1.08], p = 0.007) were found to be predictive risk factors for postoperative AKI. However, suPAR concentration did not predict prolonged mechanical ventilation. Plasma suPAR levels after cardiac valve surgery were found to be predictive of postoperative AKI and infection. Our results indicate that early postoperative suPAR measurements may be a valuable tool for identifying patients at higher risk for developing postoperative complications.
Collapse
Affiliation(s)
- Clara Erixon
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Louise Thelaus
- Department of Infection Medicine, Institute of Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Emilia Johannesson
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Johan Nilsson
- Thoracic Surgery and Bioinformatics Research Unit, Institute of Clinical Sciences, Department of Translational Medicine, Lund University, Lund, Sweden
- Department of Thoracic and Vascular Surgery, Skåne University Hospital, Lund, Sweden
| | - Karl Teurneau-Hermansson
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Adam Linder
- Department of Infection Medicine, Institute of Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sigurdur Ragnarsson
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Niklas Sterner
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Igor Zindovic
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alain Dardashti
- Department of Cardiothoracic Surgery, Institute of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
3
|
Yang H, Chen Y, He J, Li Y, Feng Y. Advances in the diagnosis of early biomarkers for acute kidney injury: a literature review. BMC Nephrol 2025; 26:115. [PMID: 40045274 PMCID: PMC11884078 DOI: 10.1186/s12882-025-04040-3] [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: 01/02/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Acute kidney injury (AKI) is a critical condition with diverse manifestations and variable outcomes. Its diagnosis traditionally relies on delayed indicators such as serum creatinine and urine output, making early detection challenging. Early identification is essential to improving patient outcomes, driving the need for novel biomarkers. Recent advancements have identified promising biomarkers across various biological processes. Tubular injury markers, including neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), N-acetyl-β-D-glucosaminidase (NAG), and liver-type fatty acid-binding protein (L-FABP), offer insights into early tubular damage. Inflammatory and repair-associated biomarkers, such as interleukin-18 (IL-18), monocyte chemotactic protein-1 (MCP-1), osteopontin (OPN), and C-C motif chemokine ligand 14 (CCL14), reflect ongoing injury and recovery processes. Additionally, stress and repair markers like tissue inhibitor of metalloproteinase-2 (TIMP-2) and insulin-like growth factor-binding protein-7 (IGFBP-7), alongside filtration markers such as cystatin C (CysC) and proenkephalin (PenKid®) e.tal, further enhance diagnostic precision. Oxidative stress-related markers, including Superoxide Dismutase 1 (SOD1), also contribute valuable information. Emerging candidates, such as microRNAs, soluble urokinase plasminogen activator receptor (SuPAR), and chitinase-3-like protein 1 (CHI3L1), hold substantial promise for AKI detection and prognosis. This review summarizes the progress in AKI biomarker research, highlighting their clinical utility and exploring their potential to refine early diagnosis and management strategies. These findings offer a new perspective for integrating novel biomarkers into routine clinical practice, ultimately improving AKI care.
Collapse
Affiliation(s)
- Hongsha Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yanqin Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Jiajia He
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yi Li
- Department of Nephrology, Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yunlin Feng
- Department of Nephrology, Institute of Nephrology, Sichuan Provincial People's Hospital, Sichuan Clinical Research Centre for Kidney Diseases, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
4
|
Peng L, Li S, Huang Q, Sun Y, Sun J, Luo T, Wang Y, Hu Z, Lai W, Peng H. Irisin-mediated muscle-renal crosstalk as a protective mechanism against contrast-induced acute kidney injury via cGAS-STING signalling inhibition. Clin Transl Med 2025; 15:e70235. [PMID: 40008481 PMCID: PMC11862893 DOI: 10.1002/ctm2.70235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/17/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Contrast-induced acute kidney injury (CI-AKI) continues to pose a pressing clinical challenge during invasive cardiovascular procedures due to the limited availability of preventative strategies. We aimed to demonstrate that irisin, a myokine induced by exercise, protects against CI-AKI by inhibiting the cGAS-STING inflammatory pathway. METHODS AND RESULTS We explored the relationship between serum irisin levels and CI-AKI incidence in patients administered the contrast media iohexol. Notably, lower serum irisin levels were strongly associated with an increased incidence of CI-AKI following contrast media administration. To establish a causal link between serum irisin levels and CI-AKI, we utilised a mouse model that simulates exercise by overexpressing muscle-specific PGC-1α. This approach showed a significant reduction in tubular injury and mitochondrial dysfunction induced by iohexol via cGAS/STING suppression, thereby diminishing inflammation. Mechanistically, irisin was found to inhibit the activation of cGAS/STING, preventing double stranded DNA (dsDNA) leakage and reducing inflammation in tubular epithelial cells (TECs). Pharmacological inhibition of STING further corroborated these observations. Moreover, we identified integrin complex αV/β5 as the irisin receptor on TECs, which is essential for irisin-mediated suppression of cGAS-STING signalling and resolution of inflammation. CONCLUSIONS Our data position irisin as a crucial factor in muscle‒kidney crosstalk, inhibiting cGAS-STING signalling and preventing dsDNA leakage via integrin αV/β5 in TECs, thus mitigating tubular injury and inflammation. These data underscore the potential of irisin as both a predictive biomarker for CI-AKI and a promising candidate for preventative strategies against CI-AKI. HIGHLIGHTS Irisin mediated muscle-kidney crosstalk mitigated tubular injury and inflammation. Irisin inhibited the cGAS-STING signalling activation via integrin αV/β5 in tubular epithelial cells. Irisin was a predictive biomarker and a promising candidate for CI-AKI.
Collapse
Affiliation(s)
- Long Peng
- Division of Cardiovascular Medicine, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Suhua Li
- Division of Cardiovascular Medicine, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Qiang Huang
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yuxiang Sun
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Juan Sun
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Ting Luo
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yanlin Wang
- Division of Nephrology, Department of MedicineUniversity of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Zhaoyong Hu
- Division of Nephrology, Department of MedicineBaylor College of MedicineHoustonTexasUSA
| | - Weiyan Lai
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Hui Peng
- Division of Nephrology, Department of MedicineThe Third Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
5
|
Dani C, Tarchi L, Rossi E, Cassioli E, Rotella F, Fanelli A, Salvadori B, Mannino R, Rossolini GM, Lucarelli S, Ricca V, Castellini G. Inflammatory biomarkers and childhood maltreatment: A cluster analysis in patients with eating disorders. Psychoneuroendocrinology 2025; 174:107405. [PMID: 39978212 DOI: 10.1016/j.psyneuen.2025.107405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/14/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Eating Disorders (EDs) are severe psychiatric disorders, with growing evidence pointing towards the role of childhood maltreatment (CM) influencing their onset, severity, and response to treatment. Preliminary evidence showed that CM could be associated with an elevation of inflammatory biomarkers across the different EDs. The objective of the study was to elucidate the interplay between CM, ED-specific psychopathology, and inflammatory biomarkers. The study involved 198 female participants, comprising 70 patients with anorexia nervosa (AN), 56 patients with bulimia nervosa (BN), and 72 healthy controls (HCs). K-means clustering was used to assess the hypothesis that latent clusters could be described between patients affected by EDs based on serum levels of inflammatory biomarkers alone (CRP, IL-6, suPAR). Additionally, the analysis included a comparison between patients with and without history of childhood maltreatment. Patients with AN exhibited significantly higher suPAR levels than HCs, regardless of the severity of psychopathology. A direct association between CM and elevated levels of inflammatory biomarkers, particularly CRP, IL-6, and suPAR were found. Cluster analysis identified two distinct populations among patients with EDs, with the group showing elevated inflammatory biomarkers likely to report more severe CM. Even though preliminary, the results of the present study support the existence of a biologically grounded "maltreated eco-phenotype" in EDs. The present study also reports results on CRP, IL-6 and suPAR, in patients with EDs. These findings might suggest future potential tailored treatments and interventions designed to target specific subgroups of patients, and potentially improving treatment efficacy.
Collapse
Affiliation(s)
- Cristiano Dani
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Livio Tarchi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Eleonora Rossi
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Emanuele Cassioli
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesco Rotella
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | | | - Roberta Mannino
- General Laboratory, Careggi University Hospital, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Valdo Ricca
- Department of Health Sciences, University of Florence, Florence, Italy
| | | |
Collapse
|
6
|
Saliba A, Debnath S, Tamayo I, Lee HJ, Ragi N, Das F, Montellano R, Tumova J, Maddox M, Trevino E, Singh P, Fastenau C, Maity S, Zhang G, Hejazi L, Venkatachalam MA, O’Connor JC, Fongang B, Hopp SC, Bieniek KF, Lechleiter JD, Sharma K. Quinolinic acid potentially links kidney injury to brain toxicity. JCI Insight 2025; 10:e180229. [PMID: 39946208 PMCID: PMC11949017 DOI: 10.1172/jci.insight.180229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/12/2025] [Indexed: 02/19/2025] Open
Abstract
Kidney dysfunction often leads to neurological impairment, yet the complex kidney-brain relationship remains elusive. We employed spatial and bulk metabolomics to investigate a mouse model of rapid kidney failure induced by mouse double minute 2 (Mdm2) conditional deletion in the kidney tubules to interrogate kidney and brain metabolism. Pathway enrichment analysis of a focused plasma metabolomics panel pinpointed tryptophan metabolism as the most altered pathway with kidney failure. Spatial metabolomics showed toxic tryptophan metabolites in the kidneys and brains, revealing a connection between advanced kidney disease and accelerated kynurenine degradation. In particular, the excitotoxic metabolite quinolinic acid was localized in ependymal cells in the setting of kidney failure. These findings were associated with brain inflammation and cell death. Separate mouse models of ischemia-induced acute kidney injury and adenine-induced chronic kidney disease also exhibited systemic inflammation and accumulating toxic tryptophan metabolites. Patients with advanced chronic kidney disease (stage 3b-4 and stage 5) similarly demonstrated elevated plasma kynurenine metabolites, and quinolinic acid was uniquely correlated with fatigue and reduced quality of life. Overall, our study identifies the kynurenine pathway as a bridge between kidney decline, systemic inflammation, and brain toxicity, offering potential avenues for diagnosis and treatment of neurological issues in kidney disease.
Collapse
Affiliation(s)
- Afaf Saliba
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Subrata Debnath
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Ian Tamayo
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Hak Joo Lee
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Nagarjunachary Ragi
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Falguni Das
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Richard Montellano
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jana Tumova
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | | | - Esmeralda Trevino
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Pragya Singh
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Caitlyn Fastenau
- Department of Pharmacology
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, and
| | - Soumya Maity
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Guanshi Zhang
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Leila Hejazi
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Manjeri A. Venkatachalam
- Center for Precision Medicine and
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jason C. O’Connor
- Department of Pharmacology
- South Texas Veterans Health Care System, Audie L. Murphy VA Hospital, San Antonio, Texas, USA
| | - Bernard Fongang
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, and
- Department of Biochemistry and Structural Biology
- Department of Population Health Sciences, and
| | - Sarah C. Hopp
- Department of Pharmacology
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, and
| | - Kevin F. Bieniek
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, and
| | - James D. Lechleiter
- Center for Precision Medicine and
- Department of Cell Systems and Anatomy, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Kumar Sharma
- Center for Precision Medicine and
- Division of Nephrology, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
7
|
Mwaba C, Munsaka S, Mwakazanga D, Rutagwerae D, Ngalamika O, Mwanza S, McCulloch M, Mpabalwani E. Clinical, immune and genetic risk factors of malaria-associated acute kidney injury in Zambian children: A study protocol. PLoS One 2025; 20:e0316205. [PMID: 39913368 PMCID: PMC11801570 DOI: 10.1371/journal.pone.0316205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/06/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) affects nearly half of children with severe malaria and increases the risk of adverse outcomes such as death and poor cognitive function. The pathogenesis and predictors of malaria-associated acute kidney injury (MAKI) are not fully described. This study aims to determine the clinical, immune, and genetic correlates of risk to AKI in Zambian children admitted with malaria. In addition, we intend to assess a modified renal angina index (mRAI), kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and soluble urokinase receptor (suPAR), when done on the first day of admission, for the ability to predict AKI two days later (day 3) in children admitted with malaria. METHODS This is an unmatched case-control study with a nested prospective observational study. A case-to-control ratio of 1:1 is used and 380 children with malaria and aged less than 16 years are being recruited from two hospitals in Zambia. Eligible children are recruited after obtaining written informed consent. Recruitment occurs during the malaria season and began on 6th March 2024 and will continue until July 2025. AKI is defined using the 2012 KIDGO AKI creatinine criteria, and cases are defined as children admitted with malaria who develop AKI within 72 hours of admission, while controls are children admitted with malaria but with no AKI. Serum creatinine is collected on Day 1 within 24 hours of admission, on Day 3 and then again on discharge or day 7, whichever comes sooner. Baseline biomarker concentrations will be determined using the Luminex multiplex Elisa system or high-sensitivity ELISA. SPSS version 29 will be used for data analysis. Descriptive statistics and inferential statistical tests will be run as appropriate. A p ≤ 0.05 will be considered as significant. The sensitivity, specificity, and estimates of the area under the curve (AUC) for the renal angina score will be determined.
Collapse
Affiliation(s)
- Chisambo Mwaba
- Department of Paediatrics and Child Health, School of Medicine, University of Zambia, Lusaka, Zambia
- Department of Paediatrics, University Teaching Hospitals-Children’s Hospital, Lusaka, Zambia
| | - Sody Munsaka
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - David Mwakazanga
- Public Health Department, Epidemiology and Statistics Unit, Tropical Diseases Research Centre, Ndola, Zambia
| | - David Rutagwerae
- Kaposi’s Sarcoma Molecular Laboratory, Paediatric Centre of Excellence, University Teaching Hospitals-Children’s Hospital, Lusaka, Zambia
| | - Owen Ngalamika
- Dermatology and Venereology Division, Department of Internal Medicine, University Teaching Hospital, University of Zambia School of Medicine, Lusaka, Zambia
| | - Suzanna Mwanza
- Department of Paediatrics and Child Health, Chipata Central Hospital, Chipata, Zambia
| | - Mignon McCulloch
- Division of Paediatric Nephrology, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
| | - Evans Mpabalwani
- Department of Paediatrics and Child Health, School of Medicine, University of Zambia, Lusaka, Zambia
- Department of Paediatrics, University Teaching Hospitals-Children’s Hospital, Lusaka, Zambia
| |
Collapse
|
8
|
Ang L, Gunaratnam S, Huang Y, Dillon BR, Martin CL, Burant A, Reiss J, Blakely P, Vasbinder A, Zhao L, Mizokami-Stout K, Tang Y, Feldman EL, Doria A, Spino C, Banerjee M, Hayek SS, Pop-Busui R. Inflammatory Markers and Measures of Cardiovascular Autonomic Neuropathy in Type 1 Diabetes. J Am Heart Assoc 2025; 14:e036787. [PMID: 39727210 DOI: 10.1161/jaha.124.036787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/28/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Cardiovascular autonomic neuropathy (CAN) and inflammation predict more severe outcomes in type 1 diabetes (T1D). However, the link between CAN and inflammation in T1D remains unclear. We examined associations between CAN measures and inflammatory biomarkers in individuals with T1D. METHODS AND RESULTS In a cross-sectional study, we measured cardiovascular autonomic reflex tests and heart rate variability (established CAN measures) and a panel of 39 inflammatory biomarkers, including soluble urokinase plasminogen activator receptor (suPAR), in T1D participants of the TINSAL-T1DN (Targeting Inflammation with Salsalate in Individuals with T1D Neuropathy) trial (n=57, discovery), and the PERL (Preventing Early Renal Loss in Diabetes) trial (n=468, validation). Amongst 39 inflammatory biomarkers measured in TINSAL-T1DN, suPAR levels had the strongest negative correlations with CAN measures: expiration/inspiration (r=-0.48), Valsalva (r=-0.28), 30:15 (r=-0.37), SD of the normal RR interval (r=-0.37), and root mean square of differences of successive RR intervals (r=-0.31) (all P<0.05). Findings were validated in PERL. In unadjusted analyses, median suPAR levels significantly differed between the lowest and highest SD of the normal RR interval tertiles (3.79 versus 3.12 ng/mL, P<0.001) and root mean square of differences of successive RR intervals (3.76 versus 3.17 ng/mL, P<0.001). After adjusting for covariates (age, sex, hemoglobin A1c, and estimated glomerular filtration rate), median suPAR values remained significantly elevated in the lowest tertiles of SD of the normal RR interval (P=0.004) and root mean square of differences of successive RR intervals (P=0.006). CONCLUSIONS Amongst several inflammatory biomarkers, suPAR, an immune-mediated glycoprotein, has a singular association with CAN measures. The potential of targeting suPAR as a disease-modifying approach for CAN in T1D warrants further exploration. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifiers: NCT02936843, NCT02017171.
Collapse
Affiliation(s)
- Lynn Ang
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes University of Michigan Ann Arbor MI USA
| | - Sejal Gunaratnam
- Life Science Informatics University of Michigan Ann Arbor MI USA
| | - Yiyuan Huang
- Department of Biostatistics University of Michigan Ann Arbor MI USA
| | - Brendan R Dillon
- Department of Medicine NYU Grossman School of Medicine New York NY USA
| | - Catherine L Martin
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes University of Michigan Ann Arbor MI USA
| | - Aaron Burant
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes University of Michigan Ann Arbor MI USA
| | - Jacob Reiss
- Quality Department University of Michigan Ann Arbor MI USA
| | - Pennelope Blakely
- Department of Internal Medicine, Division of Cardiology University of Texas Medical Branch Galveston TX USA
| | - Alexi Vasbinder
- Biobehavioral Nursing and Health Informatics University of Washington School of Nursing Seattle WA USA
| | - Lili Zhao
- Corewell Health William Beaumont University Hospital Royal Oak MI USA
| | - Kara Mizokami-Stout
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes University of Michigan Ann Arbor MI USA
- Ann Arbor Veteran Affairs Hospital Ann Arbor MI USA
| | - Yaling Tang
- Department of Epidemiology Joslin Diabetes Center Boston MA USA
| | - Eva L Feldman
- Department of Neurology University of Michigan Ann Arbor MI USA
| | | | - Cathie Spino
- Department of Biostatistics University of Michigan Ann Arbor MI USA
| | - Mousumi Banerjee
- Department of Biostatistics University of Michigan Ann Arbor MI USA
| | - Salim S Hayek
- Department of Internal Medicine, Division of Cardiology University of Texas Medical Branch Galveston TX USA
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes University of Michigan Ann Arbor MI USA
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Clinical Nutrition Oregon Health and Science University Portland OR USA
| |
Collapse
|
9
|
Yadalam AK, Gold ME, Patel KJ, Liu C, Razavi AC, Jain V, Vatsa N, Gold D, Owais M, Haroun N, Sun YV, Quyyumi AA. Proteomics-Based Soluble Urokinase Plasminogen Activator Receptor Levels Are Associated With Incident Heart Failure Risk. JACC. ADVANCES 2025; 4:101442. [PMID: 39737138 PMCID: PMC11683231 DOI: 10.1016/j.jacadv.2024.101442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/28/2024] [Accepted: 11/06/2024] [Indexed: 01/01/2025]
Abstract
Background Higher soluble urokinase plasminogen activator receptor (suPAR) levels are associated with adverse outcomes in chronic heart failure (HF). Objectives The authors assessed the association between proteomics-based suPAR levels and incident HF risk in the general population. Methods In 40,418 UK Biobank participants without HF or coronary artery disease at enrollment, the association between Olink-based suPAR levels measured as relative protein expression levels and incident all-cause, ischemic, and nonischemic HF was analyzed by competing-risk regression, while accounting for all-cause death as a competing risk. The additional variability in incident HF risk attributable to suPAR levels beyond demographics, traditional risk factors, N-terminal pro B-type natriuretic peptide (NT-proBNP), and C-reactive protein (CRP) levels was assessed with nested Cox modeling and likelihood ratio testing. Results The mean age was 56 years; 45% were male, and 94% were White. During a median follow-up of 13.7 (IQR: 1.5) years, 1,428 (3.5%) incident HF events occurred. Proteomics-based suPAR levels (per 1-SD) were independently associated with incident HF (subdistribution HR (sHR): 1.37, 95% CI: 1.29-1.46), ischemic HF (sHR: 1.40, 95% CI: 1.28-1.54), and nonischemic HF (sHR: 1.32, 95% CI: 1.21-1.44) risk, after adjustment for demographics, traditional cardiovascular risk factors, NT-proBNP, and CRP levels. The addition of suPAR levels to a base risk factor model significantly improved the explained variability of incident HF risk (R 2 = 0.76 vs 0.73, P < 0.001). Conclusions Independent of demographics, traditional risk factors, NT-proBNP, and CRP levels, proteomics-based suPAR levels were significantly associated with incident all-cause, ischemic, and nonischemic HF risk. Proteomics-based measurement of suPAR levels may underestimate the effect size of this relationship.
Collapse
Affiliation(s)
- Adithya K. Yadalam
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Matthew E. Gold
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Krishan J. Patel
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Chang Liu
- Emory University, Rollins School of Public Health, Department of Epidemiology, Atlanta, Georgia, USA
| | - Alexander C. Razavi
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Vardhmaan Jain
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Nishant Vatsa
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Daniel Gold
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Muhammad Owais
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Nisreen Haroun
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| | - Yan V. Sun
- Emory University, Rollins School of Public Health, Department of Epidemiology, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Healthcare System, Decatur, Georgia, USA
| | - Arshed A. Quyyumi
- Emory University School of Medicine, Division of Cardiology, Department of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Kyriazopoulou E, Akinosoglou K, Florou E, Kouriannidi E, Bogosian A, Tsachouridou O, Syrigos KN, Gatselis N, Milionis H, Papanikolaou IC, Sympardi S, Dafni M, Alevizou A, Amvrazi AV, Alexandrou E, Archontoulis K, Argyraki K, Alexiou Z, Georgiou Y, Gkogka D, Kyrailidi F, Kalyva V, Nikolopoulou T, Ioannou S, Bakakos P, Karathanassiou G, Koklanos K, Miletis DN, Tili AM, Vakkas L, Vila I, Panagopoulos P, Samarkos M, Chrysos G, Dalekos GN, Poulakou G, Metallidis S, Giamarellos-Bourboulis EJ. Anakinra efficacy in COVID-19 pneumonia guided by soluble urokinase plasminogen activator receptor: Association with the inflammatory burden of the host. Int J Antimicrob Agents 2025; 65:107405. [PMID: 39647797 DOI: 10.1016/j.ijantimicag.2024.107405] [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: 04/10/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Anakinra was approved by the European Medicines Agency and received Emergency Use Authorization by the United States Food and Drug Administration for patients with COVID-19 pneumonia at risk for severe respiratory failure (SRF) with blood levels of soluble urokinase plasminogen activator receptor (suPAR) ≥ 6 ng/mL. We report the final results of the phase II open-label single-arm SAVE trial in a large population. METHODS Patients with COVID-19 pneumonia and suPAR levels ≥ 6 ng/mL received subcutaneous anakinra 100 mg once daily for 10 days. The primary outcome was the incidence of SRF by day 14. Secondary outcomes were 30-day mortality, incidence of SRF according to time delay for start of treatment, safety, and associations with the inflammatory burden of the host. RESULTS From March 2020 to March 2022, a total of 992 patients were enrolled. The incidence of SRF was 18.8%, similar to the results of the phase III pivotal SAVE-MOREtrial. The overall 30-day mortality was 9.5%. Participants were divided into 4 subgroups according to time delay between symptoms onset and start of anakinra. The incidence of SRF was similar for all subgroups. Serious adverse events were reported in 15.4%; only 3 were possibly related to anakinra. The most common adverse event was increased liver function tests. A post hoc comparison with the pivotal phase III trial showed similar anakinra outcomes among patient subgroups by levels of inflammatory mediators and D-dimers. CONCLUSIONS Results support the efficacy of anakinra as being similar to that of the pivotal registrational trial for COVID-19 pneumonia. The lack of a comparator group is a limitation. TRIAL REGISTRATION ClinicalTrials.gov, NCT04357366.
Collapse
Affiliation(s)
- Evdoxia Kyriazopoulou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Eleni Florou
- Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Elli Kouriannidi
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Olga Tsachouridou
- First Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos N Syrigos
- Third Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Nikolaos Gatselis
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Haralampos Milionis
- First Department of Internal Medicine, University of Ioannina, Medical School, Ioannina, Greece
| | - Ilias C Papanikolaou
- Department of Pulmonary Medicine, General Hospital of Corfu "Agia Eirini", Greece
| | - Styliani Sympardi
- First Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Maria Dafni
- First Department of Internal Medicine, Korgialeneion-Benakeion General Hospital, Athens, Greece
| | - Antonia Alevizou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Alexia-Vasiliki Amvrazi
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Errika Alexandrou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Kyprianos Archontoulis
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Katerina Argyraki
- Department of Internal Medicine, Sotiria Athens Hospital of Chest Diseases, Athens, Greece
| | - Zoi Alexiou
- Second Department of Internal Medicine, Thriasio General Hospital of Eleusis, Athens, Greece
| | - Yakinthi Georgiou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dimitra Gkogka
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Foteini Kyrailidi
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Vassiliki Kalyva
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | | | - Sofia Ioannou
- Department of Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Bakakos
- First Department of Pulmonary Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Karathanassiou
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Kyriakos Koklanos
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dionysios-Nikolaos Miletis
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Anna-Maria Tili
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Lampros Vakkas
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioanna Vila
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Periklis Panagopoulos
- Second Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Michael Samarkos
- First Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George Chrysos
- Second Department of Internal Medicine, Tzaneio General Hospital of Piraeus, Athens, Greece
| | - George N Dalekos
- Department of Medicine and Research Laboratory of Internal Medicine, National Expertise Center of Greece in Autoimmune Liver Diseases, Full Member of the European Reference Network on Hepatological Diseases (ERN RARE-LIVER), General University Hospital of Larissa, Larissa, Greece
| | - Garyphallia Poulakou
- Third Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Symeon Metallidis
- First Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelos J Giamarellos-Bourboulis
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece.
| |
Collapse
|
11
|
Altintas MM, Agarwal S, Sudhini Y, Zhu K, Wei C, Reiser J. Pathogenesis of Focal Segmental Glomerulosclerosis and Related Disorders. ANNUAL REVIEW OF PATHOLOGY 2025; 20:329-353. [PMID: 39854184 PMCID: PMC11875227 DOI: 10.1146/annurev-pathol-051220-092001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Focal segmental glomerulosclerosis (FSGS) is the morphologic manifestation of a spectrum of kidney diseases that primarily impact podocytes, cells that create the filtration barrier of the glomerulus. As its name implies, only parts of the kidney and glomeruli are affected, and only a portion of the affected glomerulus may be sclerosed. Although the diagnosis is based primarily on microscopic features, patient stratification relies on clinical data such as proteinuria and etiological criteria. FSGS affects both children and adults and has an elevated risk of progression to end-stage renal disease. The prevalence of FSGS is rising among various populations, and the efficacy of various therapies is limited. Therefore, understanding the pathophysiology of FSGS and developing targeted therapies to address the complex needs of FSGS patients are topics of great interest that are currently being studied across various clinical trials. We discuss the etiology of FSGS, describe the major contributing pathophysiological pathways, and outline emerging therapeutic strategies along with their pitfalls.
Collapse
Affiliation(s)
- Mehmet M Altintas
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA;
| | | | - Yashwanth Sudhini
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Ke Zhu
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA;
| | - Changli Wei
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA;
| | - Jochen Reiser
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA;
| |
Collapse
|
12
|
Zhang W, Gu Y, Zhou J, Wang J, Zhao X, Deng X, Li H, Yan L, Jiao X, Shao F. Clinical value of soluble urokinase-type plasminogen activator receptor in predicting sepsis-associated acute kidney injury. Ren Fail 2024; 46:2307959. [PMID: 38289005 PMCID: PMC10829810 DOI: 10.1080/0886022x.2024.2307959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Sepsis-associated acute kidney injury (S-AKI) is a critical illness and is often associated with high morbidity and mortality rates. The soluble urokinase-type plasminogen activator receptor (suPAR) is an important immune mediator and is involved in kidney injury. However, its diagnostic value in S-AKI patients remains unclear. Therefore, we assessed the early predictive value of suPAR for S-AKI patients. METHODS We prospectively enrolled adult patients, immediately after fulfilling the sepsis-3 criteria. Plasma suPAR levels at 0-, 12-, 24-, and 48-h post-sepsis diagnosis were measured. S-AKI development was the primary outcome. S-AKI risk factors were analyzed using logistic regression, and the value of plasma suPAR for early S-AKI diagnosis was assessed using receiver operating characteristic (ROC) curves. RESULTS Of 179 sepsis patients, 63 (35.2%) developed AKI during hospitalization. At 12-, 24-, and 48-h post-sepsis diagnosis, plasma suPAR levels were significantly higher in patients with S-AKI than in patients without S-AKI (p < 0.05). The plasma suPAR had the highest area under the ROC curve of 0.700 (95% confidence interval (CI), 0.621-0.779) at 24-h post-sepsis diagnosis, at which the best discrimination ability for S-AKI was achieved with suPAR of ≥6.31 ng/mL (sensitivity 61.9% and specificity 71.6%). Logistic regression analysis showed that suPAR at 24-h post-sepsis diagnosis remained an independent S-AKI risk factor after adjusting for mechanical ventilation, blood urea nitrogen, and pH. CONCLUSIONS The findings suggest that plasma suPAR may be a potential biomarker for early S-AKI diagnosis.
Collapse
Affiliation(s)
- Wenwen Zhang
- Department of Nephrology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yue Gu
- Department of Nephrology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Key Laboratory for Kidney Disease and Immunology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Nephrology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Jing Zhou
- Department of Nephrology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Key Laboratory for Kidney Disease and Immunology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Juntao Wang
- Department of Nephrology, The First People’s Hospital of Shangqiu, Shangqiu, China
| | - Xiaoru Zhao
- Department of Nephrology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiaoyu Deng
- Department of Nephrology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Han Li
- Department of Nephrology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lei Yan
- Department of Nephrology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Key Laboratory for Kidney Disease and Immunology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiaojing Jiao
- Department of Nephrology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Key Laboratory for Kidney Disease and Immunology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Fengmin Shao
- Department of Nephrology, Henan Provincial Clinical Research Center for Kidney Disease, Henan Key Laboratory for Kidney Disease and Immunology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
13
|
Noel S, Kapoor R, Rabb H. New approaches to acute kidney injury. Clin Kidney J 2024; 17:65-81. [PMID: 39583139 PMCID: PMC11581771 DOI: 10.1093/ckj/sfae265] [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: 04/04/2024] [Indexed: 11/26/2024] Open
Abstract
Acute kidney injury (AKI) is a common and serious clinical syndrome that involves complex interplay between different cellular, molecular, metabolic and immunologic mechanisms. Elucidating these pathophysiologic mechanisms is crucial to identify novel biomarkers and therapies. Recent innovative methodologies and the advancement of existing technologies has accelerated our understanding of AKI and led to unexpected new therapeutic candidates. The aim of this review is to introduce and update the reader about recent developments applying novel technologies in omics, imaging, nanomedicine and artificial intelligence to AKI research, plus to provide examples where this can be translated to improve patient care.
Collapse
Affiliation(s)
- Sanjeev Noel
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Radhika Kapoor
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
14
|
Xu J, Chen J, Li M, Jiang Z, Fang F, Hu J, Zhou Y, Li H, Bai Z, Li X, Lu G, Li Y. Utility of plasma suPAR to identify AKI and sepsis associated AKI in critically ill children. iScience 2024; 27:111247. [PMID: 39569376 PMCID: PMC11576388 DOI: 10.1016/j.isci.2024.111247] [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: 07/22/2024] [Revised: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Current biomarkers for sepsis-associated acute kidney injury (SA-AKI) lack specificity. The role of soluble urokinase plasminogen activator receptor (suPAR) in discriminating AKI and SA-AKI in children remains elusive. This prospective multicenter study was conducted in critically ill children cohorts using a derivation-validation design, and plasma samples were collected within first 24 h after admission. Plasma suPAR was independently associated with AKI, SA-AKI, and PICU mortality, even after adjustment for confounding variables. This multiclass classification model had the micro-average AUC of 0.89 with specificity of 97.6% for discriminating non-septic AKI, and specificity of 99.0% for discriminating SA-AKI, based on the cut-off values of 1.5 and 2.3-fold baseline in serum creatinine (SCr) and 4.5 and 11.2 ng/mL in plasma suPAR. The multiclass classification model provides the cutoffs for plasma suPAR and SCr and specifically discriminates critically ill children at high risk of non-septic AKI and SA-AKI, which can facilitate clinical utility.
Collapse
Affiliation(s)
- Jing Xu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Jiao Chen
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
- Pediatric Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, P.R. China
| | - Min Li
- Pediatric Intensive Care Unit, Anhui Provincial Children's Hospital, Hefei, Anhui Province, P.R. China
| | - Zhen Jiang
- Pediatric Intensive Care Unit, Xuzhou Children's Hospital, Xuzhou, Jiangsu Province, P.R. China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Junlong Hu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Yueying Zhou
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Huiwen Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Zhenjiang Bai
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Xiaozhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| | - Guoping Lu
- Pediatric Intensive Care Unit, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - Yanhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, P.R. China
| |
Collapse
|
15
|
Rasmussen SB, Boyko Y, Ranucci M, de Somer F, Ravn HB. Cardiac surgery-Associated acute kidney injury - A narrative review. Perfusion 2024; 39:1516-1530. [PMID: 37905794 DOI: 10.1177/02676591231211503] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cardiac Surgery-Associated Acute Kidney Injury (CSA-AKI) is a serious complication seen in approximately 20-30% of cardiac surgery patients. The underlying pathophysiology is complex, often involving both patient- and procedure related risk factors. In contrast to AKI occurring after other types of major surgery, the use of cardiopulmonary bypass comprises both additional advantages and challenges, including non-pulsatile flow, targeted blood flow and pressure as well as the ability to manipulate central venous pressure (congestion). With an increasing focus on the impact of CSA-AKI on both short and long-term mortality, early identification and management of high-risk patients for CSA-AKI has evolved. The present narrative review gives an up-to-date summary on definition, diagnosis, underlying pathophysiology, monitoring and implications of CSA-AKI, including potential preventive interventions. The review will provide the reader with an in-depth understanding of how to identify, support and provide a more personalized and tailored perioperative management to avoid development of CSA-AKI.
Collapse
Affiliation(s)
- Sebastian Buhl Rasmussen
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Yuliya Boyko
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
| | - Marco Ranucci
- Department of Cardiovascular Anaesthesiology and Intensive Care, IRCCS Policlinico San Donato, Milan, Italy
| | | | - Hanne Berg Ravn
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
16
|
Mascle O, Dupuis C, Brailova M, Bonnet B, Mirand A, De Beauchene RC, Philipponnet C, Adda M, Calvet L, Cassagnes L, Henquell C, Sapin V, Evrard B, Souweine B. Clustering based on renal and inflammatory admission parameters in critically ill patients admitted to the ICU. PLoS One 2024; 19:e0307938. [PMID: 39485788 PMCID: PMC11530013 DOI: 10.1371/journal.pone.0307938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/15/2024] [Indexed: 11/03/2024] Open
Abstract
INTRODUCTION The COVID-19 pandemic has been associated with significant variability in acute kidney injury (AKI) incidence, leading to concerns regarding patient heterogeneity. The study's primary objective was a cluster analysis, to identify homogeneous subgroups of patients (clusters) using baseline characteristics, including inflammatory biomarkers. The secondary objectives were the comparisons of MAKE-90 and mortality between the different clusters at three months. METHODS This retrospective single-center study was conducted in the Medical Intensive Care Unit of the University Hospital of Clermont-Ferrand, France. Baseline data, clinical and biological characteristics on ICU admission, and outcomes at day 90 were recorded. The primary outcome was the risk of major adverse kidney events at 90 days (MAKE-90). Clusters were determined using hierarchical clustering on principal components approach based on admission characteristics, biomarkers and serum values of immune dysfunction and kidney function. RESULTS It included consecutive adult patients admitted between March 20, 2020 and February 28, 2021 for severe COVID-19. A total of 149 patients were included in the study. Three clusters were identified of which two were fully described (cluster 3 comprising 2 patients). Cluster 1 comprised 122 patients with fewer organ dysfunctions, moderate immune dysfunction, and was associated with reduced mortality and a lower incidence of MAKE-90. Cluster 2 comprised 25 patients with greater disease severity, immune dysfunction, higher levels of suPAR and L-FABP/U Creat, and greater organ support requirement, incidence of AKI, day-90 mortality and MAKE-90. CONCLUSIONS This study identified two clusters of severe COVID-19 patients with distinct biological characteristics and renal event risks. Such clusters may help facilitate the identification of targeted populations for future clinical trials. Also, it may help to understand the significant variability in AKI incidence observed in COVID-19 patients.
Collapse
Affiliation(s)
- Olivier Mascle
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Claire Dupuis
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Université Clermont Auvergne, Clermont Ferrand, France
| | - Marina Brailova
- CHU de Clermont-Ferrand, Service de Biochimie Médicale, Clermont-Ferrand, France
| | - Benjamin Bonnet
- CHU de Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Audrey Mirand
- CHU de Clermont-Ferrand, 3IHP, Service de Virologie, Clermont-Ferrand, France
- UMR CNRS 6023 LMGE, Université Clermont Auvergne, Clermont-Ferrand, France
| | | | | | - Mireille Adda
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Laure Calvet
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Lucie Cassagnes
- CHU de Clermont-Ferrand, Service de Radiologie, Clermont-Ferrand, France
| | - Cécile Henquell
- CHU de Clermont-Ferrand, 3IHP, Service de Virologie, Clermont-Ferrand, France
- UMR CNRS 6023 LMGE, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Vincent Sapin
- CHU de Clermont-Ferrand, Service de Biochimie Médicale, Clermont-Ferrand, France
| | - Bertrand Evrard
- CHU de Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Bertrand Souweine
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- UMR CNRS 6023 LMGE, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
17
|
Alkhaleq HA, Hacker I, Karram T, Hamoud S, Kabala A, Abassi Z. Potential Nephroprotective Effect of uPA against Ischemia/Reperfusion-Induced Acute Kidney Injury in αMUPA Mice and HEK-293 Cells. Biomedicines 2024; 12:2323. [PMID: 39457635 PMCID: PMC11505258 DOI: 10.3390/biomedicines12102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The incidence of acute kidney injury (AKI) has been steadily increasing. Despite its high prevalence, there is no pathogenetically rational therapy for AKI. This deficiency stems from the poor understanding of the pathogenesis of AKI. Renal ischemia/hypoxia is one of the leading causes of clinical AKI. This study investigates whether αMUPA mice, overexpressing the urokinase plasminogen activator (uPA) gene are protected against ischemic AKI, thus unraveling a potential renal damage treatment target. Methods: We utilized an in vivo model of I/R-induced AKI in αMUPA mice and in vitro experiments of uPA-treated HEK-293 cells. We evaluated renal injury markers, histological changes, mRNA expression of inflammatory, apoptotic, and autophagy markers, as compared with wild-type animals. Results: the αMUPA mice exhibited less renal injury post-AKI, as was evident by lower SCr, BUN, and renal NGAL and KIM-1 along attenuated adverse histological alterations. Notably, the αMUPA mice exhibited decreased levels pro-inflammatory, fibrotic, apoptotic, and autophagy markers like TGF-β, IL-6, STAT3, IKB, MAPK, Caspase-3, and LC3. By contrast, ACE-2, p-eNOS, and PGC1α were higher in the kidneys of the αMUPA mice. In vitro results of the uPA-treated HEK-293 cells mirrored the in vivo findings. Conclusions: These results indicate that uPA modulates key pathways involved in AKI, offering potential therapeutic targets for mitigating renal damage.
Collapse
Affiliation(s)
- Heba Abd Alkhaleq
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
| | - Israel Hacker
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
| | - Tony Karram
- Department of Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Shadi Hamoud
- Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Aviva Kabala
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel; (H.A.A.); (I.H.); (A.K.)
- Laboratory Medicine, Rambam Health Care Campus, Haifa 3109601, Israel
| |
Collapse
|
18
|
Chen MA, Chen E, Gallivan SU, Brody EJ, Passarelli V, Miller GE. Socioeconomic Disadvantage, Neighborhood Belonging, and Inflammation Among Adolescents. Psychosom Med 2024; 86:660-669. [PMID: 39109943 PMCID: PMC11444907 DOI: 10.1097/psy.0000000000001332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
OBJECTIVE Childhood socioeconomic disadvantage is associated with a host of adverse health outcomes across the lifespan. However, there is increasing interest in identifying factors that may promote resilience to disadvantage's effects on health. One promising candidate in this regard is a sense of neighborhood belonging, which could offset health risks by providing a sense of connection to others, as well as a sense of belonging to a community larger than oneself. METHODS In a sample of 245 adolescents (age: mean [standard deviation] = 15.98 [0.54] years; sex: 64.1% female; race: 41.6% White, 37.6% Black/African American, 9.8% Other; ethnicity: 68.6% non-Hispanic), we examined neighborhood belonging as a moderator of the relationship between socioeconomic disadvantage (measured on a 0- to 5-point scale, mean [standard deviation] = 1.21 [1.36]) and low-grade inflammation (measured via a composite of circulating inflammatory biomarkers including IL-6, IL-8, IL-10, TNF-α, CRP, and suPAR). Covariates included age, sex, race/ethnicity, and pubertal status. RESULTS Neighborhood belonging buffered the relationship between socioeconomic disadvantage and low-grade inflammation, a key mechanistic pathway to multiple chronic diseases. Specifically, there was a positive relationship between socioeconomic disadvantage and low-grade inflammation among individuals with low neighborhood belonging, but not among individuals with high neighborhood belonging. CONCLUSIONS These findings suggest that neighborhood belonging is one type of social connection factor that can mitigate the relationship between socioeconomic disadvantage and low-grade inflammation in youth.
Collapse
Affiliation(s)
- Michelle A. Chen
- Institute of Policy Research, Northwestern University, 1801 Maple Ave, Evanston, IL, 60201
| | - Edith Chen
- Institute of Policy Research, Northwestern University, 1801 Maple Ave, Evanston, IL, 60201
| | - Shanti U. Gallivan
- Institute of Policy Research, Northwestern University, 1801 Maple Ave, Evanston, IL, 60201
| | - Elizabeth J. Brody
- Institute of Policy Research, Northwestern University, 1801 Maple Ave, Evanston, IL, 60201
- Saint Louis University School of Medicine, 1402 S Grand Blvd, St. Louis, MO 63104
| | - Veronica Passarelli
- Institute of Policy Research, Northwestern University, 1801 Maple Ave, Evanston, IL, 60201
| | - Gregory E. Miller
- Institute of Policy Research, Northwestern University, 1801 Maple Ave, Evanston, IL, 60201
| |
Collapse
|
19
|
Zhu L, Cai J, Fang J, Ran L, Chang H, Zhang H, Zeng J, Yang Q, Fu C, Li Q, Pan Q, Zhao H. Nomogram Model for Cardiac Surgery-Associated Acute Kidney Injury Based on Clinical Characteristics Combined with Plasma suPAR. Int J Gen Med 2024; 17:3181-3192. [PMID: 39049830 PMCID: PMC11268850 DOI: 10.2147/ijgm.s464904] [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: 02/22/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Objective Analyze risk factors for cardiac surgery-associated acute kidney injury (CSA-AKI) in adults and establish a nomogram model for CSA-AKI based on plasma soluble urokinase-type plasminogen activator receptor (suPAR) and clinical characteristics. Methods In a study of 170 patients undergoing cardiac surgery with cardiopulmonary bypass, enzyme-linked immunosorbent assay (ELISA) measured plasma suPAR levels. Multivariable logistic regression analysis identified risk factors associated with CSA-AKI. Subsequently, the CSA-AKI nomogram model was developed using R software. Predictive performance was evaluated using a receiver operating characteristic (ROC) curve and the area under the curve (AUC). Internal validation was performed through the Bootstrap method with 1000 repeated samples. Additionally, decision curve analysis (DCA) assessed the clinical applicability of the model. Results Multivariable logistic regression analysis revealed that being male, age ≥ 50 years, operation time ≥ 290 minutes, postoperative plasma suPAR at 2 hours, and preoperative left ventricular ejection fraction (LVEF) were independent risk factors for CSA-AKI. Employing these variables as predictive factors, a nomogram model was constructed, an ROC curve was generated, and the AUC was computed as 0.817 (95% CI 0.726-0.907). The calibration curve indicated the accuracy of the model, and the results of DCA demonstrated that the model could benefit the majority of patients. Conclusion Being male, age ≥ 50 years, operation time ≥ 290 minutes, low preoperative LVEF, and elevated plasma suPAR at 2 hours are independent risk factors for CSA-AKI. The nomogram model established based on these risk factors has high accuracy and clinical value, serving as a predictive tool for assessing the risk of CSA-AKI.
Collapse
Affiliation(s)
- Longyin Zhu
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Juan Cai
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Jia Fang
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Lingyu Ran
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Huan Chang
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Huhai Zhang
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Jiamin Zeng
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Qin Yang
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Chunxiao Fu
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Qingping Li
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Qianguang Pan
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| | - Hongwen Zhao
- Department of Nephrology, the First Hospital Affiliated to Army Military Medical University (Southwest Hospital), Chongqing, People’s Republic of China
| |
Collapse
|
20
|
Nusshag C, Theobald V, Wortmann M, Kaimann P, Dietrich M, Gruneberg D, Tourelle K, von der Forst M, Weigand MA, Bischoff MS, Böckler D, Schmitt FCF. Biomarker-guided detection of acute kidney injury in abdominal aortic surgery: the new and the old. Front Med (Lausanne) 2024; 11:1386018. [PMID: 39021823 PMCID: PMC11251966 DOI: 10.3389/fmed.2024.1386018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Acute kidney injury (AKI) is a common complication in patients undergoing major vascular surgery. Despite significant research efforts in this area, the incidence of AKI remains high, posing a significant challenge to healthcare systems, especially in situations where resources are limited. Early prediction of AKI severity and individualized postoperative care is therefore essential. Methods The primary objective of this exploratory study was to assess the diagnostic value of urine cell-cycle arrest biomarkers [(TIMP-2) × (IGFBP7)] and soluble urokinase plasminogen activator receptor (suPAR) for predicting moderate or severe AKI within 24 h after open aortic surgery, and compared to routine kidney biomarkers. Seventy-five patients undergoing elective aortic surgery were included. Clinical parameters, urine and blood samples were collected preoperatively, immediately postoperatively, and 24 h later. AKI was defined using KDIGO criteria. Individual and combined diagnostic performance of biomarkers were evaluated. Results Of the 75 patients, 61% developed AKI, of which 28% developed moderate or severe AKI within 24 h of surgery. Baseline demographics, comorbidities and kidney parameters did not differ between patients with moderate or severe AKI (AKI II/III) and none or mild AKI (AKI 0/I), except for higher preoperative suPAR levels in later AKI II/III patients. Urine osmolality, Cystatin C and serum creatinine had the highest predictive power for AKI II/III with AUCs of 0.75-0.72. (TIMP-2) × (IGFBP7), and neither (TIMP-2) × (IGFBP7) nor suPAR individually showed superior diagnostic value. Combining CysC or SCr with urine osmolality and 6 h urine output gave the best performance with AUCs of 0.86 (95% CI, 0.74-0.96) and 0.85 (95% CI, 0.75-0.95) respectively. Conclusion Our study suggests that routine parameters like urine osmolality, CysC, SCr and 6 h urine output perform best in predicting postoperative AKI after aortic surgery compared to the new biomarkers (TIMP-2) × (IGFBP7) and suPAR. Combining biomarkers, particularly CysC or SCr with urine output, urine osmolality, may enhance diagnostic accuracy. Further validation in larger cohorts and clinical settings is warranted to establish their clinical utility.
Collapse
Affiliation(s)
- Christian Nusshag
- Department of Nephrology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Vivienne Theobald
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Markus Wortmann
- Department of Vascular, Endovascular Surgery and Transplant Surgery, Stuttgart Hospital, Stuttgart, Germany
| | - Philipp Kaimann
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Maximilian Dietrich
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Daniel Gruneberg
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Kevin Tourelle
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Maik von der Forst
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Markus A. Weigand
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Moritz S. Bischoff
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Dittmar Böckler
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix C. F. Schmitt
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
21
|
Entwistle A, Walker S, Knecht A, Strum SL, Shah AA, Pustavoitau A, Mitin N, Williams JB. A Signature of Pre-Operative Biomarkers of Cellular Senescence to Predict Risk of Cardiac and Kidney Adverse Events after Cardiac Surgery. CARDIOLOGY AND CARDIOVASCULAR MEDICINE 2024; 8:267-274. [PMID: 39328896 PMCID: PMC11426411 DOI: 10.26502/fccm.92920387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Importance Improved pre-operative risk stratification methods are needed for targeted risk mitigation and optimization of care pathways for cardiac patients. This is the first report demonstrating pre-operative, aging-related biomarkers of cellular senescence and immune system function can predict risk of common and serious cardiac surgery-related adverse events. Design Multi-center 331-patient cohort study that enrolled patients undergoing coronary artery bypass grafing (CABG) surgery with 30-day follow-up. Included a quaternary care center and two community-based hospitals. Primary outcome was KDIGO-defined acute kidney injury (AKI). Secondary outcomes: decline in eGFR ≥25% at 30d and a composite of major adverse cardiac and kidney events at 30d (MACKE30). Biomarkers were assessed in blood samples collected prior to surgery. Results A multivariate regression model of six senescence biomarkers (p16, p14, LAG3, CD244, CD28 and suPAR) identified patients at risk for AKI (NPV 86.6%, accuracy 78.6%), decline in eGFR (NPV 93.5%, accuracy 85.2%), and MACKE30 (NPV 91.4%, accuracy 79.9%). Patients in the top risk tertile had 7.8 (3.3-18.4) higher odds of developing AKI, 4.5 (1.6-12.6) higher odds of developing renal decline at 30d follow-up, and 5.7 (2.1-15.6) higher odds of developing MACKE30 versus patients in the bottom tertile. All models remained significant when adjusted for clinical variables. Conclusions A network of senescence biomarkers, a fundamental mechanism of aging, can identify patients at risk for adverse kidney and cardiac events when measured pre-operatively. These findings lay the foundation to improve pre-surgical risk assessment with measures that capture heterogeneity of aging, thereby improving clinical outcomes and resource utilization in cardiac surgery.
Collapse
Affiliation(s)
| | | | | | | | - Asad A Shah
- Hoag Memorial Hospital Presbyterian, Newport Beach, CA, USA
| | - Aliaksei Pustavoitau
- Division of Adult Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Judson B Williams
- Cardiovascular and Thoracic Surgery, Wake Med Health & Hospitals, Raleigh, NC, USA
| |
Collapse
|
22
|
Yang HH, Chen YC, Ho CC, Hsu BG. Association between the Serum Soluble Urokinase Plasminogen Activator Receptor and Peripheral Arterial Stiffness According to the Cardio-Ankle Vascular Index in Patients Undergoing Kidney Transplantation. Rev Cardiovasc Med 2024; 25:219. [PMID: 39076328 PMCID: PMC11270095 DOI: 10.31083/j.rcm2506219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/16/2024] [Accepted: 03/27/2024] [Indexed: 07/31/2024] Open
Abstract
Background High soluble urokinase plasminogen activator receptor (suPAR) levels are correlated with cardiovascular (CV) disease. Arterial stiffness is associated with aging-related vascular diseases and is an independent risk factor for CV morbidity and mortality. It can be measured by the cardio-ankle vascular index (CAVI). We evaluated the association between serum suPAR levels and arterial stiffness according to the CAVI in kidney transplantation (KT) recipients. Methods In this study, 82 patients undergoing KT were enrolled. Serum suPAR levels were analyzed using an enzyme immunoassay. The CAVI was measured using a plethysmograph waveform device, and patients with a CAVI of ≥ 9.0 were assigned to the peripheral arterial stiffness (PAS) group. Results Twenty KT patients (24.4%) had PAS, were of older age (p = 0.042), and had higher serum triglyceride (p = 0.023) and suPAR levels (p < 0.001) than the normal group. After adjusting for factors significantly associated with PAS by multivariate logistic regression analysis, serum suPAR levels (odds ratio [OR] 1.072, 95% confidence interval (CI) 1.023-1.123; p = 0.004) were independently associated with PAS in KT patients. The logarithmically transformed suPAR level (log-suPAR) was also positively correlated with the left or right CAVI values (all p < 0.001) from the results of the Spearman correlation analysis in KT patients. Conclusions Serum suPAR levels are positively associated with left or right CAVI values and are independently associated with PAS in KT patients.
Collapse
Affiliation(s)
- Hsiao-Hui Yang
- Department of Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical
Foundation, 97004 Hualien, Taiwan
- School of Medicine, Tzu Chi University, 97004 Hualien, Taiwan
| | - Yen-Cheng Chen
- Department of Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical
Foundation, 97004 Hualien, Taiwan
- School of Medicine, Tzu Chi University, 97004 Hualien, Taiwan
| | - Ching-Chun Ho
- Department of Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical
Foundation, 97004 Hualien, Taiwan
- School of Medicine, Tzu Chi University, 97004 Hualien, Taiwan
| | - Bang-Gee Hsu
- School of Medicine, Tzu Chi University, 97004 Hualien, Taiwan
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical
Foundation, 97004 Hualien, Taiwan
| |
Collapse
|
23
|
Brozat JF, Harbalioğlu N, Hohlstein P, Abu Jhaisha S, Pollmanns MR, Adams JK, Wirtz TH, Hamesch K, Yagmur E, Weiskirchen R, Tacke F, Trautwein C, Koch A. Elevated Serum KIM-1 in Sepsis Correlates with Kidney Dysfunction and the Severity of Multi-Organ Critical Illness. Int J Mol Sci 2024; 25:5819. [PMID: 38892009 PMCID: PMC11172102 DOI: 10.3390/ijms25115819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
The kidney injury molecule (KIM)-1 is shed from proximal tubular cells in acute kidney injury (AKI), relaying tubular epithelial proliferation. Additionally, KIM-1 portends complex immunoregulation and is elevated after exposure to lipopolysaccharides. It thus may represent a biomarker in critical illness, sepsis, and sepsis-associated AKI (SA-AKI). To characterise and compare KIM-1 in these settings, we analysed KIM-1 serum concentrations in 192 critically ill patients admitted to the intensive care unit. Irrespective of kidney dysfunction, KIM-1 serum levels were significantly higher in patients with sepsis compared with other critical illnesses (191.6 vs. 132.2 pg/mL, p = 0.019) and were highest in patients with urogenital sepsis, followed by liver failure. Furthermore, KIM-1 levels were significantly elevated in critically ill patients who developed AKI within 48 h (273.3 vs. 125.8 pg/mL, p = 0.026) or later received renal replacement therapy (RRT) (299.7 vs. 146.3 pg/mL, p < 0.001). KIM-1 correlated with markers of renal function, inflammatory parameters, hematopoietic function, and cholangiocellular injury. Among subcomponents of the SOFA score, KIM-1 was elevated in patients with hyperbilirubinaemia (>2 mg/dL, p < 0.001) and thrombocytopenia (<150/nL, p = 0.018). In univariate and multivariate regression analyses, KIM-1 predicted sepsis, the need for RRT, and multi-organ dysfunction (MOD, SOFA > 12 and APACHE II ≥ 20) on the day of admission, adjusting for relevant comorbidities, bilirubin, and platelet count. Additionally, KIM-1 in multivariate regression was able to predict sepsis in patients without prior (CKD) or present (AKI) kidney injury. Our study suggests that next to its established role as a biomarker in kidney dysfunction, KIM-1 is associated with sepsis, biliary injury, and critical illness severity. It thus may offer aid for risk stratification in these patients.
Collapse
Affiliation(s)
- Jonathan Frederik Brozat
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Neval Harbalioğlu
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Philipp Hohlstein
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Samira Abu Jhaisha
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Maike Rebecca Pollmanns
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Jule Katharina Adams
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Theresa Hildegard Wirtz
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Karim Hamesch
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Eray Yagmur
- Institute of Laboratory Medicine, Western Palatine Hospital, Hellmut-Hartert-Straße 1, 67655 Kaiserslautern, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany;
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Christian Trautwein
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Alexander Koch
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| |
Collapse
|
24
|
Hamada M, Varkoly KS, Riyadh O, Beladi R, Munuswamy-Ramanujam G, Rawls A, Wilson-Rawls J, Chen H, McFadden G, Lucas AR. Urokinase-Type Plasminogen Activator Receptor (uPAR) in Inflammation and Disease: A Unique Inflammatory Pathway Activator. Biomedicines 2024; 12:1167. [PMID: 38927374 PMCID: PMC11201033 DOI: 10.3390/biomedicines12061167] [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: 03/26/2024] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 06/28/2024] Open
Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a unique protease binding receptor, now recognized as a key regulator of inflammation. Initially, uPA/uPAR was considered thrombolytic (clot-dissolving); however, recent studies have demonstrated its predominant immunomodulatory functions in inflammation and cancer. The uPA/uPAR complex has a multifaceted central role in both normal physiological and also pathological responses. uPAR is expressed as a glycophosphatidylinositol (GPI)-linked receptor interacting with vitronectin, integrins, G protein-coupled receptors, and growth factor receptors within a large lipid raft. Through protein-to-protein interactions, cell surface uPAR modulates intracellular signaling, altering cellular adhesion and migration. The uPA/uPAR also modifies extracellular activity, activating plasminogen to form plasmin, which breaks down fibrin, dissolving clots and activating matrix metalloproteinases that lyse connective tissue, allowing immune and cancer cell invasion and releasing growth factors. uPAR is now recognized as a biomarker for inflammatory diseases and cancer; uPAR and soluble uPAR fragments (suPAR) are increased in viral sepsis (COVID-19), inflammatory bowel disease, and metastasis. Here, we provide a comprehensive overview of the structure, function, and current studies examining uPAR and suPAR as diagnostic markers and therapeutic targets. Understanding uPAR is central to developing diagnostic markers and the ongoing development of antibody, small-molecule, nanogel, and virus-derived immune-modulating treatments that target uPAR.
Collapse
Affiliation(s)
- Mostafa Hamada
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Kyle Steven Varkoly
- Department of Internal Medicine, McLaren Macomb Hospital, Michigan State University College of Human Medicine, 1000 Harrington St., Mt Clemens, MI 48043, USA
| | - Omer Riyadh
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Roxana Beladi
- Department of Neurosurgery, Ascension Providence Hospital, Michigan State University College of Human Medicine, 16001 W Nine Mile Rd, Southfield, MI 48075, USA;
| | - Ganesh Munuswamy-Ramanujam
- Molecular Biology and Immunobiology Division, Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur 603203, India;
| | - Alan Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Hao Chen
- Department of Tumor Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Grant McFadden
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| |
Collapse
|
25
|
Montecillo J, Pirker T, Pemberton C, Chew-Harris J. suPAR in cardiovascular disease. Adv Clin Chem 2024; 121:89-131. [PMID: 38797545 DOI: 10.1016/bs.acc.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR), the soluble counterpart of urokinase plasminogen activator receptor, is found in the circulation at various levels. suPAR and its parent molecule, cell surface uPAR, exhibit similar structure and extracellular functional roles facilitating fibrinolysis, cellular adhesion, and migration. Studies have assessed the correlation between suPAR in cardiovascular disease (CVD). It is postulated that suPAR may serve as an indicator of inflammatory activation and burden during CVD progression. Increased suPAR independently predicts poorer outcomes in acute coronary syndromes, in heart failure, as well as in coronary artery disease and atherosclerosis. To guide translation into clinical utization, suPAR has been assessed in numerous CVD settings for improved risk discrimination independently or in association with established traditional risk factors. Whilst the involvement of suPAR has been explored in other diseases such as kidney diseases and cancer, there is only emerging evidence of suPAR's mechanistic involvement in cardiovascular disease. In this review, we provide a background into suPAR and its potential role as a biomarker in CVD.
Collapse
Affiliation(s)
- Jaya Montecillo
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Thomas Pirker
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | | | - Janice Chew-Harris
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
26
|
Füller D, Liu C, Ko YA, Alkhoder AA, Desai SR, Almuwaqqat Z, Patel SA, Ejaz K, Kauser T, Martini MA, Alvi Z, Mehta PK, Sperling LS, Quyyumi AA. Soluble urokinase Plasminogen Activator Receptor (suPAR) mediates the effect of a lower education level on adverse outcomes in patients with coronary artery disease. Eur J Prev Cardiol 2024; 31:521-528. [PMID: 37788634 PMCID: PMC10972630 DOI: 10.1093/eurjpc/zwad311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/16/2023] [Accepted: 09/17/2023] [Indexed: 10/05/2023]
Abstract
AIMS To investigate whether the adverse impact of lower educational attainment on mortality risk in patients with coronary artery disease (CAD) is mediated by the activation of inflammatory and immune pathways, estimated as elevated soluble urokinase plasminogen activator receptor levels. METHODS AND RESULTS In 3164 patients undergoing coronary angiography, we investigated multivariable associations between suPAR and educational attainment and assessed the relationship between a lower educational level (defined as a high-school degree or less as the highest educational qualification) and outcomes using Cox proportional hazard and Fine and Gray's subdistribution competing risk models. The potential mediating effect through suPAR and high-sensitivity C-reactive protein (hs-CRP) was assessed using mediation analysis. A total of 1814 patients (57.3%) had achieved a higher (≥college) education level and 1350 patients (42.7%) a lower (≤high school) education level. Soluble urokinase plasminogen activator receptor levels were 9.0% [95% confidence interval (CI) 6.3-11.8, P ≤ 0.0001] higher in patients with lower educational qualifications than in those with higher educational qualifications after covariate adjustment. Lower educational attainment was associated with a higher risk of cardiovascular death after adjustment for demographic, clinical, and behavioural covariates, including CAD severity and heart failure history, medication use, and hs-CRP levels [hazard ratio 1.26 (95% CI 1.02-1.55, P = 0.03)]. However, after adjustment for suPAR levels, the effect of a lower educational level on cardiovascular death became insignificant. Values were similar for all-cause death. Soluble urokinase plasminogen activator receptor levels mediated 49% and hs-CRP levels 17% of the cardiovascular death risk attributable to lower educational attainment. CONCLUSION Circulating suPAR levels importantly mediate the effects of lower educational attainment on mortality, indicating the importance of systemic inflammation and immune dysregulation as biologic mediators of adverse social determinants of health.
Collapse
Affiliation(s)
- David Füller
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
- Brandenburg Medical School (Theodor Fontane), Brandenburg an der Havel, Germany
| | - Chang Liu
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Ayman A Alkhoder
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Shivang R Desai
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Zakaria Almuwaqqat
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Shivani A Patel
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Kiran Ejaz
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Tanveer Kauser
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Mohamed Afif Martini
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Zain Alvi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Puja K Mehta
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Laurence S Sperling
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road NE, Suite 507, Atlanta, GA 30322, USA
| |
Collapse
|
27
|
Singh R, Watchorn JC, Zarbock A, Forni LG. Prognostic Biomarkers and AKI: Potential to Enhance the Identification of Post-Operative Patients at Risk of Loss of Renal Function. Res Rep Urol 2024; 16:65-78. [PMID: 38476861 PMCID: PMC10928916 DOI: 10.2147/rru.s385856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Acute kidney injury (AKI) is a common complication after surgery and the more complex the surgery, the greater the risk. During surgery, patients are exposed to a combination of factors all of which are associated with the development of AKI. These include hypotension and hypovolaemia, sepsis, systemic inflammation, the use of nephrotoxic agents, tissue injury, the infusion of blood or blood products, ischaemia, oxidative stress and reperfusion injury. Given the risks of AKI, it would seem logical to conclude that early identification of patients at risk of AKI would translate into benefit. The conventional markers of AKI, namely serum creatinine and urine output are the mainstay of defining chronic kidney disease but are less suited to the acute phase. Such concerns are compounded in surgical patients given they often have significantly reduced mobility, suboptimal levels of nutrition and reduced muscle bulk. Many patients may also have misleadingly low serum creatinine and high urine output due to aggressive fluid resuscitation, particularly in intensive care units. Over the last two decades, considerable information has accrued with regard to the performance of what was termed "novel" biomarkers of AKI, and here, we discuss the most examined molecules and performance in surgical settings. We also discuss the application of biomarkers to guide patients' postoperative care.
Collapse
Affiliation(s)
- Rishabh Singh
- Department of Surgery, Royal Surrey Hospital, Guildford, Surrey, UK
| | - James C Watchorn
- Intensive Care Unit, Royal Berkshire NHS Foundation Trust, Reading, Berkshire, UK
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Lui G Forni
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surrey, UK
- School of Medicine, Kate Granger Building, University of Surrey, Guildford, UK
| |
Collapse
|
28
|
Asia LK, Van Vuren EJ, Kruger IM, Williams ME. A Pilot Investigation of the Association Between Vpr Amino Acid Substitutions and Peripheral Immune Marker Levels in People With Human Immunodeficiency Virus: Implications for Neurocognitive Impairment. Open Forum Infect Dis 2024; 11:ofae111. [PMID: 38524224 PMCID: PMC10960601 DOI: 10.1093/ofid/ofae111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Background Subtype-specific amino acid variations in viral proteins of human immunodeficiency virus type 1 (HIV-1) influence disease progression. Furthermore, Vpr sequence variation correlates with chronic inflammation, a central mechanism in HIV-1 (neuro)pathogenesis. Nevertheless, no clinical study has investigated the link between Vpr sequence variation and peripheral inflammation in people with HIV (PWH). The aim of this pilot study was to ascertain whether specific Vpr amino acid variants were associated with immune markers in PWH. Methods We included a unique cohort of 48 treatment-naive South African PWH to determine the association between blood-derived Vpr sequence variation and peripheral immune marker levels using Sanger sequencing and enzyme-linked immunosorbent assay analysis, respectively. Results Our findings indicate that among the many neuropathogenic Vpr amino acid variants and immune markers examined, after applying Bonferroni corrections (P = .05/3) and adjusting for sex and locality, soluble urokinase plasminogen activator receptor (suPAR) was nearing significance for higher levels in participants with the G41 amino acid variant compared to those with the S41 variant (P = .035). Furthermore, amino acid variations at position 41 (between G41 and S41) exhibited a significant association with suPAR (adjusted R2 = 0.089, β = .386 [95% confidence interval, .125-3.251]; P = .035). Conclusions These findings suggest that Vpr amino acid sequence variations might contribute to dysregulated inflammation, which could explain the observed association between specific Vpr variants and HIV-1 (neuro)pathogenesis found in prior research. These Vpr variants merit further investigation to fully understand their roles in HIV-1 pathogenesis and neuropathogenesis.
Collapse
Affiliation(s)
- Levanco K Asia
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Esmé Jansen Van Vuren
- Hypertension in Africa Research Team, North-West University, Potchefstroom, South Africa
- South African Medical Research Council, Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Iolanthé M Kruger
- Africa Unit for Transdisciplinary Health Research, North-West University, Potchefstroom, South Africa
| | - Monray E Williams
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| |
Collapse
|
29
|
Garcia NA, Gonzalez-King H, Mellergaard M, Nair S, Salomon C, Handberg A. Comprehensive strategy for identifying extracellular vesicle surface proteins as biomarkers for chronic kidney disease. Front Physiol 2024; 15:1328362. [PMID: 38379702 PMCID: PMC10877036 DOI: 10.3389/fphys.2024.1328362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Chronic kidney disease (CKD) poses a significant health burden worldwide. Especially, obesity-induced chronic kidney disease (OCKD) is associated with a lack of accuracy in disease diagnostic methods. The identification of reliable biomarkers for the early diagnosis and monitoring of CKD and OCKD is crucial for improving patient outcomes. Extracellular vesicles (EVs) have emerged as potential biomarkers in the context of CKD. In this review, we focused on the role of EVs as potential biomarkers in CKD and OCKD and developed a comprehensive list of EV membrane proteins that could aid in the diagnosis and monitoring of the disease. To assemble our list, we employed a multi-step strategy. Initially, we conducted a thorough review of the literature on EV protein biomarkers in kidney diseases. Additionally, we explored papers investigating circulating proteins as biomarkers in kidney diseases. To further refine our list, we utilized the EV database Vesiclepedia.org to evaluate the qualifications of each identified protein. Furthermore, we consulted the Human Protein Atlas to assess the localization of these candidates, with a particular focus on membrane proteins. By integrating the information from the reviewed literature, Vesiclepedia.org, and the Human Protein Atlas, we compiled a comprehensive list of potential EV membrane protein biomarkers for CKD and OCKD. Overall, our review underscores the potential of EVs as biomarkers in the field of CKD research, providing a foundation for future studies aimed at improving CKD and OCKD diagnosis and treatment.
Collapse
Affiliation(s)
| | - Hernan Gonzalez-King
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland, Brisbane, QLD, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland, Brisbane, QLD, Australia
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, The Faculty of Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
30
|
Reisinger AC, Hatzl S, Prattes J, Hackl G, Schilcher G, Eisner F, Niedrist T, Raggam R, Krause R, Eller P. Soluble urokinase plasminogen activator receptor (suPAR) in bronchoalveolar fluid and blood in critically ill patients-a prospective cohort study. Infection 2024; 52:249-252. [PMID: 37973717 PMCID: PMC10811150 DOI: 10.1007/s15010-023-02127-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Soluble urokinase plasminogen activator receptor (suPAR) is a biologically active protein and increased levels are associated with worse outcomes in critically ill patients. suPAR in bronchoalveolar fluid (BALF) may be helpful to differentiate between types of acute respiratory distress syndrome (ARDS) and may have potential for early detection of fungal infection. METHODS We prospectively investigated levels of suPAR in BALF and serum in critically ill patients who underwent bronchoscopy for any reason at the ICU of the Department of Internal Medicine, Medical University of Graz, Graz, Austria. RESULTS Seventy-five patients were available for analyses. Median age was 60 [25th-75th percentile: 50-69] years, 27% were female, and median SOFA score was 12 [11-14] points. Serum suPAR levels were significantly associated with ICU mortality in univariable logistic regression analysis. There was no correlation between BALF and serum suPAR. Serum suPAR was higher in ARDS patients at 11.2 [8.0-17.2] ng/mL compared to those without ARDS at 7.1 [3.7-10.1] (p < 0.001). BALF-suPAR was significantly higher in patients with evidence of fungal lung infection compared to patients without fungal infection both in the general cohort (7.6 [3.2-9.4] vs 2.5 [1.1-5.3], p = 0.013) and in the subgroup of ARDS (7.2 [3.1-39.2] vs 2.5 [1.0-5.2], p = 0.022). All patients were classified as putative/probable invasive aspergillosis. CONCLUSION We found significant higher levels of serum suPAR in ARDS patients compared to those not fulfilling ARDS criteria. Serum and BALF-suPAR were significantly higher in those patients with evidence for invasive pulmonary aspergillosis. These findings may suggest testing this biomarker for early diagnosis of fungal infection in a greater cohort.
Collapse
Affiliation(s)
- Alexander C Reisinger
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Stefan Hatzl
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Juergen Prattes
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gerald Hackl
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Gernot Schilcher
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Florian Eisner
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Tobias Niedrist
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Reinhard Raggam
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Robert Krause
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| |
Collapse
|
31
|
Guo Z, Guo Q, Li X, Gao X, Zhang L, Xu K. Urinary biomarkers associated with podocyte injury in lupus nephritis. Front Pharmacol 2024; 15:1324540. [PMID: 38313309 PMCID: PMC10834635 DOI: 10.3389/fphar.2024.1324540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
The most prevalent and devastating form of organ damage in systemic lupus erythematosus (SLE) is lupus nephritis (LN). LN is characterized by glomerular injury, inflammation, cell proliferation, and necrosis, leading to podocyte injury and tubular epithelial cell damage. Assays for urine biomarkers have demonstrated significant promise in the early detection of LN, evaluation of disease activity, and tracking of reaction to therapy. This is because they are non-invasive, allow for frequent monitoring and easy self-collection, transport and storage. Podocyte injury is believed to be a essential factor in LN. The extent and type of podocyte injury could be connected to the severity of proteinuria, making podocyte-derived cellular debris and injury-related urinary proteins potential markers for the diagnosis and monitoring of LN. This article focuses on studies examining urinary biomarkers associated with podocyte injury in LN, offering fresh perspectives on the application of biomarkers in the early detection and management of LN.
Collapse
Affiliation(s)
| | | | | | | | | | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
32
|
Kim EY, Dryer SE. Role of Formyl Peptide Receptors and β-Arrestin-1 in suPAR Signal Transduction in Mouse Podocytes: Interactions with αVβ3-Integrin. Cells 2024; 13:172. [PMID: 38247863 PMCID: PMC10814688 DOI: 10.3390/cells13020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024] Open
Abstract
The soluble urokinase plasminogen activator receptor (suPAR) has been implicated in a wide range of pathological conditions including primary nephrotic syndromes and acute kidney injuries. suPAR can trigger transduction cascades in podocytes by outside-in activation of αVβ3-integrin, but there is evidence that the functional cell surface response element is actually a complex of different types of receptors, which may also include the receptor for advanced glycation end-products (RAGE) and formyl peptide receptors (FPRs). Here we observed that ROS accumulation and Src activation could be evoked by continuous 24 h exposure to either suPAR or the FPR agonist fMLF. Responses to suPAR and fMLF were completely blocked by either the FPR antagonist WRW4 or by the αV-integrin inhibitor cilengitide. Moreover, endogenous podocyte mouse Fpr1 co-immunoprecipitates with β3-integrin, suggesting that these receptors occur as a complex on the cell surface. suPAR- and fMLF-evoked activation of Src and ROS differed in time course. Thus, robust pertussis toxin (PTX)-sensitive responses were evoked by 60 min exposures to fMLF but not to suPAR. By contrast, responses to 24 h exposures to either suPAR or fMLF were PTX-resistant and were instead abolished by knockdown of β-arrestin-1 (BAR1). FPRs, integrins, and RAGE (along with various Toll-like receptors) can all function as pattern-recognition receptors that respond to "danger signals" associated with infections and tissue injury. The fact that podocytes express such a wide array of pattern-recognition receptors suggests that the glomerular filter is designed to change its function under certain conditions, possibly to facilitate clearance of toxic macromolecules.
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, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
33
|
Rossiter A, La A, Koyner JL, Forni LG. New biomarkers in acute kidney injury. Crit Rev Clin Lab Sci 2024; 61:23-44. [PMID: 37668397 DOI: 10.1080/10408363.2023.2242481] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/14/2023] [Accepted: 07/26/2023] [Indexed: 09/06/2023]
Abstract
Acute kidney injury (AKI) is a commonly encountered clinical syndrome. Although it often complicates community acquired illness, it is more common in hospitalized patients, particularly those who are critically ill or who have undergone major surgery. Approximately 20% of hospitalized adult patients develop an AKI during their hospital care, and this rises to nearly 60% in the critically ill, depending on the population being considered. In general, AKI is more common in older adults, in those with preexisting chronic kidney disease and in those with known risk factors for AKI (including diabetes and hypertension). The development of AKI is associated with an increase in both mortality and morbidity, including the development of post-AKI chronic kidney disease. Currently, AKI is defined by a rise in serum creatinine from either a known or derived baseline value and/or oliguria or anuria. However, clinicians may fail to recognize the initial development of AKI because of a delay in the rise of serum creatinine or because of inaccurate urine output monitoring. This, in turn, delays any putative measures to treat AKI or to limit its degree. Consequently, efforts have focused on new biomarkers associated with AKI that may allow early recognition of this syndrome with the intent that this will translate into improved patient outcomes. Here we outline current biomarkers associated with AKI and explore their potential in aiding diagnosis, understanding the pathophysiology and directing therapy.
Collapse
Affiliation(s)
- Adam Rossiter
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
| | - Ashley La
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Jay L Koyner
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Lui G Forni
- Critical Care Unit, Royal Surrey Hospital, Guildford, Surry, UK
- School of Medicine, Department of Clinical & Experimental Medicine, Faculty of Health Sciences, University of Surrey, Surry, UK
| |
Collapse
|
34
|
Vasbinder A, Padalia K, Pizzo I, Machado K, Catalan T, Presswalla F, Anderson E, Ismail A, Hutten C, Huang Y, Blakely P, Azam TU, Berlin H, Feroze R, Launius C, Meloche C, Michaud E, O'Hayer P, Pan M, Shadid HR, Rasmussen LJH, Roberts DA, Zhao L, Banerjee M, Murthy V, Loosen SH, Chalkias A, Tacke F, Reiser J, Giamarellos-Bourboulis EJ, Eugen-Olsen J, Pop-Busui R, Hayek SS. SuPAR, biomarkers of inflammation, and severe outcomes in patients hospitalized for COVID-19: The International Study of Inflammation in COVID-19. J Med Virol 2024; 96:e29389. [PMID: 38235904 PMCID: PMC10829525 DOI: 10.1002/jmv.29389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/13/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024]
Abstract
Severe coronavirus disease 2019 (COVID-19) is a hyperinflammatory syndrome. The biomarkers of inflammation best suited to triage patients with COVID-19 are unknown. We conducted a prospective multicenter observational study of adult patients hospitalized specifically for COVID-19 from February 1, 2020 to October 19, 2022. Biomarkers measured included soluble urokinase plasminogen activator receptor (suPAR), C-reactive protein, interleukin-6, procalcitonin, ferritin, and D-dimer. In-hospital outcomes examined include death and the need for mechanical ventilation. Patients admitted in the United States (US, n = 1962) were used to compute area under the curves (AUCs) and identify biomarker cutoffs. The combined European cohorts (n = 1137) were used to validate the biomarker cutoffs. In the US cohort, 356 patients met the composite outcome of death (n = 197) or need for mechanical ventilation (n = 290). SuPAR was the most important predictor of the composite outcome and had the highest AUC (0.712) followed by CRP (0.642), ferritin (0.619), IL-6 (0.614), D-dimer (0.606), and lastly procalcitonin (0.596). Inclusion of other biomarkers did not improve discrimination. A suPAR cutoff of 4.0 ng/mL demonstrated a sensitivity of 95.4% (95% CI: 92.4%-98.0%) and negative predictive value (NPV) of 92.5% (95% CI: 87.5%-96.9%) for the composite outcome. Patients with suPAR < 4.0 ng/mL comprised 10.6% of the cohort and had a 0.8% probability of the composite outcome. Applying this cutoff to the validation cohort yielded a sensitivity of 93.8% (90.4%-96.7%) and NPV of 95.5% (93.1%-97.8%) for the composite outcome. Among commonly measured biomarkers, suPAR offered stronger discriminatory ability and may be useful in triaging low-risk patients with COVID-19.
Collapse
Affiliation(s)
- Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kishan Padalia
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Pizzo
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kristen Machado
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Tonimarie Catalan
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Feriel Presswalla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth Anderson
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christina Hutten
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yiyuan Huang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Tariq U Azam
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hanna Berlin
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rafey Feroze
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Launius
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Chelsea Meloche
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Erinleigh Michaud
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Patrick O'Hayer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Husam R Shadid
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Donald A Roberts
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Mousumi Banerjee
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Venkatesh Murthy
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sven H Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Outcomes Research Consortium, Cleveland, Ohio, USA
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | | | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Rodica Pop-Busui
- Division of Endocrinology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
35
|
Rotbain Curovic V, Tavenier J, Ferreira-Divino LF, Poulsen CG, Houlind MB, Pedersen OB, Urbak L, Hansen TW, Sillesen H, Frimodt-Møller M, Hvas AM, Rossing P. Soluble urokinase plasminogen activator receptor, platelet aggregation, and carotid plaque thickness in diabetes: A cross-sectional analysis. J Diabetes Complications 2024; 38:108654. [PMID: 38042098 DOI: 10.1016/j.jdiacomp.2023.108654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/04/2023]
Affiliation(s)
| | - Juliette Tavenier
- Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark
| | | | | | - Morten B Houlind
- Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark; University of Copenhagen, Copenhagen, Denmark
| | - Oliver B Pedersen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Lærke Urbak
- Department of Vascular Surgery, Rigshospitalet, Copenhagen, Denmark
| | | | - Henrik Sillesen
- Department of Vascular Surgery, Rigshospitalet, Copenhagen, Denmark
| | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark; University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Gavriilaki E, Bousiou Z, Batsis I, Vardi A, Mallouri D, Koravou EE, Konstantinidou G, Spyridis N, Karavalakis G, Noli F, Patriarcheas V, Masmanidou M, Touloumenidou T, Papalexandri A, Poziopoulos C, Yannaki E, Sakellari I, Politou M, Papassotiriou I. Soluble Urokinase-Type Plasminogen Activator Receptor (suPAR) and Growth Differentiation Factor-15 (GDF-15) Levels Are Significantly Associated with Endothelial Injury Indices in Adult Allogeneic Hematopoietic Cell Transplantation Recipients. Int J Mol Sci 2023; 25:231. [PMID: 38203404 PMCID: PMC10778584 DOI: 10.3390/ijms25010231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Hematopoietic stem cell transplantation-associated thrombotic microangiopathy (HSCT-TMA) and graft-versus-host disease (GvHD) represent life-threatening syndromes after allogeneic hematopoietic stem cell transplantation (allo-HSCT). In both conditions, endothelial dysfunction is a common denominator, and development of relevant biomarkers is of high importance for both diagnosis and prognosis. Despite the fact that soluble urokinase plasminogen activator receptor (suPAR) and growth differentiation factor-15 (GDF-15) have been determined as endothelial injury indices in various clinical settings, their role in HSCT-related complications remains unexplored. In this context, we used immunoenzymatic methods to measure suPAR and GDF-15 levels in HSCT-TMA, acute and/or chronic GVHD, control HSCT recipients, and apparently healthy individuals of similar age and gender. We found considerably greater SuPAR and GDF-15 levels in HSCT-TMA and GVHD patients compared to allo-HSCT and healthy patients. Both GDF-15 and suPAR concentrations were linked to EASIX at day 100 and last follow-up. SuPAR was associated with creatinine and platelets at day 100 and last follow-up, while GDF-15 was associated only with platelets, suggesting that laboratory values do not drive EASIX. SuPAR, but not GDF-15, was related to soluble C5b-9 levels, a sign of increased HSCT-TMA risk. Our study shows for the first time that suPAR and GDF-15 indicate endothelial damage in allo-HSCT recipients. Rigorous validation of these biomarkers in many cohorts may provide utility for their usefulness in identifying and stratifying allo-HSCT recipients with endothelial cell impairment.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Zoi Bousiou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Ioannis Batsis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Anna Vardi
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Despina Mallouri
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Evaggelia-Evdoxia Koravou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Georgia Konstantinidou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Nikolaos Spyridis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Georgios Karavalakis
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Foteini Noli
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Vasileios Patriarcheas
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Marianna Masmanidou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Tasoula Touloumenidou
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Apostolia Papalexandri
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Christos Poziopoulos
- Department of Hematology, Metropolitan Hospital, Neo Faliro, 18547 Athens, Greece;
| | - Evangelia Yannaki
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Ioanna Sakellari
- BMT Unit, Hematology Department, George Papanicolaou General Hospital, 57010 Thessaloniki, Greece; (Z.B.); (I.B.); (A.V.); (D.M.); (E.-E.K.); (G.K.); (N.S.); (G.K.); (F.N.); (V.P.); (M.M.); (T.T.); (A.P.); (E.Y.); (I.S.)
| | - Marianna Politou
- Hematology Laboratory-Blood Bank, Aretaieion Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Ioannis Papassotiriou
- First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| |
Collapse
|
37
|
Helena C, Giulia T, Alessandra C, Roberto G, Caterina S, Sabrina G, Simone B, Tommaso B, Alessandra B, Roberto L. Detection of Inflammatory Biomarker suPAR in COVID-19 Disease With CHORUS TRIO Instrument. Biomark Insights 2023; 18:11772719231210407. [PMID: 38035182 PMCID: PMC10687937 DOI: 10.1177/11772719231210407] [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: 02/26/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Background Deregulation in the urokinase-type plasminogen activator receptor (uPA/uPAR) system is reported in many diseases where the immune system is activated. During SARS-CoV-2 infection, a rise in soluble uPAR (suPAR) levels has been detected and its concentration above 6 µg/L predicts worsening to severe respiratory failure 14 days earlier, with a positive predictive value of 85.9%, and was the prerequisite for a treatment with anakinra, a recombinant IL-1 receptor antagonist that blocks the activity of both IL-1α and IL-1β. Objectives To compare suPAR concentrations measured by CHORUS suPAR on CHORUS TRIO instrument of DIESSE with the commercially available suPARnostic (ViroGates) ELISA assay. Design A single-centre, non-pharmacological, diagnostic study was performed. Results A total of 522 serum samples from patients with COVID-19 were tested for suPAR. CHORUS suPAR resulted accurate and reliable, with a high grade of specificity (97.9%), accuracy (97.3%) and sensitivity (96.9%). The median concentration of suPAR, as determined with CHORUS suPAR, was 6.8 µg/L (interquartile range 4.5-9.7) in patients with moderate disease (n = 465) and 8.5 µg/L (interquartile range 5.4-10.6) in patients with severe disease. Among patients with moderate and severe disease, 60.6% and 71.9%, respectively, reached the cut-off concentration of suPAR ⩾6 µg/L, defining their illness severity and suggesting eligibility to anakinra treatment. Conclusion CHORUS suPAR kit resulted as sensitive, specific, accurate and able to quantify suPAR concentrations in patients with moderate and severe COVID-19.
Collapse
Affiliation(s)
- Cerutti Helena
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Tesi Giulia
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | | | - Guerranti Roberto
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Silvestrini Caterina
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Gori Sabrina
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Bianciardi Simone
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Bandini Tommaso
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Brogi Alessandra
- R&D DIESSE Diagnostica Senese S.p.A. Società Benefit, Monteriggioni, Siena, Italy
| | - Leoncini Roberto
- Laboratorio Patologia Clinica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| |
Collapse
|
38
|
Rehan ST, Hussain HU, Ali E, Kumar KA, Tabassum S, Hasanain M, Shaikh A, Ali G, Yousaf Z, Asghar MS. Role of soluble urokinase type plasminogen activator receptor (suPAR) in predicting mortality, readmission, length of stay and discharge in emergency patients: A systematic review and meta analysis. Medicine (Baltimore) 2023; 102:e35718. [PMID: 37960735 PMCID: PMC10637562 DOI: 10.1097/md.0000000000035718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/28/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Soluble urokinase plasminogen activator receptor (suPAR) is an inflammatory biomarker that is used to predict mortality, readmission, early discharge, and LOS, thus, serves as a useful tool for ED physicians. Our study aims to analyze the efficacy of suPAR in predicting these prognostic markers in ED. METHODS We performed a comprehensive search on 6 databases from the inception to 30th November 2022, to select the following eligibility criteria; a) observation or triage trial studies investigating the role of suPAR levels in predicting: 30 day and 90-day mortality, 30-day readmission, early discharge (within 24hr), and LOS in patients coming to AMU. RESULTS A total of 13 studies were included, with a population size of 35,178, of which 52.9% were female with a mean age of 62.93 years. Increased risk of 30-day mortality (RR = 10.52; 95% CI = 4.82-22.95; I2 = 38%; P < .00001), and risk of 90-day mortality (RR = 5.76; 95% CI = 3.35-9.91; I2 = 36%; P < .00001) was observed in high suPAR patients. However, a slightly increased risk was observed for 30-day readmission (RR = 1.50; 95% CI = 1.16-1.94; I2 = 54%; P = .002). More people were discharged within 24hr in the low suPAR level group compared to high suPAR group (RR = 0.46; 95% CI = 0.40-0.53; I2 = 41%; P < .00001). LOS was thrice as long in high suPAR level patients than in patients with low suPAR (WMD = 3.20; 95% CI = 1.84-4.56; I2 = 99%; P < .00001). CONCLUSION suPAR is proven to be a significant marker in predicting 30-day and 90-day mortality in ED patients.
Collapse
Affiliation(s)
| | | | - Eman Ali
- Dow University of Health Sciences, Karachi, Pakistan
| | | | | | | | - Asim Shaikh
- Department of Medicine, The Aga Khan University, Karachi, Pakistan
| | - Gibran Ali
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic - Rochester, NY, USA
| | - Zohaib Yousaf
- Department of Internal Medicine, Tower Health - Reading Hospital, PA, USA
| | | |
Collapse
|
39
|
Vallianou NG, Kounatidis D, Panagopoulos F, Evangelopoulos A, Stamatopoulos V, Papagiorgos A, Geladari E, Dalamaga M. Gut Microbiota and Its Role in the Brain-Gut-Kidney Axis in Hypertension. Curr Hypertens Rep 2023; 25:367-376. [PMID: 37632662 DOI: 10.1007/s11906-023-01263-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2023] [Indexed: 08/28/2023]
Abstract
PURPOSE OF REVIEW The role of the gut microbiota in modulating blood pressure is increasingly being recognized, currently. The purpose of this review is to summarize recent findings about the mechanisms involved in hypertension with regard to the phenomenon of "gut dysbiosis." RECENT FINDINGS Gut dysbiosis, i.e., the imbalance between the gut microbiota and the host, is characterized by a disruption of the tight junction proteins, such as occludins, claudins, and JAMs (junctional adhesion molecules), resulting in increased gut permeability or the so called "leaky gut." Due to the influence of genetic as well as environmental factors, various metabolites produced by the gut microbiota, such as indole and p-cresol, are increased. Thereby, uremic toxins, such as indoxyl sulfates and p-cresol sulfates, accumulate in the blood and the urine, causing damage in the podocytes and the tubular cells. In addition, immunological mechanisms are implicated as well. In particular, a switch from M2 macrophages to M1 macrophages, which produce pro-inflammatory cytokines, occurs. Moreover, a higher level of Th17 cells, releasing large amounts of interleukin-17 (IL-17), has been reported, when a diet rich in salt is consumed. Therefore, apart from the aggravation of uremic toxins, which may account for direct harmful effects on the kidney, there is inflammation not only in the gut, but in the kidneys as well. This crosstalk between the gut and the kidney is suggested to play a crucial role in hypertension. Notably, the brain is also implicated, with an increasing sympathetic output. The brain-gut-kidney axis seems to be deeply involved in the development of hypertension and chronic kidney disease (CKD). The notion that, by modulating the gut microbiota, we could regulate blood pressure is strongly supported by the current evidence. A healthy diet, low in animal protein and fat, and low in salt, together with the utilization of probiotics, prebiotics, synbiotics, or postbiotics, may contribute to our fight against hypertension.
Collapse
Affiliation(s)
| | | | - Fotis Panagopoulos
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | | | | | | | - Eleni Geladari
- Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias str, Athens, Greece
| |
Collapse
|
40
|
Rotbain Curovic V, Houlind MB, Kroonen MYAM, Jongs N, Zobel EH, Hansen TW, Tavenier J, Eugen-Olsen J, Laverman GD, Kooy A, Persson F, Rossing P, Heerspink HJL. Overall and inter-individual effect of four different drug classes on soluble urokinase plasminogen activator receptor in type 1 and type 2 diabetes. Diabetes Obes Metab 2023; 25:3152-3160. [PMID: 37417375 DOI: 10.1111/dom.15209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023]
Abstract
AIM To evaluate the effect of four different drug classes on soluble urokinase plasminogen activator receptor (suPAR), a biomarker active in multiple inflammatory processes and a risk factor for complications, in people with type 1 and type 2 diabetes. METHODS We conducted post hoc analyses of a randomized, open-label, crossover trial including 26 adults with type 1 and 40 with type 2 diabetes with urinary albumin-creatinine ratio ≥30 and ≤500 mg/g assigned to 4-week treatments with telmisartan 80 mg, empagliflozin 10 mg, linagliptin 5 mg and baricitinib 2 mg, separated by 4-week washouts. Plasma suPAR was measured before and after each treatment. SuPAR change after each treatment was calculated and, for each individual, the best suPAR-reducing drug was identified. Subsequently, the effect of the best individual drug was compared against the mean of the other three drugs. Repeated-measures linear mixed-effects models were employed. RESULTS The baseline median (interquartile range) plasma suPAR was 3.5 (2.9, 4.3) ng/mL. No overall effect on suPAR levels was observed for any one drug. The individual best-performing drug varied, with baricitinib being selected for 20 participants (30%), followed by empagliflozin for 19 (29%), linagliptin for 16 (24%) and telmisartan for 11 (17%). The individual best-performing drug reduced suPAR by 13.3% (95% confidence interval [CI] 3.7, 22.8; P = 0.007). The difference in suPAR response between the individual best-performing drug and the other three was -19.7% (95% CI -23.1, -16.3; P < 0.001). CONCLUSIONS We demonstrated no overall effect of 4-week treatment with telmisartan, empagliflozin, linagliptin or baricitinib on suPAR. However, individualization of treatment might significantly reduce suPAR levels.
Collapse
Affiliation(s)
| | - Morten B Houlind
- Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | | | - Niels Jongs
- University Medical Center Groningen, Groningen, Netherlands
| | | | | | - Juliette Tavenier
- Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
| | | | | | - Adriaan Kooy
- University Medical Center Groningen, Groningen, Netherlands
- Bethesda Diabetes Research Center, Hoogeveen, Netherlands
| | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
41
|
Sommerer C, Müller-Krebs S, Nadal J, Schultheiss UT, Friedrich N, Nauck M, Schmid M, Nußhag C, Reiser J, Eckardt KU, Zeier M, Hayek SS. Prospective Cohort Study of Soluble Urokinase Plasminogen Activation Receptor and Cardiovascular Events in Patients With CKD. Kidney Int Rep 2023; 8:2265-2275. [PMID: 38025216 PMCID: PMC10658273 DOI: 10.1016/j.ekir.2023.08.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/28/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Soluble urokinase plasminogen activation receptor (suPAR) is an immune-derived pathogenic factor for kidney and atherosclerotic disease. Whether the association between suPAR and cardiovascular (CV) outcomes is dependent on the severity of underlying kidney disease is unclear. Methods We measured serum suPAR levels in 4994 participants (mean age 60 years; 60% men; 36% with diabetes mellitus; mean estimated glomerular filtration rate (eGFR) 49 ml/min per 1.73 m2, SD 18) of the German Chronic Kidney Disease (GCKD) cohort and examined its association with all-cause death, CV death, and major CV events (MACE) across the range of eGFR and urine albumin-to-creatinine ratio (UACR). Results The median suPAR level was 1771 pg/ml (interquartile range [IQR] 1447-2254 pg/ml). SuPAR levels were positively and independently correlated with age, eGFR, UACR, and parathyroid hormone levels. There were 573 deaths, including 190 CV deaths and 683 MACE events at a follow-up time of 6.5 years. In multivariable analyses, suPAR levels (log2) were associated with all-cause death (hazard ratio [HR] 1.36, 95% confidence interval [CI] 1.21-1.53), CV death (HR 1.27, 95% CI 1.03-1.57), and MACE (HR 1.13, 95% CI 1.00-1.28), and were not found to differ according to diabetes mellitus status, baseline eGFR, UACR, or parathyroid hormone levels. In mediation analysis, suPAR's direct effect on all-cause death, CV death, and MACE accounted for 77%, 67%, and 60% of the total effect, respectively; whereas the effect mediated through eGFR accounted for 23%, 34%, and 40%, respectively. Conclusion In a large cohort of individuals with chronic kidney disease (CKD), suPAR levels were associated with mortality and CV outcomes independently of indices of kidney function, consistent with its independent role in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Claudia Sommerer
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Sandra Müller-Krebs
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jennifer Nadal
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Ulla T. Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Medicine IV, Nephrology and Primary Care, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Schmid
- Department of Medical Biometry, Informatics, and Epidemiology (IMBIE), University Hospital Bonn, Bonn, Germany
| | - Christian Nußhag
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Zeier
- Department of Nephrology, University Hospital Heidelberg, Renal Center, Heidelberg, Germany
| | - Salim S. Hayek
- Department of Medicine, Division of Cardiology, University of Michigan, Michigan, USA
| |
Collapse
|
42
|
Samoni S, De Rosa S, Ronco C, Castellano G. Update on persistent acute kidney injury in critical illnesses. Clin Kidney J 2023; 16:1813-1823. [PMID: 37915904 PMCID: PMC10616499 DOI: 10.1093/ckj/sfad107] [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: 02/03/2023] [Indexed: 11/03/2023] Open
Abstract
Acute kidney injury (AKI) affects about half of patients admitted to the intensive care unit (ICU), and worsens their short- and long-term outcomes. Apparently self-limiting AKI episodes initiate a progression toward chronic kidney disease (CKD) through cellular and molecular mechanisms that are yet to be explained. In particular, persistent AKI, defined in 2016 by the Acute Dialysis Quality Initiative as an AKI which lasts more than 48 h from its onset, has been correlated with higher morbidity and mortality, and with a higher progression to acute kidney disease (AKD) and CKD than transient AKI (i.e. AKI with a reversal within 48 h). This classification has been also used in the setting of solid organ transplantation, demonstrating similar outcomes. Due to its incidence and poor prognosis and because prompt interventions seem to change its course, persistent AKI should be recognized early and followed-up also after its recovery. However, while AKI and CKD are well-described syndromes, persistent AKI and AKD are relatively new entities. The purpose of this review is to highlight the key phases of persistent AKI in ICU patients in terms of both clinical and mechanistic features in order to offer to clinicians and researchers an updated basis from which to start improving patients' care and direct future research.
Collapse
Affiliation(s)
- Sara Samoni
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia De Rosa
- Centre for Medical Sciences – CISMed, University of Trento, Trento, Italy
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS Trento, Trento, Italy
| | - Claudio Ronco
- Department of Medicine, University of Padova, Padova, Italy
- International Renal Research Institute of Vicenza (IRRIV), Vicenza, Italy
- Department of Nephrology, Dialysis and Renal Transplantation, San Bortolo Hospital, Vicenza, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli studi di Milano, Milan, Italy
| |
Collapse
|
43
|
Alkhaleq HA, Karram T, Fokra A, Hamoud S, Kabala A, Abassi Z. The Protective Pathways Activated in Kidneys of αMUPA Transgenic Mice Following Ischemia\Reperfusion-Induced Acute Kidney Injury. Cells 2023; 12:2497. [PMID: 37887341 PMCID: PMC10605904 DOI: 10.3390/cells12202497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/12/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023] Open
Abstract
Despite the high prevalence of acute kidney injury (AKI), the therapeutic approaches for AKI are disappointing. This deficiency stems from the poor understanding of the pathogenesis of AKI. Recent studies demonstrate that αMUPA, alpha murine urokinase-type plasminogen activator (uPA) transgenic mice, display a cardioprotective pathway following myocardial ischemia. We hypothesize that these mice also possess protective renal pathways. Male and female αMUPA mice and their wild type were subjected to 30 min of bilateral ischemic AKI. Blood samples and kidneys were harvested 48 h following AKI for biomarkers of kidney function, renal injury, inflammatory response, and intracellular pathways sensing or responding to AKI. αMUPA mice, especially females, exhibited attenuated renal damage in response to AKI, as was evident from lower SCr and BUN, normal renal histology, and attenuated expression of NGAL and KIM-1. Notably, αMUPA females did not show a significant change in renal inflammatory and fibrotic markers following AKI as compared with wild-type (WT) mice and αMUPA males. Moreover, αMUPA female mice exhibited the lowest levels of renal apoptotic and autophagy markers during normal conditions and following AKI. αMUPA mice, especially the females, showed remarkable expression of PGC1α and eNOS following AKI. Furthermore, MUPA mice showed a significant elevation in renal leptin expression before and following AKI. Pretreatment of αMUPA with leptin-neutralizing antibodies prior to AKI abolished their resistance to AKI. Collectively, the kidneys of αMUPA mice, especially those of females, are less susceptible to ischemic I/R injury compared to WT mice, and this is due to nephroprotective actions mediated by the upregulation of leptin, eNOS, ACE2, and PGC1α along with impaired inflammatory, fibrotic, and autophagy processes.
Collapse
Affiliation(s)
- Heba Abd Alkhaleq
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
| | - Tony Karram
- Department of Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Ahmad Fokra
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
| | - Shadi Hamoud
- Internal Medicine, Rambam Health Care Campus, Haifa 3109601, Israel;
| | - Aviva Kabala
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (H.A.A.); (A.F.); (A.K.)
- Laboratory Medicine, Rambam Health Care Campus, Haifa 3109601, Israel
| |
Collapse
|
44
|
Leth JM, Newcombe EA, Grønnemose AL, Jørgensen JT, Qvist K, Clausen AS, Knudsen LBS, Kjaer A, Kragelund BB, Jørgensen TJD, Ploug M. Targeted imaging of uPAR expression in vivo with cyclic AE105 variants. Sci Rep 2023; 13:17248. [PMID: 37821532 PMCID: PMC10567728 DOI: 10.1038/s41598-023-43934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/30/2023] [Indexed: 10/13/2023] Open
Abstract
A comprehensive literature reports on the correlation between elevated levels of urokinase-type plasminogen activator receptor (uPAR) and the severity of diseases with chronic inflammation including solid cancers. Molecular imaging is widely used as a non-invasive method to locate disease dissemination via full body scans and to stratify patients for targeted treatment. To date, the only imaging probe targeting uPAR that has reached clinical phase-II testing relies on a high-affinity 9-mer peptide (AE105), and several studies by positron emission tomography (PET) scanning or near-infra red (NIR) fluorescence imaging have validated its utility and specificity in vivo. While our previous studies focused on applying various reporter groups, the current study aims to improve uPAR-targeting properties of AE105. We successfully stabilized the small uPAR-targeting core of AE105 by constraining its conformational landscape by disulfide-mediated cyclization. Importantly, this modification mitigated the penalty on uPAR-affinity typically observed after conjugation to macrocyclic chelators. Cyclization did not impair tumor targeting efficiency of AE105 in vivo as assessed by PET imaging and a trend towards increased tracer uptake was observed. In future studies, we predict that this knowledge will aid development of new fluorescent AE105 derivatives with a view to optical imaging of uPAR to assist precision guided cancer surgery.
Collapse
Affiliation(s)
- Julie Maja Leth
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, 2200, Copenhagen N, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen N, Denmark
| | - Estella Anne Newcombe
- Structural Biology and NMR Laboratory, Copenhagen N, Denmark
- REPIN, Copenhagen N, Denmark
- The Linderstrøm Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, 2200, Copenhagen N, Denmark
| | - Anne Louise Grønnemose
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, 2200, Copenhagen N, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen N, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230, Odense M, Denmark
| | - Jesper Tranekjær Jørgensen
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Katrine Qvist
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Anne Skovsbo Clausen
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Line Bruhn Schneider Knudsen
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine and Cluster for Molecular Imaging, Copenhagen University Hospital - Rigshospitalet, Copenhagen N, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Birthe Brandt Kragelund
- Structural Biology and NMR Laboratory, Copenhagen N, Denmark
- REPIN, Copenhagen N, Denmark
- The Linderstrøm Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, 2200, Copenhagen N, Denmark
| | | | - Michael Ploug
- Finsen Laboratory, Copenhagen University Hospital - Rigshospitalet, 2200, Copenhagen N, Denmark.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen N, Denmark.
| |
Collapse
|
45
|
Zhu K, Mukherjee K, Wei C, Hayek SS, Collins A, Gu C, Corapi K, Altintas MM, Wang Y, Waikar SS, Bianco AC, Koch A, Tacke F, Reiser J, Sever S. The D2D3 form of uPAR acts as an immunotoxin and may cause diabetes and kidney disease. Sci Transl Med 2023; 15:eabq6492. [PMID: 37729431 DOI: 10.1126/scitranslmed.abq6492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is a risk factor for kidney diseases. In addition to suPAR, proteolysis of membrane-bound uPAR results in circulating D1 and D2D3 proteins. We showed that when exposed to a high-fat diet, transgenic mice expressing D2D3 protein developed progressive kidney disease marked by microalbuminuria, elevated serum creatinine, and glomerular hypertrophy. D2D3 transgenic mice also exhibited insulin-dependent diabetes mellitus evidenced by decreased levels of insulin and C-peptide, impaired glucose-stimulated insulin secretion, decreased pancreatic β cell mass, and high fasting blood glucose. Injection of anti-uPAR antibody restored β cell mass and function in D2D3 transgenic mice. At the cellular level, the D2D3 protein impaired β cell proliferation and inhibited the bioenergetics of β cells, leading to dysregulated cytoskeletal dynamics and subsequent impairment in the maturation and trafficking of insulin granules. D2D3 protein was predominantly detected in the sera of patients with nephropathy and insulin-dependent diabetes mellitus. These sera inhibited glucose-stimulated insulin release from human islets in a D2D3-dependent manner. Our study showed that D2D3 injures the kidney and pancreas and suggests that targeting this protein could provide a therapy for kidney diseases and insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- Ke Zhu
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kamalika Mukherjee
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Agnieszka Collins
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Changkyu Gu
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Kristin Corapi
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Yong Wang
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
| | - Sushrut S Waikar
- Section of Nephrology, Boston University Chobanian & Avedisian School of Medicine and Boston Medical Center, Boston, MA 02129, USA
| | - Antonio C Bianco
- Division of Endocrinology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Alexander Koch
- Department of Gastroenterology, Metabolic Diseases and Internal Intensive Care Medicine, University Hospital Aachen, 52072 Aachen, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sanja Sever
- Harvard Medical School and Division of Nephrology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
46
|
Desai SR, Dhindsa DS, Ko YA, Sandesara PB, Mehta A, Liu C, Tahhan AS, Hayek SS, Ejaz K, Hooda A, Alkhoder A, Islam SJ, Rogers SC, Beshiri A, Murtagh G, Kim JH, Wilson P, Almuwaqqat Z, Sperling LS, Quyyumi AA. Aggregate Clinical and Biomarker-Based Model Predicts Adverse Outcomes in Patients With Coronary Artery Disease. Am J Cardiol 2023; 203:315-324. [PMID: 37517126 PMCID: PMC10900119 DOI: 10.1016/j.amjcard.2023.06.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023]
Abstract
Despite guideline-based therapy, patients with coronary artery disease (CAD) are at widely variable risk for cardiovascular events. This variability demands a more individualized risk assessment. Herein, we evaluate the prognostic value of 6 biomarkers: high-sensitivity C-reactive protein, heat shock protein-70, fibrin degradation products, soluble urokinase plasminogen activator receptor, high-sensitivity troponin I, and B-type natriuretic peptide. We then develop a multi-biomarker-based cardiovascular event prediction model for patients with stable CAD. In total, 3,115 subjects with stable CAD who underwent cardiac catheterization at Emory (mean age 62.8 years, 17% Black, 35% female, 57% obstructive CAD, 31% diabetes mellitus) were randomized into a training cohort to identify biomarker cutoff values and a validation cohort for prediction assessment. Main outcomes included (1) all-cause death and (2) a composite of cardiovascular death and nonfatal myocardial infarction (MI) within 5 years. Elevation of each biomarker level was associated with higher event rates in the training cohort. A biomarker risk score was created using optimal cutoffs, ranging from 0 to 6 for each biomarker exceeding its cutoff. In the validation cohort, each unit increase in the biomarker risk score was independently associated with all-cause death (hazard ratio 1.62, 95% confidence interval [CI] 1.45 to 1.80) and cardiovascular death/MI (hazard ratio 1.52, 95% CI 1.35 to 1.71). A biomarker risk prediction model for cardiovascular death/MI improved the c-statistic (∆ 6.4%, 95% CI 3.9 to 8.8) and net reclassification index by 31.1% (95% CI 24 to 37), compared with clinical risk factors alone. Integrating multiple biomarkers with clinical variables refines cardiovascular risk assessment in patients with CAD.
Collapse
Affiliation(s)
- Shivang R Desai
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Devinder S Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Pratik B Sandesara
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Anurag Mehta
- Department of Preventive Cardiology, VCU Health Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Chang Liu
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ayman S Tahhan
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Salim S Hayek
- Department of Medicine, Division of Cardiology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Kiran Ejaz
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ananya Hooda
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ayman Alkhoder
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Shabatun J Islam
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Steven C Rogers
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Agim Beshiri
- Diagnostics Division, Abbott Laboratories, North Chicago, Illinois
| | - Gillian Murtagh
- Diagnostics Division, Abbott Laboratories, North Chicago, Illinois
| | - Jonathan H Kim
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Peter Wilson
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Zakaria Almuwaqqat
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Laurence S Sperling
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
47
|
Landi A, Chiarito M, Branca M, Frigoli E, Gagnor A, Calabrò P, Briguori C, Andò G, Repetto A, Limbruno U, Sganzerla P, Lupi A, Cortese B, Ausiello A, Ierna S, Esposito G, Ferrante G, Santarelli A, Sardella G, Varbella F, Heg D, Mehran R, Valgimigli M. Validation of a Contemporary Acute Kidney Injury Risk Score in Patients With Acute Coronary Syndrome. JACC Cardiovasc Interv 2023; 16:1873-1886. [PMID: 37587595 DOI: 10.1016/j.jcin.2023.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/23/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND A simple, contemporary risk score for the prediction of contrast-associated acute kidney injury (CA-AKI) after percutaneous coronary intervention (PCI) was recently updated, although its external validation is lacking. OBJECTIVES The aim of this study was to validate the updated CA-AKI risk score in a large cohort of acute coronary syndrome patients from the MATRIX (Minimizing Adverse Haemorrhagic Events by Transradial Access Site and Systemic Implementation of angioX) trial. METHODS The risk score identifies 4 risk categories for CA-AKI. The primary endpoint was to appraise the receiver-operating characteristics of an 8-component and a 12-component CA-AKI model. Independent predictors of Kidney Disease Improving Global Outcomes-based acute kidney injury and the impact of CA-AKI on 1-year mortality and bleeding were also investigated. RESULTS The MATRIX trial included 8,201 patients with complete creatinine values and no end-stage renal disease. CA-AKI occurred in 5.5% of the patients, with a stepwise increase of CA-AKI rates from the lowest to the highest of the 4 risk categories. The receiver-operating characteristic area under the curve was 0.67 (95% CI: 0.64-0.70) with model 1 and 0.71 (95% CI: 0.68-0.74) with model 2. CA-AKI risk was systematically overestimated with both models (Hosmer-Lemeshow goodness-of-fit test: P < 0.05). The 1-year risks of all-cause mortality and bleeding were higher in CA-AKI patients (HR: 7.03 [95% CI: 5.47-9.05] and HR: 3.20 [95% CI: 2.56-3.99]; respectively). There was a gradual risk increase for mortality and bleeding as a function of the CA-AKI risk category for both models. CONCLUSIONS The updated CA-AKI risk score identifies patients at incremental risks of acute kidney injury, bleeding, and mortality. (Minimizing Adverse Haemorrhagic Events by Transradial Access Site and Systemic Implementation of angioX [MATRIX]; NCT01433627).
Collapse
Affiliation(s)
- Antonio Landi
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland; Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland
| | - Mauro Chiarito
- Department of Biomedical Sciences, Humanitas University, Emanuele, Italy
| | | | - Enrico Frigoli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Andrea Gagnor
- Department of Invasive Cardiology, Maria Vittoria Hospital, Turin, Italy
| | - Paolo Calabrò
- Division of Cardiology, "Sant'Anna e San Sebastiano" Hospital, Caserta, Italy; Department of Translational Medicine, University of Campania "Luigi Vanvitelli," Caserta, Italy
| | - Carlo Briguori
- Interventional Cardiology Unit, Mediterranea Cardiocentro, Naples, Italy
| | - Giuseppe Andò
- Cardiology Unit, Azienda Ospedaliera Universitaria Policlinico "Gaetano Martino," University of Messina, Messina, Italy
| | | | - Ugo Limbruno
- Cardiology Department, Misericordia Hospital, Grosseto, Italy
| | - Paolo Sganzerla
- IRCCS Istituto Auxologico Italiano, Ospedale San Luca, Milan, Italy
| | - Alessandro Lupi
- Division of Cardiology, Hospital of Domodossola, Domodossola, Verbano-Cusio-Ossola, Italy
| | - Bernardo Cortese
- Cardiovascular Research Center, Fondazione Ricerca e Innovazione Cardiovascolare, Milan, Italy
| | | | - Salvatore Ierna
- Interventional Cardiology Unit, Ospedale di Carbonia, Carbonia, Italy
| | - Giovanni Esposito
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Giuseppe Ferrante
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; Department of Cardiovascular Medicine, Humanitas Research Hospital IRCCS, Rozzano-Milan, Italy
| | | | - Gennaro Sardella
- Department of Cardiovascular Sciences, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Ferdinando Varbella
- Cardiology Unit, Azienda Ospedaliera Universitaria San Luigi Gonzaga Orbassano, Turin, Italy
| | - Dik Heg
- CTU Bern, University of Bern, Bern, Switzerland
| | - Roxana Mehran
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marco Valgimigli
- Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland; Department of Biomedical Sciences, University of Italian Switzerland, Lugano, Switzerland.
| |
Collapse
|
48
|
Wei C, Datta PK, Siegerist F, Li J, Yashwanth S, Koh KH, Kriho NW, Ismail A, Luo S, Fischer T, Amber KT, Cimbaluk D, Landay A, Endlich N, Rappaport J, Hayek SS, Reiser J. SuPAR mediates viral response proteinuria by rapidly changing podocyte function. Nat Commun 2023; 14:4414. [PMID: 37479685 PMCID: PMC10362037 DOI: 10.1038/s41467-023-40165-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023] Open
Abstract
Elevation in soluble urokinase receptor (suPAR) and proteinuria are common signs in patients with moderate to severe coronavirus disease 2019 (COVID-19). Here we characterize a new type of proteinuria originating as part of a viral response. Inoculation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes increased suPAR levels and glomerulopathy in African green monkeys. Using an engineered mouse model with high suPAR expression, inhaled variants of SARS-CoV-2 spike S1 protein elicite proteinuria that could be blocked by either suPAR antibody or SARS-CoV-2 vaccination. In a cohort of 1991 COVID-19 patients, suPAR levels exhibit a stepwise association with proteinuria in non-Omicron, but not in Omicron infections, supporting our findings of biophysical and functional differences between variants of SARS-CoV-2 spike S1 protein and their binding to podocyte integrins. These insights are not limited to SARS-CoV-2 and define viral response proteinuria (VRP) as an innate immune mechanism and co-activation of podocyte integrins.
Collapse
Affiliation(s)
- Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
| | - Prasun K Datta
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany
- NIPOKA GmbH, 17489, Greifswald, Germany
| | - Jing Li
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Sudhini Yashwanth
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kwi Hye Koh
- Morphic Therapeutic, Waltham, MA, 02451, USA
| | - Nicholas W Kriho
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shengyuan Luo
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Tracy Fischer
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Kyle T Amber
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
- Department of Dermatology, Rush University Medical Center, Chicago, IL, USA
| | - David Cimbaluk
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Alan Landay
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17487, Greifswald, Germany
- NIPOKA GmbH, 17489, Greifswald, Germany
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
49
|
Agborbesong E, Bissler J, Li X. Liquid Biopsy at the Frontier of Kidney Diseases: Application of Exosomes in Diagnostics and Therapeutics. Genes (Basel) 2023; 14:1367. [PMID: 37510273 PMCID: PMC10379367 DOI: 10.3390/genes14071367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/08/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
In the era of precision medicine, liquid biopsy techniques, especially the use of urine analysis, represent a paradigm shift in the identification of biomarkers, with considerable implications for clinical practice in the field of nephrology. In kidney diseases, the use of this non-invasive tool to identify specific and sensitive biomarkers other than plasma creatinine and the glomerular filtration rate is becoming crucial for the diagnosis and assessment of a patient's condition. In recent years, studies have drawn attention to the importance of exosomes for diagnostic and therapeutic purposes in kidney diseases. Exosomes are nano-sized extracellular vesicles with a lipid bilayer structure, composed of a variety of biologically active substances. In the context of kidney diseases, studies have demonstrated that exosomes are valuable carriers of information and are delivery vectors, rendering them appealing candidates as biomarkers and drug delivery vehicles with beneficial therapeutic outcomes for kidney diseases. This review summarizes the applications of exosomes in kidney diseases, emphasizing the current biomarkers of renal diseases identified from urinary exosomes and the therapeutic applications of exosomes with reference to drug delivery and immunomodulation. Finally, we discuss the challenges encountered when using exosomes for therapeutic purposes and how these may affect its clinical applications.
Collapse
Affiliation(s)
- Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - John Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38105, USA
- Pediatric Medicine Department, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
50
|
Brody GH, Yu T, Miller GE, Chen E. Longitudinal links between early adolescent temperament and inflammation among young black adults. Psychoneuroendocrinology 2023; 152:106077. [PMID: 36931166 PMCID: PMC10201910 DOI: 10.1016/j.psyneuen.2023.106077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/19/2023]
Abstract
A large body of research demonstrates that inflammation is involved in physical health problems that cause substantial morbidity and early mortality. Given inflammation's role in the etiology of chronic diseases, pediatric scientists have begun to study childhood factors that presage elevation of inflammatory biomarkers later in life. The purpose of this study was to test hypotheses designed to determine whether early adolescent emotionally intense and low attention temperaments forecast (a) inflammation at ages 25 and 29 years and (b) worsening levels of inflammation between these two data points. Toward this end, 307 Black children from the rural southeastern United States participated in an 18-year longitudinal study (mean age at baseline, 11.2 years) to determine whether and how early adolescent's behavioral styles or emotionally intense and low attention temperaments may be associated with absolute and worsening levels of inflammation in young adulthood. When children were 11-13 years of age, different teachers at each age provided assessments of emotionally intense and low attention temperaments. Thus, multiple measures of the same temperament constructs were obtained across 3 years for each participant. At age 25, participants provided data on their self-regulation abilities. Peripheral blood was collected at ages 25 and 29 years from which inflammation was quantified, using soluble urokinase plasminogen activator (suPAR), the proinflammatory cytokines interleukin (IL) IL-6, IL-10, and tumor necrosis factor-alpha (TNF-α). Covariates associated with inflammation in prior studies were also assessed; these included socioeconomic risk, gender, cigarette smoking, body mass index (BMI), adverse childhood experiences (ACEs), depressive symptoms, and medication use. An early adolescent emotionally intense temperament was associated directly with higher suPAR and cytokine levels at age 29, and with worsening cytokine levels between ages 25 and 29. A low attention temperament was associated with suPAR levels at age 29. Collectively, these observations highlight pathways that could underlie health risks associated with early adolescent temperaments. The findings suggest that emotionally intense and low-attention early adolescent temperaments forecast higher and worsening inflammation levels across young adulthood.
Collapse
Affiliation(s)
- Gene H Brody
- Center for Family Research, University of Georgia, Athens, GA, USA.
| | - Tianyi Yu
- Center for Family Research, University of Georgia, Athens, GA, USA
| | - Gregory E Miller
- Department of Psychology, Northwestern University, Evanston, IL, USA; Institute for Policy Research, Northwestern University, Evanston, IL, USA
| | - Edith Chen
- Department of Psychology, Northwestern University, Evanston, IL, USA; Institute for Policy Research, Northwestern University, Evanston, IL, USA
| |
Collapse
|