1
|
Bahrami HSZ, Jørgensen PG, Hove JD, Dixen U, Rasmussen LJH, Eugen-Olsen J, Rossing P, Jensen MT. Association between interleukin-6, suPAR, and hsCRP with subclinical left ventricular dysfunction in type 1 diabetes: The Thousand & 1 study. Diabetes Res Clin Pract 2025; 222:112071. [PMID: 40043809 DOI: 10.1016/j.diabres.2025.112071] [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: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/18/2025]
Abstract
AIMS To investigate the association between chronic inflammation and subclinical left ventricular dysfunction in type 1 diabetes (T1D). METHODS In a cross-sectional study of individuals with T1D without known heart disease, interleukin-6 (IL-6), soluble-urokinase-plasminogen-activator-receptor (suPAR), and high-sensitivity C-reactive-protein (hsCRP) were examined for associations with echocardiographic E/e' (primary outcome) and global longitudinal strain (GLS) (secondary outcome). We adjusted for several clinical variables in linear regression analysis, including N-terminal pro-B-type natriuretic peptide (NT-proBNP). The biomarkers were categorized as elevated/non-elevated based on their upper quartiles. RESULTS Of 962 individuals (52 % male, mean age 49 ± 14 years), mean E/e' was 7 ± 3 and GLS 18 ± 3. In fully adjusted models, all biomarkers were each associated with increased E/e': beta coefficients for IL-6 0.2 (95 % confidence intervals: 0.1-0.3, P = 0.001), suPAR 0.5 (0.1-0.7, P = 0.011), and hsCRP 0.1 (0.0-0.2, P = 0.023). Combining biomarkers showed stronger associations: elevated IL-6 and suPAR 1.3 (0.7-2.0, P < 0.001), elevated all three 1.9 (1.1-2.7, P < 0.001). Results were similar for decreased GLS with IL-6-0.4 (-0.7 to 0.0, P = 0.039), IL-6 and hsCRP -1.0 (-1.7 to -0.4, P = 0.007), all three -1.1 (-2.0 to -0.3, P = 0.009). CONCLUSIONS Inflammatory biomarkers are independently associated with subclinical left ventricular dysfunction. Chronic inflammation may contribute to the development of myocardial dysfunction in T1D.
Collapse
Affiliation(s)
- Hashmat Sayed Zohori Bahrami
- Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark; Department of Cardiology, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark.
| | - Peter Godsk Jørgensen
- Department of Cardiology, Copenhagen University Hospital, Herlev & Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark
| | - Jens Dahlgaard Hove
- Department of Cardiology, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Ulrik Dixen
- Department of Cardiology, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark; Department of Psychology & Neuroscience, Duke University, 2020 W Main St, Durham, NC 27708, USA
| | - Jesper Eugen-Olsen
- Department of Cardiology, Copenhagen University Hospital, Herlev & Gentofte, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark; Department of Clinical Research, Copenhagen University Hospital, Amager & Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark
| | - Peter Rossing
- Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Magnus T Jensen
- Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Borgmester Ib Juuls Vej 83, 2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark; William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
2
|
Wisborg FD, El Caidi NO, Taraldsen IA, Tonning S, Kandiah A, El‐Sheikh M, Bahrami HSZ, Andersen O, Rasmussen LJH, Hove J, Dixen U, Grand J. Soluble urokinase plasminogen activator receptor (suPAR) as a prognostic biomarker in acutely admitted patients with atrial fibrillation. J Arrhythm 2025; 41:e70077. [PMID: 40271386 PMCID: PMC12017082 DOI: 10.1002/joa3.70077] [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: 11/06/2024] [Revised: 03/12/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025] Open
Abstract
Background Atrial fibrillation (AF) is associated with a higher incidence of stroke, heart failure, and mortality. Risk assessment of clinical outcomes in patients hospitalized acutely with AF remains a challenge. Purpose To investigate if soluble urokinase plasminogen activator receptor (suPAR) levels at admission to the Emergency Department (ED) are associated with 1-year all-cause mortality in patients admitted with AF. Methods A prospective cohort study of patients consecutively admitted to the medical ED of a university hospital in Copenhagen, Denmark, between 2020 and 2022 with symptoms of COVID-19. Patients were included if they were admitted with AF as the primary or secondary diagnosis. All patients had suPAR measured at the index admission, and follow-up was up to 1 year. The association between suPAR and 1-year mortality was investigated with multivariate Cox regression. We adjusted for age, sex, smoking, C-reactive protein, creatinine, hemoglobin, albumin, and comorbidities. Results Of the 7,258 patients included during the period, 362 (5.0%) patients were admitted with AF as the primary or secondary diagnosis. Due to missing data, 23 (6.4%) patients were excluded. Among the remaining 339 patients, 68 (20.1%) patients were dead at follow-up. The multivariate Cox regression showed that elevated suPAR was independently associated with an increased risk of 1-year mortality, with a hazard ratio of 1.12 (95% confidence interval: 1.05-1.20, p < 0.001). Conclusion Elevated suPAR levels were significantly associated with 1-year all-cause mortality in patients acutely admitted with AF to the ED.
Collapse
Affiliation(s)
| | - Nora Olsen El Caidi
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Ida Arentz Taraldsen
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Sandra Tonning
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Aginsha Kandiah
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Mohammed El‐Sheikh
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Hashmat S. Z. Bahrami
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Clinical and Translational ResearchSteno Diabetes Center CopenhagenHerlevDenmark
| | - Ove Andersen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Emergency MedicineCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical ResearchCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Department of Emergency MedicineCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jens Hove
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Ulrik Dixen
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| | - Johannes Grand
- Department of CardiologyCopenhagen University Hospital, Amager and HvidovreHvidovreDenmark
| |
Collapse
|
3
|
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
|
4
|
Kostopoulou E, Kalavrizioti D, Davoulou P, Sinopidis X, Papachristou E, Goumenos DS, Dimitriou G, Spiliotis BE, Papasotiriou M. Soluble urokinase plasminogen activator receptor (suPAR) in children with obesity or type 1 diabetes as a marker of endothelial dysfunction: a cross-sectional study. Eur J Pediatr 2024; 183:2383-2389. [PMID: 38448612 DOI: 10.1007/s00431-024-05496-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Pediatric obesity and type 1 diabetes mellitus (T1DM) represent two common chronic diseases associated with chronic inflammation, endothelial dysfunction and long-term complications. The aim of the present study was to assess the possible diagnostic and prognostic value of soluble urokinase plasminogen activator receptor (suPAR), a marker of inflammation and impaired endothelial function, in children with the diseases. In this cross-sectional study, children and adolescents with T1DM (N = 41) or obesity (N = 37), aged < 18 years old, and without proteinuria were included, together with children of similar age and without evident morbidity that served as controls (N = 42). Serum samples were obtained during standard outpatient follow up and the urokinase-type plasminogen activator receptor (suPAR) concentrations were measured using a commercially available sandwich ELISA kit (DUP00, R&D systems). Clinical and biochemical indices that were also assessed include body mass index (BMI) z-score, Tanner stages, glycosylated haemoglobin (HbA1c), fasting lipid profile and serum creatinine. Mean serum suPAR levels were significantly higher in patients with obesity compared to patients with T1DM and controls, while children with T1DM had similar suPAR levels to controls. Also, serum suPAR levels showed a negative correlation with age (Spearman rho -0.359, p < 0.001) and serum creatinine levels (Spearman rho -0.334, p = 0.005), and a positive correlation with BMI z-score (Spearman rho 0.354, p = 0.009) in the whole cohort. Conclusion: Serum suPAR may be a useful predictive marker of inflammation or endothelial dysfunction for children with obesity and T1DM, as well as a promising therapeutic target. Further studies are needed in order to clarify whether the reported differences in suPAR levels could reflect a greater impairment of the inflammation status and endothelial function in children with obesity compared to children with T1DM. What is Known: • Paediatric obesity and type 1 diabetes are characterised by chronic inflammation and metabolic dysregulation. • Urokinase plasminogen activator receptor (uPAR) has been proposed as a useful biomarker for chronic inflammation and cardiovascular risk in adults. What is New: • Serum suPAR levels were increased in children and adolescents with obesity compared to those with T1DM and healthy controls; thus, obesity may affect the inflammatory status and endothelial function to a higher degree than T1DM during childhood. • Serum suPAR may serve as a diagnostic and predictive marker of inflammation and endothelial dysfunction for children and adolescents with obesity and T1DM.
Collapse
Affiliation(s)
| | - Dimitra Kalavrizioti
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | - Panagiota Davoulou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | | | - Evangelos Papachristou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | - Dimitrios S Goumenos
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece
| | - Gabriel Dimitriou
- Department of Pediatrics, University Hospital of Patras, Patras, Greece
| | | | - Marios Papasotiriou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, Patras, 26504, Greece.
| |
Collapse
|
5
|
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
|
6
|
Matthews T, Rasmussen LJH, Ambler A, Danese A, Eugen-Olsen J, Fancourt D, Fisher HL, Iversen KK, Schultz M, Sugden K, Williams B, Caspi A, Moffitt TE. Social isolation, loneliness, and inflammation: A multi-cohort investigation in early and mid-adulthood. Brain Behav Immun 2024; 115:727-736. [PMID: 37992788 PMCID: PMC11194667 DOI: 10.1016/j.bbi.2023.11.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/09/2023] [Accepted: 11/19/2023] [Indexed: 11/24/2023] Open
Abstract
Social isolation and loneliness have been associated with poor health and increased risk for mortality, and inflammation might explain this link. We used data from the Danish TRIAGE Study of acutely admitted medical patients (N = 6,144, mean age 60 years), and from two population-representative birth cohorts: the New Zealand Dunedin Longitudinal Study (N = 881, age 45) and the UK Environmental Risk (E-Risk) Longitudinal Twin Study (N = 1448, age 18), to investigate associations of social isolation with three markers of systemic inflammation: C-reactive protein (CRP), interleukin-6 (IL-6), and a newer inflammation marker, soluble urokinase plasminogen activator receptor (suPAR), which is thought to index systemic chronic inflammation. In the TRIAGE Study, socially isolated patients (those living alone) had significantly higher median levels of suPAR (but not CRP or IL-6) compared with patients not living by themselves. Social isolation prospectively measured in childhood was longitudinally associated with higher CRP, IL-6, and suPAR levels in adulthood (at age 45 in the Dunedin Study and age 18 in the E-Risk Study), but only suPAR remained associated after controlling for covariates. Dunedin Study participants who reported loneliness at age 38 or age 45 had elevated suPAR at age 45. In contrast, E-Risk Study participants reporting loneliness at age 18 did not show any elevated markers of inflammation. In conclusion, social isolation was robustly associated with increased inflammation in adulthood, both in medical patients and in the general population. It was associated in particular with systemic chronic inflammation, evident from the consistently stronger associations with suPAR than other inflammation biomarkers.
Collapse
Affiliation(s)
- Timothy Matthews
- School of Human Sciences, Faculty of Education, Health and Human Sciences, University of Greenwich, London, United Kingdom.
| | - Line Jee Hartmann Rasmussen
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Antony Ambler
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Andrea Danese
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; National and Specialist CAMHS Trauma and Anxiety Clinic, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark; ViroGates A/S, Birkerød, Denmark
| | - Daisy Fancourt
- Department of Behavioural Science and Health, University College London, United Kingdom
| | - Helen L Fisher
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; ESRC Centre for Society and Mental Health, King's College London, London, United Kingdom
| | - Kasper Karmark Iversen
- Department of Emergency Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark; Department of Cardiology, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Martin Schultz
- Department of Emergency Medicine, Copenhagen University Hospital Herlev and Gentofte, Herlev, Denmark
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Benjamin Williams
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
7
|
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
|
8
|
Rasmussen VF, Hirschberg Jensen V, Thrysøe M, Vestergaard ET, Størling J, Kristensen K. Cross-sectional study investigating the association between inflammatory biomarkers and neuropathy in adolescents with type 1 diabetes. BMJ Open 2023; 13:e074992. [PMID: 37802616 PMCID: PMC10565182 DOI: 10.1136/bmjopen-2023-074992] [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: 04/22/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023] Open
Abstract
OBJECTIVES The aims of this study were to investigate circulating levels of inflammatory markers in adolescents with type 1 diabetes with and without different types of neuropathies and evaluate the association between inflammatory biomarkers, nerve function and clinical parameters. DESIGN Cross-sectional study. SETTING Hospitals and Steno Diabetes Center in Denmark. PARTICIPANTS Adolescents with more than 5 years of diabetes duration were investigated for large fibre, small fibre and autonomic neuropathy as a part of the T1DANES study. Blood samples from the participants were analysed for inflammatory biomarkers by Meso Scale Discovery multiplexing technology. PRIMARY AND SECONDARY OUTCOME MEASURES Inflammatory biomarkers and results of diagnostic nerve tests. RESULTS Fifty-six adolescents with type 1 diabetes and 23 healthy controls were included. The adolescents with diabetes had significantly higher interferon-gamma, tumour necrosis factor-alpha (TNF-a), interleukin (IL)-10 and soluble urokinase plasminogen activator receptor (suPAR) compared with healthy controls (p values<0.05). TNF-a was higher in the adolescents with large fibre neuropathy (LFN) (p=0.03) compared with those without LFN in the group with diabetes. A negative correlation was seen between TNF-a and conduction velocity in nervus tibialis (p=0.04), and higher TNF-a and IL-6 were associated with higher gastric motility index (TNF-a, p value=0.03; IL-6, p value=0.02). There were no significant associations between inflammatory markers and expressed symptoms, haemoglobin A1c, diabetes duration or body mass index standard derivation score (p values>0.05). The receiver operating characteristic (ROC) curves for the inflammatory markers suggested them as poor screening methods for all types of neuropathies with an area under the curve between 0.47 and 0.67. CONCLUSION Our results confirm increased low-grade inflammation in adolescents with type 1 diabetes. TNF-a was higher in adolescents with LFN and correlated negatively with nervus tibialis conduction velocity. The other inflammatory biomarkers fail to support differences in those with and without different types of diabetic neuropathies. However, TNF-a and IL-6 were positively correlated to gastric motility index.
Collapse
Grants
- Steno Diabetes Center
- The entire project was sponsored by the following: Skibsreder Per Henriksen og Hustrus Fond, Tømrermester Jørgen Holm og Hustru Lisa F. Hansens Mindelegat, Vissing Fonden, Rissfort Fonden, Kirsten Dyrløv Madsens legat, Lipperts Fond, Reinholdt W. Jorck og Hustrus fond, Helga og Peter Kornings Fond, Beckett Fonden, Dagmar Marschall Fond. Danske lægers Forsikring under Danica Pension, William Demant Fonden. Professor Iversens Rejsefond, Diabetesforeningen.
- Novo Nordisk
- Aarhus University
Collapse
Affiliation(s)
- Vinni Faber Rasmussen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Pediatrics and Adolescents, Randers Regional Hospital, Randers, Denmark
| | | | - Mathilde Thrysøe
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Joachim Størling
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Kristensen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
9
|
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
|
10
|
Erbak Yılmaz H, Aksun S, Günay S, Elmalı F, Çekiç C. Evaluation of plasma soluble urokinase plasminogen activator receptor (SuPAR) levels in ulcerative colitis. Arab J Gastroenterol 2023; 24:175-179. [PMID: 37045728 DOI: 10.1016/j.ajg.2023.03.001] [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: 03/14/2022] [Revised: 02/13/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND AND STUDY AIM Soluble urokinase plasminogen activator receptor (SuPAR), a soluble form of the urokinase-type plasminogen activator receptor, is a biomarker produced by macrophages, monocytes, neutrophils, active T cells, endothelial cells, and circulating tumor cells. SuPAR is a novel biomarker showing altered inflammation in many inflammatory diseases. This study aims to investigate the SuPAR level in ulcerative colitis (UC) patients, and to evaluate the SuPAR level in active, and remission patients. PATIENTS AND METHODS Patient and healthy control SuPAR levels were analyzed by immunoassay method. SuPAR levels between UC patients and control group were compared. The difference between SuPAR levels in patients with active UC and UC in remission was analyzed. The relationship between C-reactive protein level, Total Mayo score, Mayo Endoscopic score used to predict disease activity, and amount of SuPAR were evaluated. RESULTS SuPAR levels were determined in the UC patient group (2170,3 ± 121,0 pg/ml), and healthy controls (2130,7 ± 164,8 pg/ml) (p = 0. 805). Median SuPAR levels were determined in moderate UC (2479 pg/ml), mild UC (1944 pg/ml), and patients in remission (1774 pg/ml) (p = 0,207). There were no significant relationships between SuPAR levels and CRP levels, Total Mayo score, disease duration in the UC group (r = 0.177, r = 0.267, and r = 0,007; respectively p > 0.05). A slightly positive correlation was found between Mayo Endoscopic Score and SuPAR level (r = 0.303; p = 0.031). CONCLUSION SuPAR is of limited value in the diagnosis of ulcerative colitis and in the assessment of disease activation.
Collapse
Affiliation(s)
- Huriye Erbak Yılmaz
- Izmir Katip Çelebi University Atatürk Training and Research Hospital, Biochemistry Laboratory Izmir, Turkey; Izmir Biomedicine and Genom Center, Izmir, Turkey.
| | - Saliha Aksun
- Izmir Katip Çelebi University, Department of Biochemistry, Izmir, Turkey
| | - Süleyman Günay
- Izmir Katip Çelebi University, Department of Gastroenterology, Izmir, Turkey
| | - Ferhan Elmalı
- Izmir Katip Çelebi University, Department of Biostatistics, Izmir, Turkey
| | - Cem Çekiç
- Izmir Tinaztepe University, Department of Gastroenterology, Izmir, Turkey
| |
Collapse
|
11
|
Manfredi M, Van Hoovels L, Benucci M, De Luca R, Coccia C, Bernardini P, Russo E, Amedei A, Guiducci S, Grossi V, Bossuyt X, Perricone C, Infantino M. Soluble Urokinase Plasminogen Activator Receptor (suPAR) in Autoimmune Rheumatic and Non Rheumatic Diseases. J Pers Med 2023; 13:688. [PMID: 37109074 PMCID: PMC10140982 DOI: 10.3390/jpm13040688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The soluble urokinase plasminogen activator receptor (suPAR) is the bioactive form of uPAR, a membrane-bound glycoprotein, and it is primarily expressed on the surface of immunologically active cells. Mirroring local inflammation and immune activation, suPAR has gained interest as a potential prognostic biomarker in several inflammatory diseases. Indeed, in many diseases, including cancer, diabetes, cardiovascular diseases, kidney diseases, and inflammatory disorders, higher suPAR concentrations have been associated with disease severity, disease relapse, and mortality. Our review describes and discusses the supporting literature concerning the promising role of suPAR as a biomarker in different autoimmune rheumatic and non-rheumatic diseases.
Collapse
Affiliation(s)
- Mariangela Manfredi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy; (M.M.)
| | - Lieve Van Hoovels
- Department of Microbiology, Immunology and Transplantation, University of Leuven, 3000 Leuven, Belgium
- Department of Laboratory Medicine, OLV Hospital, 9300 Aalst, Belgium
| | - Maurizio Benucci
- Rheumatology Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy
| | - Riccardo De Luca
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Carmela Coccia
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Pamela Bernardini
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Serena Guiducci
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Valentina Grossi
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy; (M.M.)
| | - Xavier Bossuyt
- Department of Microbiology, Immunology and Transplantation, University of Leuven, 3000 Leuven, Belgium
- Department of Laboratory Medicine, University Hospital Leuven, 3000 Leuven, Belgium
| | - Carlo Perricone
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, 06122 Perugia, Italy
| | - Maria Infantino
- Immunology and Allergology Laboratory Unit, S. Giovanni di Dio Hospital, 50143 Florence, Italy; (M.M.)
| |
Collapse
|
12
|
Zhu K, Reiser J. XOR risk variants drive diabetic kidney disease. Nat Metab 2023; 5:536-537. [PMID: 37024753 DOI: 10.1038/s42255-023-00768-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Affiliation(s)
- Ke Zhu
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
13
|
Association between Serum Soluble Urokinase-Type Plasminogen Activator Receptor Level and Arterial Stiffness in Chronic Hemodialysis Patients. J Pers Med 2023; 13:jpm13030470. [PMID: 36983652 PMCID: PMC10051056 DOI: 10.3390/jpm13030470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Cardiovascular diseases (CVDs) remain a significant cause of death in hemodialysis (HD) patients. To explore their associations, we examine the role of soluble urokinase-type plasminogen activator receptor (suPAR) in arterial stiffness in chronic HD patients. From June to August 2020, we recruited 135 chronic HD patients. The arterial stiffness group included patients with a carotid–femoral pulse-wave velocity (cfPWV) of >10 m/s. Fifty-five HD patients (40.7%) were in the arterial stiffness group. They had a higher prevalence of diabetes (p = 0.001) and hypertension (p = 0.039), were older (p = 0.007) and had higher aortic systolic blood pressure (p = 0.034), brachial systolic blood pressure (p = 0.025), glucose (p = 0.019), C-reactive protein (p = 0.039), and AIx75 (p = 0.003) and suPAR (p < 0.001) levels than the control group. After we performed multivariable logistic regression analysis, except age and glucose, serum suPAR (odds ratio [OR]: 2.05; 95% confidence interval [CI]: 1.48–2.70, p < 0.001) was independently associated with arterial stiffness in chronic HD patients. In the multivariable linear regression analysis, suPAR positively correlated with cfPWV (β = 0.475, p < 0.001) and could serve as a biomarker for arterial stiffness development in patients undergoing HD.
Collapse
|
14
|
Pelle MC, Provenzano M, Busutti M, Porcu CV, Zaffina I, Stanga L, Arturi F. Up-Date on Diabetic Nephropathy. Life (Basel) 2022; 12:1202. [PMID: 36013381 PMCID: PMC9409996 DOI: 10.3390/life12081202] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 12/11/2022] Open
Abstract
Diabetes is one of the leading causes of kidney disease. Diabetic kidney disease (DKD) is a major cause of end-stage kidney disease (ESKD) worldwide, and it is linked to an increase in cardiovascular (CV) risk. Diabetic nephropathy (DN) increases morbidity and mortality among people living with diabetes. Risk factors for DN are chronic hyperglycemia and high blood pressure; the renin-angiotensin-aldosterone system blockade improves glomerular function and CV risk in these patients. Recently, new antidiabetic drugs, including sodium-glucose transport protein 2 inhibitors and glucagon-like peptide-1 agonists, have demonstrated additional contribution in delaying the progression of kidney disease and enhancing CV outcomes. The therapeutic goal is regression of albuminuria, but an atypical form of non-proteinuric diabetic nephropathy (NP-DN) is also described. In this review, we provide a state-of-the-art evaluation of current treatment strategies and promising emerging treatments.
Collapse
Affiliation(s)
- Maria Chiara Pelle
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Provenzano
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS—Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Marco Busutti
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS—Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Clara Valentina Porcu
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS—Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Isabella Zaffina
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Lucia Stanga
- Oncology Unit, IRCCS—Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Franco Arturi
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Research Centre for the Prevention and Treatment of Metabolic Diseases (CR METDIS), University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
15
|
Hyperglycemia Promotes Endothelial Cell Senescence through AQR/PLAU Signaling Axis. Int J Mol Sci 2022; 23:ijms23052879. [PMID: 35270021 PMCID: PMC8911151 DOI: 10.3390/ijms23052879] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/23/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023] Open
Abstract
Hyperglycemia is reported to accelerate endothelial cell senescence that contributes to diabetic complications. The underlying mechanism, however, remains elusive. We previously demonstrated AQR as a susceptibility gene for type 2 diabetes mellitus (T2DM) and showed that it was increased in multiple tissues in models with T2DM or metabolic syndrome. This study aimed to investigate the role of AQR in hyperglycemia-induced senescence and its underlying mechanism. Here, we retrieved several datasets of the aging models and found the expression of AQR was increased by high glucose and by aging across species, including C. elegans (whole-body), rat (cardiac tissues), and monkey (blood). we validated the increased AQR expression in senescent human umbilical vein endothelial cells (HUVECs). When overexpressed, AQR promoted the endothelial cell senescence, confirmed by an increased number of cells stained with senescence-associated beta-galactosidase and upregulation of CDKN1A (P21) as well as the prohibited cellular colony formation and G2/M phase arrest. To explore the mechanism by which AQR regulated the cellular senescence, transcriptomic analyses of HUVECs with the overexpression and knockdown of the AQR were performed. We identified 52 co-expressed genes that were enriched, in the terms of plasminogen activation, innate immunity, immunity, and antiviral defense. Among co-expressed genes, PLAU was selected to evaluate its contribution to senescence for its highest strength in the enrichment of the biological process. We demonstrated that the knockdown of PLAU rescued senescence-related phenotypes, endothelial cell activation, and inflammation in models induced by AQR or TNF-α. These findings, for the first time, indicate that AQR/PLAU is a critical signaling axis in the modulation of endothelial cell senescence, revealing a novel link between hyperglycemia and vascular dysfunction. The study may have implications in the prevention of premature vascular aging associated with T2DM.
Collapse
|
16
|
Manshad AS, Ballout FA, Borgia JA, Reiser J, Okwuosa TM. Soluble Urokinase Plasminogen Activator Receptor Is Associated With Subclinical Myocardial Impairment by Speckle Tracking Echocardiography in Lung Cancer Patients. Front Cardiovasc Med 2022; 8:659524. [PMID: 35155590 PMCID: PMC8831744 DOI: 10.3389/fcvm.2021.659524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022] Open
Abstract
Background Plasma cardiac biomarkers have emerged as a cost-effective diagnostic tool aimed at early identification of cardiotoxicity. Soluble urokinase plasminogen activator receptor (suPAR) is a bone marrow cell derived signaling molecule that is associated with cardiovascular disease outcomes. Objectives We investigated associations between suPAR and global longitudinal strain (GLS) as a marker of early myocardial impairment in lung cancer patients. Methods We retrospectively analyzed 52 patients with stage IV non-small cell lung cancer with normal left ventricular ejection fraction (LVEF >55%) and without known heart disease or end-stage renal disease (ESRD). We studied associations between cardiac biomarkers and echocardiographic measures of systolic and diastolic function. GLS was analyzed using 2D speckle-tracking echocardiography via vendor-independent software (TomTec). Results Median plasma suPAR was 7.0 ng/mL (interquartile range: 5.4–9.0). Mean LVEF was 61.9 ± 8.3% and mean GLS was-19.3 ± 2.1%. Inter-observer reproducibility was excellent for GLS as determined by Intraclass Correlation Coefficient analysis, ICC = 0.81 (0.68–0.89). After multivariate analysis, suPAR was the only biomarker associated with GLS (p = 0.009). suPAR was also associated with diastolic parameters E velocity (p = 0.018), A velocity (p = 0.017), and E/E' ratio (p = 0.033). Interestingly, suPAR was not associated with LVEF (p = 0.916). In addition, suPAR and GLS were found to be age-independent predictors of all-cause mortality, though only GLS remained significant after multivariate adjustment. Conclusions In this cohort of stage IV non-small cell lung cancer patients with normal LVEF and without known heart disease or ESRD, suPAR was associated with GLS and diastolic impairment. suPAR is a readily available inexpensive biomarker; further research is required to evaluate the possible role of suPAR in screening for subclinical LV dysfunction in the high-risk oncological population.
Collapse
Affiliation(s)
- Ahmad S. Manshad
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Fatima A. Ballout
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Jeffrey A. Borgia
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
- Department of Pathology, Rush University Medical Center, Chicago, IL, United States
| | - Jochen Reiser
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Tochukwu M. Okwuosa
- Division of Cardiology, Rush University Medical Center, Chicago, IL, United States
- *Correspondence: Tochukwu M. Okwuosa
| |
Collapse
|
17
|
Rasmussen LJH, Petersen JEV, Eugen-Olsen J. Soluble Urokinase Plasminogen Activator Receptor (suPAR) as a Biomarker of Systemic Chronic Inflammation. Front Immunol 2021; 12:780641. [PMID: 34925360 PMCID: PMC8674945 DOI: 10.3389/fimmu.2021.780641] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/09/2021] [Indexed: 01/08/2023] Open
Abstract
Systemic chronic inflammation (SCI) is persistent, health-damaging, low-grade inflammation that plays a major role in immunosenescence and in development and progression of many diseases. But currently, there are no recognized standard biomarkers to assess SCI levels alone, and SCI is typically measured by combining biomarkers of acute inflammation and infection, e.g., CRP, IL-6, and TNFα. In this review, we highlight 10 properties and characteristics that are shared by the blood protein soluble urokinase plasminogen activator receptor (suPAR) and SCI, supporting the argument that suPAR is a biomarker of SCI: (1) Expression and release of suPAR is upregulated by immune activation; (2) uPAR and suPAR exert pro-inflammatory functions; (3) suPAR is associated with the amount of circulating immune cells; (4) Blood suPAR levels correlate with the levels of established inflammatory biomarkers; (5) suPAR is minimally affected by acute changes and short-term influences, in contrast to many currently used markers of systemic inflammation; (6) Like SCI, suPAR is non-specifically associated with multiple diseases; (7) suPAR and SCI both predict morbidity and mortality; (8) suPAR and SCI share the same risk factors; (9) suPAR is associated with risk factors and outcomes of inflammation above and beyond other inflammatory biomarkers; (10) The suPAR level can be reduced by anti-inflammatory interventions and treatment of disease. Assessing SCI has the potential to inform risk for morbidity and mortality. Blood suPAR is a newer biomarker which may, in fact, be a biomarker of SCI since it is stably associated with inflammation and immune activation; shares the same risk factors as many age-related diseases; is both elevated by and predicts age-related diseases. There is strong evidence that suPAR is a prognostic marker of adverse events, morbidity, and mortality. It is associated with immune activity and prognosis across diverse conditions, including kidney disease, cardiovascular disease, cancer, diabetes, and inflammatory disorders. Thus, we think it likely represents a common underlying disease-process shared by many diseases; that is, SCI. We review the supporting literature and propose a research agenda that can help test the hypothesis that suPAR indexes SCI, with the potential of becoming the new gold standard for measuring SCI.
Collapse
Affiliation(s)
- Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| | - Jens Emil Vang Petersen
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, United States
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| |
Collapse
|
18
|
Lupușoru G, Ailincăi I, Sorohan BM, Andronesi A, Achim C, Micu G, Caragheorgheopol A, Manda D, Lupușoru M, Ismail G. Serum soluble urokinase plasminogen activator receptor as a potential biomarker of renal impairment severity in diabetic nephropathy. Diabetes Res Clin Pract 2021; 182:109116. [PMID: 34728182 DOI: 10.1016/j.diabres.2021.109116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 12/28/2022]
Abstract
AIMS To investigate serum soluble form of urokinase-type plasminogen activator receptor (suPAR) in patients with diabetic kidney disease (DKD) and biopsy-proven diabetic nephropathy (DN), its correlation with histological parameters and its capacity as a biomarker for renal impairment severity. METHODS We conducted a cross-sectional study on 75 patients with diabetes mellitus (DM) and DKD, among whom 28 had biopsy-proven DN. RESULTS Among the 75 patients, 9 (12%) had type 1 and 66 (88%) type 2 DM. The median value of the serum suPAR level was 2857.2 pg/mL (1916.4-3700) in the entire cohort and 2472.1 pg/mL (1782.6-3745.8) in the biopsy-proven DN subgroup, respectively. suPAR was significantly correlated with diabetes duration, diabetic retinopathy, anti-proteinuric treatment, albuminuria, kidney function, DN class, interstitial fibrosis and tubular atrophy (IFTA) score and with interstitial inflammation score. suPAR had a good accuracy for the association with chronic kidney disease (CKD) stages G3b-5, macroalbuminuria, DN class IV, IFTA score 3 and interstitial inflammation score 2. CONCLUSIONS Serum suPAR was increased in DN patients and was associated with DM duration, diabetic retinopathy, renoprotective treatment, kidney function, proteinuria, DN class, IFTA and interstitial inflammation scores. Also, suPAR had a good capacity as a biomarker for advanced renal impairment and severe histological lesions of DN.
Collapse
Affiliation(s)
- Gabriela Lupușoru
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Ioana Ailincăi
- Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Bogdan Marian Sorohan
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania.
| | - Andreea Andronesi
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Camelia Achim
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Georgia Micu
- Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| | - Andra Caragheorgheopol
- Research Department, "C.I.Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Dana Manda
- Research Department, "C.I.Parhon" National Institute of Endocrinology, Bucharest, Romania
| | - Mircea Lupușoru
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Gener Ismail
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania; Department of Nephrology, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
19
|
Qin Y, Qiao Y, Wang D, Yan G, Tang C, Ma G. The Predictive Value of Soluble Urokinase-Type Plasminogen Activator Receptor in Contrast-Induced Acute Kidney Injury in Patients Undergoing Percutaneous Coronary Intervention. Int J Gen Med 2021; 14:6497-6504. [PMID: 34675617 PMCID: PMC8504866 DOI: 10.2147/ijgm.s339075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/01/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Soluble urokinase-type plasminogen activator receptor (SuPAR) is a circulating protein and a novel identified promising biomarker for various renal diseases and kidney injury. However, it remains unknown on the predictive value of suPAR in contrast induced acute kidney injury (CI-AKI) in patients undergoing percutaneous coronary intervention (PCI). Methods A total of 399 patients undergoing PCI were enrolled in the research from June 2020 to June 2021 in Zhongda Hospital. Patients were divided into CI-AKI and non-CI-AKI groups according to the preoperative and postoperative serum creatinine levels. Plasma suPAR level was detected through enzyme-linked immunosorbent assay on admission. Demographic data, hematological parameters, coronary angiography data and medications were recorded and compared between CI-AKI and non-CI-AKI groups. Logistic regression analysis and receiver operator characteristic (ROC) curve analysis were performed for identifying the independent risk factors of CI-AKI and assessment of the predictive value of suPAR for CI-AKI. Results CI-AKI occurred in 65 (16.3%) patients undergoing PCI. The level of suPAR in CI-AKI group was significantly higher than that in the non-CI-AKI group. Multivariate logistic regression indicated diabetes, lower LVEF, lower hydration rate, lower baseline eGFR, higher plasma suPAR (OR = 2.875, 95% CI = 2.038–3.672, P < 0.001) and volume of contrast media were all independent risk factors for CI-AKI after adjustment of the confounding factors. ROC analysis illustrated that the optimal area under the curve was 0.765, indicating plasma suPAR was a splendid predictor for CI-AKI. The corresponding cut-off value was 3.305 ng/mL, and the sensitivity and specificity were 63.1% and 82.3%, respectively. Conclusion Increased suPAR level is independently associated with elevated risk of suffering CI-AKI, and suPAR is a strong predictor for CI-AKI in patients undergoing PCI. SuPAR might act as a novel biomarker for CI-AKI in clinical practice.
Collapse
Affiliation(s)
- Yuhan Qin
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
20
|
Kerget B, Kerget F, Aksakal A, Aşkın S, Uçar EY, Sağlam L. Evaluation of the relationship between KIM-1 and suPAR levels and clinical severity in COVID-19 patients: A different perspective on suPAR. J Med Virol 2021; 93:5568-5573. [PMID: 34019703 PMCID: PMC8242801 DOI: 10.1002/jmv.27099] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 12/21/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is one of the most pressing health problems of this century, but our knowledge of the disease is still limited. In this study, we aimed to examine serum-soluble urokinase plasminogen activator receptor (suPAR) and kidney injury molecule 1 (KIM-1) levels based on the clinical course of COVID-19. Our study included 102 patients over the age of 18 who were diagnosed as having COVID-19 between September 2020 and December 2020 and a control group of 50 health workers over the age of 18 whose severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PCR results were negative. KIM-1 was measured by ELISA and suPAR by suPARnostic™ assay. Analysis of previously identified variables of prognostic significance in COVID-19 revealed high neutrophil to lymphocyte ratio, lactose dehydrogenase, prothrombin time, C-reactive protein, PaO2 /FiO2 , D-dimer, ferritin, and fibrinogen levels in patients with severe disease (p < 0.05 for all). KIM-1 and suPAR levels were significantly higher in COVID-19 patients compared to the control group (p = 0.001 for all). KIM-1 level was higher in severe patients compared to moderate patients (p = 0.001), while suPAR level was lower (p = 0.001). KIM-1, which is believed to play an important role in the endocytosis of SARS-CoV-2, was elevated in patients with severe COVID-19 and may be a therapeutic target in the future. SuPAR may have a role in defense mechanism and fibrinolysis, and low levels in severe patients may be associated with poor prognosis in the early period.
Collapse
Affiliation(s)
- Buğra Kerget
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Ferhan Kerget
- Department of Infection Diseases and Clinical Microbiology, Health Sciences University Erzurum Regional Education and Research Hospital, Yakutiye, Erzurum, Turkey
| | - Alperen Aksakal
- Department of Pulmonary Diseases, Health Sciences University Erzurum Regional Education and Research Hospital, Yakutiye, Erzurum, Turkey
| | - Seda Aşkın
- Department of Biochemistry, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Elif Yılmazel Uçar
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| | - Leyla Sağlam
- Department of Pulmonary Diseases, Ataturk University School of Medicine, Yakutiye, Erzurum, Turkey
| |
Collapse
|
21
|
Höbaus C, Ursli M, Yussef SM, Wrba T, Koppensteiner R, Schernthaner GH. Soluble urokinase-type plasminogen activator receptor predicts peripheral artery disease severity and outcomes. Vasc Med 2021; 26:11-17. [PMID: 33448911 PMCID: PMC7879231 DOI: 10.1177/1358863x20982077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Soluble urokinase-type plasminogen activator receptor (suPAR) is associated with chronic kidney disease (CKD) severity and peripheral artery disease (PAD). We hypothesize an association of PAD severity and suPAR in patients without advanced CKD and further risk stratification according to the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines. For study purposes, suPAR was measured in 334 PAD patients (34% women, age 69 (62-78) years, eGFR 68 ± 20 mL/min/1.72 m2) by commercial ELISA. Patients were followed for 10 years to assess long-term all-cause survival by Cox regression. Higher suPAR levels were associated with lower ankle-brachial index (R = -0.215, p = 0.001) in patients with PAD without media-sclerosis (n = 236). suPAR levels inversely correlated with decreased glomerular filtration rate (R = -0.476, p < 0.001) and directly correlated with urinary albumin-to-creatinine ratio (R = 0.207, p < 0.001). Furthermore, higher suPAR levels associated with a higher KDIGO risk score (p < 0.001). Baseline suPAR was significantly associated with all-cause mortality (HR 1.40 (95% CI 1.16-1.68), p < 0.001) over 10 years. suPAR remained associated with mortality (HR 1.29 (1.03-1.61), p = 0.026) after multivariable adjustment for age, sex, cardiovascular risk factors, and eGFR. Future research may define a standard role for suPAR assessment in PAD's work-up and treatment, especially in patients with CKD.
Collapse
Affiliation(s)
- Clemens Höbaus
- Division of Angiology, Medicine II, Medical University Vienna, Vienna, Austria
| | - Martin Ursli
- Department of Internal Medicine I, University Hospital of St Poelten, Karl Landsteiner University of Health Sciences, Karl Landsteiner Institute for Nephrology and Hematooncology, St Poelten, Austria
| | | | - Thomas Wrba
- IT4Science, IT-Systems & Communications, Medical University Vienna, Vienna, Austria
| | | | | |
Collapse
|
22
|
Jhee JH, Nam BY, Lee CJ, Park JT, Han SH, Kang SW, Park S, Yoo TH. Soluble Urokinase-Type Plasminogen Activator Receptor, Changes of 24-Hour Blood Pressure, and Progression of Chronic Kidney Disease. J Am Heart Assoc 2020; 10:e017225. [PMID: 33325248 PMCID: PMC7955457 DOI: 10.1161/jaha.120.017225] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Soluble urokinase‐type plasminogen activator receptor (suPAR) is associated with cardiovascular risks and poor renal outcomes. However, whether elevated suPAR levels are associated with 24‐hour blood pressure patterns or kidney disease progression in patients with chronic kidney disease (CKD) is unclear. Methods and Results A total of 751 patients with CKD stage 1 to 5 were recruited from CMERC‐HI (Cardiovascular and Metabolic Disease Etiology Research Center–High Risk) cohort study (2013–2018). The relationship of serum suPAR levels to 24‐hour blood pressure parameters and CKD progression was analyzed. The median serum suPAR level was 1439.0 (interquartile range, 1026.2–2150.1) pg/mL, and the mean estimated glomerular filtration rate was 52.8±28.5 mL/min per 1.73 m2 at baseline. Patients with higher suPAR levels had significantly higher levels of office, 24‐hour, daytime, and nighttime systolic blood pressure and nighttime diastolic blood pressure than those with lower suPAR levels. The highest suPAR tertile was associated with an increased risk of a reverse dipping pattern (odds ratio, 2.93; 95% CI, 1.27–6.76; P=0.01). During a follow‐up of 43.2 (interquartile range, 27.0–55.6) months, the CKD progression occurred in 271 (36.1%) patients. The highest suPAR tertile was significantly associated with higher risk of CKD progression than the lowest tertile (hazard ratio [HR], 2.09; 95% CI, 1.37–3.21; P=0.001). When the relationship was reevaluated with respect to each dipping pattern (dipper, extreme dipper, nondipper, and reverse dipper), this association was consistent only in reverse dippers in whom the risk of CKD progression increased (HR, 1.43; 95% CI, 1.02–2.01; P=0.03) with every 1‐unit increase in serum suPAR levels. Conclusions Elevated suPAR levels are independently associated with CKD progression, and this association is prominent in reverse dippers.
Collapse
Affiliation(s)
- Jong Hyun Jhee
- Division of Nephrology Department of Internal Medicine Gangnam Severance HospitalYonsei University College of Medicine Seoul Korea
| | - Bo Young Nam
- Department of Internal Medicine College of Medicine Severance Biomedical Science Institute Brain Korea 21 PLUS Yonsei University Seoul Korea
| | - Chan Joo Lee
- Division of Cardiology Severance Cardiovascular Hospital and Severance Cardiovascular Hospital and Integrated Research Center for Cerebrovascular and Cardiovascular Diseases Yonsei University College of Medicine Seoul Korea
| | - Jung Tak Park
- Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Seung Hyeok Han
- Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Shin-Wook Kang
- Department of Internal Medicine College of Medicine Severance Biomedical Science Institute Brain Korea 21 PLUS Yonsei University Seoul Korea.,Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| | - Sungha Park
- Division of Cardiology Severance Cardiovascular Hospital and Severance Cardiovascular Hospital and Integrated Research Center for Cerebrovascular and Cardiovascular Diseases Yonsei University College of Medicine Seoul Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine College of Medicine Institute of Kidney Disease Research Yonsei University Seoul Korea
| |
Collapse
|
23
|
Abstract
The traditional chronic kidney disease (CKD) biomarkers (eGFR based on serum creatinine, sex and age and albuminuria) cannot predict a patient's individual risk for developing progressive CKD. For this reason, it is necessary to identify novel CKD biomarkers that will be able to predict which patients are prone to develop progressive disease and discriminate between disease processes in different parts of the nephron (glomeruli or tubules). A good biomarker should change before or simultaneously with lesion development and its changes should correlate strongly with lesion development. Also, there should be a close relationship between severity of injury and amount of detectable biomarker and its levels should decrease with diminishing injury. Among the large number of molecules under investigation, we have reviewed the most promising ones: NGAL and KIM-1, MCP-1, MMP-9, clusterin, MMP-9, TIMP-1, Procollagen I alpha 1 and suPAR. All these, have been studied as biomarkers for prediction of CKD progression in cohorts of patients with chronic kidney disease of different stages and various aetiologies (proteinuric and non-proteinuric, glomerulonephritides, diabetic, hypertensive and polycystic kidney disease). There is evidence that these molecules could be useful as biomarkers for progressive chronic kidney disease, however, the available data are not enough to draw final conclusions. Further studies with large cohorts and long follow-up are required to identify appropriate biomarkers, that will be able to accurately and reliably define the risk for progressive chronic kidney disease.
Collapse
|
24
|
Petersen JEV, Kallemose T, Barton KD, Caspi A, Rasmussen LJH. Soluble urokinase plasminogen activator receptor (suPAR) as a prognostic marker of mortality in healthy, general and patient populations: protocol for a systematic review and meta-analysis. BMJ Open 2020; 10:e036125. [PMID: 32690515 PMCID: PMC7371134 DOI: 10.1136/bmjopen-2019-036125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/11/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Chronic inflammation is increasingly recognised as a major contributor to disease, disability and ultimately death, but measuring the levels of chronic inflammation remains non-canonised, making it difficult to relate chronic inflammation and mortality. Soluble urokinase plasminogen activator receptor (suPAR), an emerging biomarker of chronic inflammation, has been proposed as a prognostic biomarker associated with future incidence of chronic disease and mortality in general as well as patient populations. Proper prognostic biomarkers are important as they can help improve risk stratification in clinical settings and provide guidance in treatment or lifestyle decisions as well as in the design of randomised trials. Here, we wish to summarise the evidence about the overall association of the biomarker suPAR with mortality in healthy, general and patient populations across diseases. METHODS AND ANALYSIS The search will be conducted using Medline, Embase and Scopus databases from their inception to 03 June 2020 to identify studies investigating 'suPAR' and 'mortality'. Observational studies and control groups from intervention studies written in English or Danish will be included. The 'Quality In Prognosis Studies' tool will be used to assess the risk of bias for the studies included. Unadjusted and adjusted mortality outcome measures (eg, risk ratios, ORs, HRs) with 95% CIs will be extracted for healthy individuals, general and patient populations. The primary outcome is all-cause mortality within any given follow-up. Subgroup analyses will be performed based on time of outcome, cause of death, population type, adjustments for conventional risk factors and inflammation markers. ETHICS AND DISSEMINATION This systematic review will synthesise evidence on the use of suPAR as a prognostic marker for mortality. The results will be disseminated by publication in a peer-reviewed journal. Data used will be obtained from published studies, and ethics approval is therefore not necessary for this systematic review. TRIAL REGISTRATION NUMBER PROSPERO CRD42020167401.
Collapse
Affiliation(s)
- Jens Emil Vang Petersen
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Karen D Barton
- Duke University Medical Center Library & Archives, Duke University, Durham, North Carolina, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| |
Collapse
|
25
|
D'Alonzo D, De Fenza M, Pavone V. COVID-19 and pneumonia: a role for the uPA/uPAR system. Drug Discov Today 2020; 25:1528-1534. [PMID: 32562843 PMCID: PMC7299864 DOI: 10.1016/j.drudis.2020.06.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/14/2020] [Accepted: 06/11/2020] [Indexed: 12/29/2022]
Abstract
Here, we highlight recent findings on the urokinase plasminogen activator (uPA)/uPA receptor (uPAR) system that suggest its potential role as a main orchestrator of fatal progression to pulmonary, kidney, and heart failure in patients with coronavirus. Patients with prolonged background inflammation can present aberrant inflammatory reactions, well recognized as the main factors that can result in death and probably sustained by a dysregulated uPA/uPAR system. SuPAR, the soluble form of uPAR, represents a biomarker of disease progression, and its levels correlate well with comorbidities associated with the death of patients with coronavirus. New drugs that regulate the uPA/uPAR system could help treat the severe complications of highly pathogenic human coronaviruses (hCoVs), including pandemic coronavirus 2019 (COVID-19).
Collapse
Affiliation(s)
- Daniele D'Alonzo
- University of Naples 'Federico II', Department of Chemical Sciences, Complesso Universitario di Monte Sant'Angelo, Via Cintia 46, 80126 Naples, Italy
| | - Maria De Fenza
- University of Naples 'Federico II', Department of Chemical Sciences, Complesso Universitario di Monte Sant'Angelo, Via Cintia 46, 80126 Naples, Italy
| | - Vincenzo Pavone
- University of Naples 'Federico II', Department of Chemical Sciences, Complesso Universitario di Monte Sant'Angelo, Via Cintia 46, 80126 Naples, Italy.
| |
Collapse
|
26
|
Skovsted TA, Petersen ERB, Fruekilde MB, Pedersen AK, Pielak T, Eugen-Olsen J. Validation of suPAR turbidimetric assay on Cobas® (c502 and c702) and comparison to suPAR ELISA. Scandinavian Journal of Clinical and Laboratory Investigation 2020; 80:327-335. [PMID: 32186407 DOI: 10.1080/00365513.2020.1741674] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
suPAR is a plasma marker of chronic inflammation, and an elevated suPAR is consistently associated with worse outcome in a variety of clinical conditions. Quantification of suPAR is useful for determining patient risk in triage, but there is no fast automatized method for quick determination of suPAR. We developed and validated a rapid latex particle-enhanced turbidimetric immunoassay for quantification of plasma suPAR on the c502 and the c702 Roche Cobas® 8000 measurment systems. The turbidimetric assay was validated against the suPARnostic® ELISA (ViroGates, Denmark). This validation demonstrates suPAR can be analysed by turbidimetry giving very similar results (<15% difference) compared to the ELISA method and the observed correlations (n = 103) were strong, r > 0.95. Roche Cobas® 8000 instruments demonstrated repeatability and repoducibility, CV % at 3.4-4.1 and 5.7-11.4, respectively. The estimated limit of detection was 1.30 µg/L and 1.31 µg/L for the Cobas® c502 and c702, respectively. Dilution tests showed linearity of suPAR from 1.8 to 26.5 μg/L. The acceptable concentrations of Bilirubin, Intralipid and Hemoglobin, were 350 µmol/L, 3.3 g/L and 1.4 g/L, respectively. suPAR can be quantified reproducibly within 10 min using a turbidimetry assay. This assay is faster than ELISA with similar results, making it suitable for clinical routine analysis.
Collapse
Affiliation(s)
- Thor A Skovsted
- Department of Biochemistry and Immunology, Hospital of Southern Jutland, Region of Southern Denmark, Aabenraa, Denmark
| | - Eva Rabing Brix Petersen
- Department of Biochemistry and Immunology, Hospital of Southern Jutland, Region of Southern Denmark, Aabenraa, Denmark
| | - Maj-Britt Fruekilde
- Department of Biochemistry and Immunology, Hospital of Southern Jutland, Region of Southern Denmark, Aabenraa, Denmark
| | | | - Tomasz Pielak
- ViroGates A/S, Birkerød, Denmark.,NUTOPI Sp. z o. o, Poznan, Poland
| | - Jesper Eugen-Olsen
- ViroGates A/S, Birkerød, Denmark.,Clinical Research Center, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| |
Collapse
|
27
|
Lv L, Wang F, Wu L, Wang JW, Cui Z, Hayek SS, Wei C, Reiser J, He K, Zhang L, Chen M, Zhao MH. Soluble urokinase-type plasminogen activator receptor and incident end-stage renal disease in Chinese patients with chronic kidney disease. Nephrol Dial Transplant 2020; 35:465-470. [PMID: 30124995 PMCID: PMC9214641 DOI: 10.1093/ndt/gfy265] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/28/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Soluble urokinase-type plasminogen activator receptor (suPAR), a marker of immune activation, was shown to be associated with outcomes and kidney disease among various patient populations. The prognostic role of circulating suPAR levels in patients with chronic kidney disease (CKD) needs to be investigated in a cohort with large sample size of renal diseases. METHODS We measured serum suPAR concentration in 2391 CKD patients in the multicenter Chinese Cohort Study of Chronic Kidney Disease, and investigated the association of serum suPAR with the prespecified endpoint event, end-stage renal disease (ESRD), using Cox proportional hazards regression model. RESULTS Altogether, 407 ESRD events occurred during the median follow-up of 54.8 (interquartile range: 47.5-62.2) months. The higher levels of serum suPAR were independently associated with increased risk of incident ESRD after adjusting for potential confounders including the baseline estimated glomerular filtration rate categories, with the hazard ratios (HRs) of 1.53 [95% confidence intervals (CIs) 1.10-2.12] for the top tertile (≥3904 pg/mL) compared with the bottom tertile (<2532 pg/mL). When stratified by the etiologies of CKD, among patients with glomerulonephritis (GN), serum suPAR levels were also independently associated with the higher risk of ESRD, with an HR of 1.61 (95% CI 1.03-2.53) in the top tertile compared with the bottom tertile. CONCLUSIONS Circulating suPAR level was independently associated with an increased risk of progression to ESRD in Chinese CKD patients, especially in those with an etiology of GN.
Collapse
Affiliation(s)
| | | | - Liang Wu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Jin-Wei Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Zhao Cui
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Salim S Hayek
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kevin He
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Luxia Zhang
- Correspondence and offprint requests to: Min Chen; E-mail: Luxia Zhang; E-mail:
| | - Min Chen
- Correspondence and offprint requests to: Min Chen; E-mail: Luxia Zhang; E-mail:
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Institute of Nephrology, Peking University, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
28
|
Rasmussen LJH, Moffitt TE, Arseneault L, Danese A, Eugen-Olsen J, Fisher HL, Harrington H, Houts R, Matthews T, Sugden K, Williams B, Caspi A. Association of Adverse Experiences and Exposure to Violence in Childhood and Adolescence With Inflammatory Burden in Young People. JAMA Pediatr 2020; 174:38-47. [PMID: 31682707 PMCID: PMC6830440 DOI: 10.1001/jamapediatrics.2019.3875] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
IMPORTANCE Childhood stress exposure is associated with inflammation as measured by C-reactive protein (CRP) and interleukin 6 (IL-6). However, findings are inconsistent and effect sizes are small. The addition of soluble urokinase plasminogen activator receptor (suPAR), a new biomarker of chronic inflammation, may improve measurement of stress-related inflammatory burden. OBJECTIVES To assess whether exposure to adverse experiences, stress, and violence is associated with an increase in suPAR levels in young people and to test the hypothesis that measuring suPAR in addition to CRP or IL-6 levels improves the assessment of the inflammatory burden associated with early-life stress. DESIGN, SETTING, AND PARTICIPANTS This cohort study included 1391 participants from a 1994 to 1995 birth cohort of twins from the nationally representative Environmental Risk Longitudinal Twin Study in the United Kingdom. Participants were followed up until 18 years of age (93% retention). Plasma samples were analyzed in July 2018, and statistical analysis was performed from October 1, 2018, to May 31, 2019. EXPOSURES Adverse childhood experiences and childhood and adolescent experience of stress and violence exposure. MAIN OUTCOMES AND MEASURES Plasma CRP, IL-6, and suPAR levels at 18 years of age. RESULTS Among 1391 young people (mean [SD] age, 18.4 [0.36] years; 733 [52.7%] female), those who had been exposed to stressful experiences had elevated suPAR levels by 18 years of age after controlling for sex, body mass index, and smoking: 0.03-ng/mL (95% CI, 0.01-0.05 ng/mL) increase in suPAR per each additional adverse childhood experience, 0.09-ng/mL (95% CI, 0.01-0.17 ng/mL) increase in suPAR per each additional severe childhood experience of stress or violence, and 0.04-ng/mL (95% CI, -0.02 to 0.10 ng/mL) increase in suPAR per each additional severe adolescent experience of stress or violence. Individuals exposed to multiple types of violence in both childhood and adolescence had 0.26-ng/mL (95% CI, 0.07-0.45 ng/mL) higher suPAR levels compared with children who did not experience stress or violence. These stress-exposed young people were significantly more likely to have elevated suPAR levels at 18 years of age even if they did not have elevated CRP or IL-6 levels. Measuring suPAR in addition to CRP or IL-6 increased the association between stress exposure and inflammatory burden. For example, after adjusting for CRP and IL-6 levels, each additional adverse childhood experience was associated with a 0.05-mL (95% CI, 0.03-0.07 ng/mL) increase in suPAR, each additional severe childhood experience of stress or violence was associated with a 0.14-ng/mL (95% CI, 0.06-0.22 ng/mL) increase in suPAR, and each additional severe adolescent experience of stress or violence was associated with a 0.10-ng/mL (95% CI, 0.04-0.16 ng/mL) increase in suPAR. CONCLUSIONS AND RELEVANCE The results suggest that adult inflammation is associated with childhood exposure to stress. Adding information about suPAR to traditional biomarkers of inflammation may improve the measurement of inflammatory burden associated with exposure to stress and violence.
Collapse
Affiliation(s)
- Line Jee Hartmann Rasmussen
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina,Clinical Research Centre, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Terrie E. Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina,Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Louise Arseneault
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Andrea Danese
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom,Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom,National and Specialist Child and Adolescent Mental Health Services Trauma, Anxiety, and Depression Clinic, South London and Maudsley National Health Service Foundation Trust, London, United Kingdom
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Helen L. Fisher
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - HonaLee Harrington
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Renate Houts
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Timothy Matthews
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Benjamin Williams
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, North Carolina,Center for Genomic and Computational Biology, Duke University, Durham, North Carolina,Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
29
|
Pérez-López L, Boronat M, Melián C, Brito-Casillas Y, Wägner AM. Animal Models and Renal Biomarkers of Diabetic Nephropathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1307:521-551. [PMID: 32329028 DOI: 10.1007/5584_2020_527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Diabetes mellitus (DM) is the first cause of end stage chronic kidney disease (CKD). Animal models of the disease can shed light on the pathogenesis of the diabetic nephropathy (DN) and novel and earlier biomarkers of the condition may help to improve diagnosis and prognosis. This review summarizes the most important features of animal models used in the study of DN and updates the most recent progress in biomarker research.
Collapse
Affiliation(s)
- Laura Pérez-López
- Institute of Biomedical and Health Research (IUIBS), University of Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Mauro Boronat
- Institute of Biomedical and Health Research (IUIBS), University of Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
- Department of Endocrinology and Nutrition, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - Carlos Melián
- Institute of Biomedical and Health Research (IUIBS), University of Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
- Department of Animal Pathology, Veterinary Faculty, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Arucas, Las Palmas, Spain
| | - Yeray Brito-Casillas
- Institute of Biomedical and Health Research (IUIBS), University of Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain
| | - Ana M Wägner
- Institute of Biomedical and Health Research (IUIBS), University of Las Palmas de Gran Canaria (ULPGC), Las Palmas de Gran Canaria, Spain.
- Department of Endocrinology and Nutrition, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain.
| |
Collapse
|
30
|
Zhou Y, Ren J, Li P, Ma R, Zhou M, Zhang N, Kong X, Hu Z, Xiao X. Expression of Urokinase-type Plasminogen Activator Receptor and its Soluble Form in Type 2 Diabetic Kidney Disease. Arch Med Res 2019; 50:249-256. [PMID: 31593848 DOI: 10.1016/j.arcmed.2019.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Urokinase-type plasminogen activator receptor (uPAR) and its soluble form (suPAR) are new injury biomarkers that have been recently suggested to play a vital role in renal diseases. AIM AND METHODS We evaluated the expression of uPAR and the serum concentration of suPAR in type 2 diabetes (T2DM) patients with diabetic kidney disease (DKD) to determine the role of this molecule as a biomarker in DKD. The uPAR immunohistochemical staining was performed in biopsy-confirmed DKD renal tissues. Meanwhile, the serum suPAR, Interleukin-18 (IL-18) and C-reactive protein (CRP) levels of 70 diabetic patients with or without DKD and 15 healthy controls were measured. RESULTS The uPAR expression in DKD patients was significantly increased compared to that in healthy controls and was widely colocalized with the podocyte marker WT1. Meanwhile, serum suPAR and IL-18 levels gradually increased as DKD progressed to the advanced stage. Moreover, serum suPAR and IL-18 levels were negatively correlated with eGFR (ρ = ‒0.734, ρ = ‒0.462, p <0.01) and positively correlated with the urine protein to creatinine ratio (UP/CR) (ρ = 0.730, ρ = 0.440, p <0.01). The suPAR AUC performed better than the IL-18 AUC for the diagnosis of proteinuria (0.845 vs. 0.753, p <0.01) and the decline of renal function (0.895 vs. 0.796, p <0.01). CONCLUSIONS The uPAR expressed in the renal tissues of DKD patients. The soluble form of uPAR, suPAR, can be detected in the serum of DKD patients and has a better diagnostic efficiency in the diagnosis of proteinuria and renal dysfunction in patients with T2DM than that of IL-18.
Collapse
Affiliation(s)
- Yujing Zhou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, China; Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine and Health, Jinan, China; Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianmin Ren
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, China; Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine and Health, Jinan, China
| | - Peng Li
- Department of Nephrology, Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Rong Ma
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, China; Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine and Health, Jinan, China
| | - Mengkun Zhou
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, China
| | - Ningxin Zhang
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, China; Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine and Health, Jinan, China
| | - Xiangguo Kong
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China; Institute of Endocrinology and Metabolism, Shandong University, Jinan, China; Key Laboratory of Endocrinology and Metabolism, Shandong Province in Medicine and Health, Jinan, China
| | - Zhao Hu
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyan Xiao
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
31
|
Sherif EM, El Maksood AAA, Youssef OI, Salah El-Din NY, Khater OKM. Soluble urokinase plasminogen activator receptor in type 1 diabetic children, relation to vascular complications. J Diabetes Complications 2019; 33:628-633. [PMID: 31301955 DOI: 10.1016/j.jdiacomp.2019.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/12/2019] [Accepted: 06/02/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Endothelial dysfunction caused by chronic inflammation is the cornerstone of vascular complications in type 1 Diabetes-Mellitus (T1DM). Soluble Urokinase Plasminogen Activator Receptor (SuPAR) is a novel marker of inflammation and endothelial dysfunction. AIM To evaluate SuPAR in T1DM children and correlate it to diabetic vascular complications. METHODS Seventy T1DM children and 40 matched healthy controls were studied focusing on disease duration, insulin therapy and symptoms of diabetic complications. Blood-pressure, fundus and screening for peripheral-neuropathy were done. Fasting lipid profile, fraction-C of glycosylated hemoglobin (HbA1c%), Urinary albumin excretion (UAE), estimated-glomerular filtration rate (eGFR) and SuPAR were measured. Internal aortic diameter was measured with calculation of aortic distensibility and stiffness index. RESULTS Sixteen T1DM patients(22.9%) had peripheral neuropathy, 12(17%) had nephropathy and none had retinopathy. SuPAR was significantly elevated in diabetic nephropathy (p < 0.01) and neuropathy (p < 0.01). Aortic stiffness index was significantly higher (p < 0.01) whereas, aortic strain and distensibility were significantly lower (p < 0.01) in T1DM than controls. SuPAR was significantly correlated to disease duration (p < 0.01), systolic blood pressure (p < 0.01), total cholesterol (p < 0.01), triglycerides (p < 0.01), UAER (p < 0.01) and aortic strain (0.013). CONCLUSION Increased SuPAR early in diabetes might become a useful indicator of developing vascular complications. Further prospective studies are needed to determine the cut-off level of SuPAR for detection of T1DM and its complications.
Collapse
Affiliation(s)
- Eman Mounir Sherif
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | | | | |
Collapse
|
32
|
Rotbain Curovic V, Theilade S, Winther SA, Tofte N, Eugen-Olsen J, Persson F, Hansen TW, Jeppesen J, Rossing P. Soluble Urokinase Plasminogen Activator Receptor Predicts Cardiovascular Events, Kidney Function Decline, and Mortality in Patients With Type 1 Diabetes. Diabetes Care 2019; 42:1112-1119. [PMID: 30885954 DOI: 10.2337/dc18-1427] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 02/25/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Soluble urokinase plasminogen activator receptor (suPAR) is an important inflammatory biomarker implicated in endothelial and podocyte dysfunction. However, suPAR's predictive qualities for complications in type 1 diabetes have yet to be determined. We investigated the prognostic value of suPAR for the development of cardiovascular events, decline in renal function, and mortality in patients with type 1 diabetes. RESEARCH DESIGN AND METHODS We included 667 patients with type 1 diabetes with various degrees of albuminuria in a prospective study. End points were cardiovascular events (cardiovascular death, nonfatal acute myocardial infarction, nonfatal stroke, or coronary or peripheral arterial interventions), estimated glomerular filtration rate (eGFR) decline ≥30%, progression from lower to higher albuminuric state, development of end-stage renal disease (ESRD), and mortality. Follow-up was 5.2-6.2 years. Results were adjusted for known risk factors. Hazard ratios (HRs) are presented per doubling of suPAR with 95% CI. Relative integrated discrimination improvement (rIDI) was calculated. RESULTS Quantification of suPAR was available in all participants; median (interquartile range) was 3.4 ng/mL (2.7-4.5). The adjusted HR (95% CI) for cardiovascular events (n = 94), progression in albuminuria (n = 36), eGFR decline (n = 93), ESRD (n = 23), and mortality (n = 58) were 3.13 (1.96-5.45, P < 0.001), 1.27 (0.51-3.19, P = 0.61), 2.93 (1.68-5.11, P < 0.001), 2.82 (0.73-11.9, P = 0.13), and 4.13 (1.96-8.69, P < 0.001), respectively. rIDI was significant for cardiovascular events (22.6%, P < 0.001), eGFR decline (14.4%, P < 0.001), and mortality (23.9%, P < 0.001). CONCLUSIONS In patients with type 1 diabetes and a broad range of albuminuria, a higher level of suPAR is a significant and independent risk factor for cardiovascular events, decline in eGFR ≥30%, and mortality. In addition, suPAR contributes significantly to discrimination for the end points.
Collapse
Affiliation(s)
| | | | | | - Nete Tofte
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | | | | | - Jørgen Jeppesen
- Department of Medicine, Amager Hvidovre Hospital, Glostrup, Denmark.,University of Copenhagen, Copenhagen, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Early Discharge from the Emergency Department Based on Soluble Urokinase Plasminogen Activator Receptor (suPAR) Levels: A TRIAGE III Substudy. DISEASE MARKERS 2019; 2019:3403549. [PMID: 31236143 PMCID: PMC6545801 DOI: 10.1155/2019/3403549] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/21/2019] [Accepted: 04/17/2019] [Indexed: 12/20/2022]
Abstract
Objective Using biomarkers for early and accurate identification of patients at low risk of serious illness may improve the flow in the emergency department (ED) by classifying these patients as nonurgent or even suitable for discharge. A potential biomarker for this purpose is soluble urokinase plasminogen activator receptor (suPAR). We hypothesized that availability of suPAR might lead to a higher proportion of early discharges. Design A substudy of the interventional TRIAGE III trial, comparing patients with a valid suPAR measurement at admission to those without. The primary endpoint was the proportion of patients discharged alive from the ED within 24 hours. Secondary outcomes were length of hospital stay, readmissions, and mortality within 30 days. Setting EDs at two university hospitals in the Capital Region of Denmark. Participants 16,801 acutely admitted patients were included. Measurements and Main Results The suPAR level was available in 7,905 patients (suPAR group), but not in 8,896 (control group). The proportion of patients who were discharged within 24 hours of admittance was significantly higher in the suPAR group compared to the control group (50.2% (3,966 patients) vs. 48.6% (4,317 patients), P = 0.04). Furthermore, the mean length of hospital stay in the suPAR group was significantly shorter compared to that in the control group (4.3 days (SD 7.4) vs. 4.6 days (SD 9.4), P = 0.04). In contrast, the readmission rate within 30 days was significantly higher in the suPAR group (10.6% (839 patients) vs. 8.8% (785 patients), P < 0.001). Among patients discharged within 24 hours, there was no significant difference in the readmission rate or mortality within 30 days. Readmission occurred in 8.5% (336 patients) vs. 7.7% (331 patients) (P = 0.18) and mortality in 1.3% (52 patients) vs. 1.8% (77 patients) (P = 0.08) for the suPAR group and control group, respectively. Conclusion These post hoc analyses demonstrate that the availability of the prognostic biomarker suPAR was associated with a higher proportion of discharge within 24 hours and reduced length of stay, but more readmissions. In patients discharged within 24 hours, there was no difference in readmission or mortality. Trial Registration of the Main Trial This trial is registered with NCT02643459.
Collapse
|
34
|
Hjortkjær HØ, Jensen T, Hilsted J, Mogensen UM, Corinth H, Rossing P, Køber L, Kofoed KF. Left ventricular remodelling and cardiac chamber sizes in long-term, normoalbuminuric type 1 diabetes patients with and without cardiovascular autonomic neuropathy. J Diabetes Complications 2019; 33:171-177. [PMID: 30355473 DOI: 10.1016/j.jdiacomp.2018.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 01/19/2023]
Abstract
AIMS Type 1 diabetes is associated with increased cardiovascular (CV) morbidity and mortality, and cardiovascular autonomic neuropathy (CAN) is an important CV risk factor. The study aimed to explore associations between CAN and altered cardiac chamber sizes in persons with type 1 diabetes. METHODS This was a cross-sectional study of 71 asymptomatic, normoalbuminuric participants with long-term type 1 diabetes (39 with CAN, determined by >1 abnormal autonomic function test) examined with cardiac multi detector computed tomography scans, which allowed measurements of left ventricular mass and all four cardiac chamber volumes. Cardiac chambers were indexed according to body surface area (ml/m2 or g/m2). RESULTS Persons with and without CAN had mean ± SD age of 57 ± 7 and 50 ± 8 years (p < 0.001) and diabetes duration of 36 ± 11 and 32 ± 9 years (p < 0.05), respectively. Increasing autonomic dysfunction, evaluated by decrease in heart rate variability during deep breathing (in beats per minute), was associated with larger right (-0.5, 95% CI -1.0 to -0.0, p < 0.05) and trend towards larger left (-0.4, 95% CI -0.8-0.0, p < 0.1) ventricular volumes in multivariable linear regression. CONCLUSIONS Our results suggest that impaired autonomic function may be associated with modest enlargement of ventricular volumes; this might be an early sign of progression towards heart failure.
Collapse
Affiliation(s)
- Henrik Ø Hjortkjær
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | - Tonny Jensen
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jannik Hilsted
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Ulrik M Mogensen
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Helle Corinth
- Department of Endocrinology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Klaus F Kofoed
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Denmark; Department of Radiology, Rigshospitalet, Copenhagen University Hospital, Denmark
| |
Collapse
|
35
|
Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg AU. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev 2018; 47:214-277. [PMID: 30071357 DOI: 10.1016/j.arr.2018.07.004] [Citation(s) in RCA: 322] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Use of the frailty index to measure an accumulation of deficits has been proven a valuable method for identifying elderly people at risk for increased vulnerability, disease, injury, and mortality. However, complementary molecular frailty biomarkers or ideally biomarker panels have not yet been identified. We conducted a systematic search to identify biomarker candidates for a frailty biomarker panel. METHODS Gene expression databases were searched (http://genomics.senescence.info/genes including GenAge, AnAge, LongevityMap, CellAge, DrugAge, Digital Aging Atlas) to identify genes regulated in aging, longevity, and age-related diseases with a focus on secreted factors or molecules detectable in body fluids as potential frailty biomarkers. Factors broadly expressed, related to several "hallmark of aging" pathways as well as used or predicted as biomarkers in other disease settings, particularly age-related pathologies, were identified. This set of biomarkers was further expanded according to the expertise and experience of the authors. In the next step, biomarkers were assigned to six "hallmark of aging" pathways, namely (1) inflammation, (2) mitochondria and apoptosis, (3) calcium homeostasis, (4) fibrosis, (5) NMJ (neuromuscular junction) and neurons, (6) cytoskeleton and hormones, or (7) other principles and an extensive literature search was performed for each candidate to explore their potential and priority as frailty biomarkers. RESULTS A total of 44 markers were evaluated in the seven categories listed above, and 19 were awarded a high priority score, 22 identified as medium priority and three were low priority. In each category high and medium priority markers were identified. CONCLUSION Biomarker panels for frailty would be of high value and better than single markers. Based on our search we would propose a core panel of frailty biomarkers consisting of (1) CXCL10 (C-X-C motif chemokine ligand 10), IL-6 (interleukin 6), CX3CL1 (C-X3-C motif chemokine ligand 1), (2) GDF15 (growth differentiation factor 15), FNDC5 (fibronectin type III domain containing 5), vimentin (VIM), (3) regucalcin (RGN/SMP30), calreticulin, (4) PLAU (plasminogen activator, urokinase), AGT (angiotensinogen), (5) BDNF (brain derived neurotrophic factor), progranulin (PGRN), (6) α-klotho (KL), FGF23 (fibroblast growth factor 23), FGF21, leptin (LEP), (7) miRNA (micro Ribonucleic acid) panel (to be further defined), AHCY (adenosylhomocysteinase) and KRT18 (keratin 18). An expanded panel would also include (1) pentraxin (PTX3), sVCAM/ICAM (soluble vascular cell adhesion molecule 1/Intercellular adhesion molecule 1), defensin α, (2) APP (amyloid beta precursor protein), LDH (lactate dehydrogenase), (3) S100B (S100 calcium binding protein B), (4) TGFβ (transforming growth factor beta), PAI-1 (plasminogen activator inhibitor 1), TGM2 (transglutaminase 2), (5) sRAGE (soluble receptor for advanced glycosylation end products), HMGB1 (high mobility group box 1), C3/C1Q (complement factor 3/1Q), ST2 (Interleukin 1 receptor like 1), agrin (AGRN), (6) IGF-1 (insulin-like growth factor 1), resistin (RETN), adiponectin (ADIPOQ), ghrelin (GHRL), growth hormone (GH), (7) microparticle panel (to be further defined), GpnmB (glycoprotein nonmetastatic melanoma protein B) and lactoferrin (LTF). We believe that these predicted panels need to be experimentally explored in animal models and frail cohorts in order to ascertain their diagnostic, prognostic and therapeutic potential.
Collapse
|
36
|
Cimeno A, Munley J, Drachenberg C, Weir M, Haririan A, Bromberg J, Barth RN, Scalea JR. Diabetic nephropathy after kidney transplantation in patients with pretransplantation type II diabetes: A retrospective case series study from a high-volume center in the United States. Clin Transplant 2018; 32:e13425. [PMID: 30326148 DOI: 10.1111/ctr.13425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/31/2018] [Accepted: 09/18/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Patients with type II diabetes mellitus (DM) undergoing renal transplantation are at risk of diabetic nephropathy (DN) in the transplanted kidney. The true risk of developing post-transplantation DN is unknown, and post-transplantation DN is poorly characterized in the literature. METHODS The biopsy database at the University of Maryland Medical Center was queried for kidney transplant biopsies which demonstrated evidence of DN. The time from transplantation to biopsy-proven DN (time to diagnosis, TTD) was calculated and analyzed in the context of demographics, serum creatinine, and onset of diabetes. By extrapolating the total number of patients who developed DN in the last 2 years, we estimated the recurrence rate of DN. RESULTS Sixty patients whose renal biopsies met criteria were identified. The mean age was 56.6 (±1.58) years, and the mean creatinine level at time of biopsy was 1.65 (±0.12) mg/dL. Simultaneous pathological diagnoses were frequent on kidney biopsy; rejection was present at variable rates: classes I, IIA, IIB, and III were 5.0%, 66.7%, 18.4%, and 10%, respectively. The mean TTD was 1456 (±206) days. TTD was significantly shorter for patients receiving a cadaveric vs living donor renal transplant (1118 ± 184 vs 2470 ± 547 days, P = 0.004). Older patients (r = 0.378, P = 0.003) and patients with higher serum creatinine (r = 0.282, P = 0.029) had shorter TTDs. Extrapolations showed that 74.7% of patients would be free of DN 10 years after renal transplantation. CONCLUSIONS Diabetic nephropathy occurs after transplantation, and this appears to be due to both donor and recipient-derived factors. Encouragingly, our estimates suggest that as many as 75% of patients may be free of DN at 10 years following kidney transplantation.
Collapse
Affiliation(s)
- Arielle Cimeno
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jennifer Munley
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Cinthia Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew Weir
- Department of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Abdolreza Haririan
- Department of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jonathan Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rolf N Barth
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joseph R Scalea
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
37
|
Mechanisms underlying modulation of podocyte TRPC6 channels by suPAR: Role of NADPH oxidases and Src family tyrosine kinases. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3527-3536. [PMID: 30293571 DOI: 10.1016/j.bbadis.2018.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/02/2018] [Accepted: 08/06/2018] [Indexed: 01/10/2023]
Abstract
The soluble urokinase receptor (suPAR) has been implicated in the pathogenesis of chronic kidney diseases (CKD) and may function as a circulating "permeability factor" driving primary focal and segmental glomerulosclerosis (FSGS). Here we examined the mechanisms whereby suPAR causes mobilization and increased activation of Ca2+-permeable TRPC6 channels, which are also implicated in FSGS. Treatment of immortalized mouse podocytes with recombinant suPAR for 24 h caused a marked increase in cytosolic reactive oxygen species (ROS) that required signaling through integrins. This effect was associated with increased assembly of active cell surface NADPH oxidase 2 (Nox2) complexes and was blocked by the Nox2 inhibitor apoycynin. Treatment with suPAR also evoked a functionally measurable increase in TRPC6 channels that was blocked by concurrent treatment with the ROS-quencher TEMPOL as well as by inhibition of Rac1, an essential component of active Nox2 complexes. Elevated ROS evoked by exposing cells to suPAR or H2O2 caused a marked increase in the abundance of tyrosine-phosphorylated proteins including Src, and suPAR-evoked Src activation was blocked by TEMPOL. Moreover, mobilization and increased activation of TRPC6 by suPAR or H2O2 was blocked by concurrent exposure to PP2, an inhibitor of Src family tyrosine kinases. These data suggest that suPAR induces oxidative stress in podocytes that in turn drives signaling through Src family kinases to upregulate TRPC6 channels. The combination of oxidative stress and altered Ca2+ signaling may contribute to loss of podocytes and progression of various forms of CKD.
Collapse
|
38
|
Luo Q, Ning P, Zheng Y, Shang Y, Zhou B, Gao Z. Serum suPAR and syndecan-4 levels predict severity of community-acquired pneumonia: a prospective, multi-centre study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:15. [PMID: 29368632 PMCID: PMC5784729 DOI: 10.1186/s13054-018-1943-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022]
Abstract
Background Community-acquired pneumonia (CAP) is a major cause of death worldwide and occurs with variable severity. There are few studies focused on the expression of soluble urokinase-type plasminogen activator receptor (suPAR) and syndecan-4 in patients with CAP. Methods A prospective, multi-centre study was conducted between January 2014 and December 2016. A total of 103 patients with severe CAP (SCAP), 149 patients with non-SCAP, and 30 healthy individuals were enrolled. Clinical data were recorded for all enrolled patients. Serum suPAR and syndecan-4 levels were determined by quantitative enzyme-linked immunosorbent assay. The t test and Mann–Whitney U test were used to compare between two groups; one-way analysis of variance and the Kruskal–Wallis test were used to compare multiple groups. Correlations were assessed using Pearson and Spearman tests. Area under the curve (AUCs), optimal threshold values, sensitivity, and specificity were calculated. Survival curves were constructed and compared by log-rank test. Regression analyses assessed the effect of multiple variables on 30-day survival. Results suPAR levels increased in all patients with CAP, especially in severe cases. Syndecan-4 levels decreased in patients with CAP, especially in non-survivors. suPAR and syndecan-4 levels were positively and negatively correlated with severity scores, respectively. suPAR exhibited high accuracy in predicting SCAP among patients with CAP with an AUC of 0.835 (p < 0.001). In contrast, syndecan-4 exhibited poor diagnostic value for predicting SCAP (AUC 0.550, p = 0.187). The AUC for predicting mortality in patients with SCAP was 0.772 and 0.744 for suPAR and syndecan-4, respectively; the respective prediction threshold values were 10.22 ng/mL and 6.68 ng/mL. Addition of both suPAR and syndecan-4 to the Pneumonia Severity Index significantly improved their prognostic accuracy, with an AUC of 0.885. Regression analysis showed that suPAR ≥10.22 ng/mL and syndecan-4 ≤ 6.68 ng/mL were reliable independent markers for prediction of 30-day survival. Conclusion suPAR exhibits high accuracy for both diagnosis and prognosis of SCAP. Syndecan-4 can reliably predict mortality in patients with SCAP. Addition of both suPAR and syndecan-4 to a clinical scoring method could improve prognostic accuracy. Trial registration ClinicalTrials.gov, NCT03093220. Registered on 28 March 2017 (retrospectively registered).
Collapse
Affiliation(s)
- Qiongzhen Luo
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, People's Republic of China
| | - Pu Ning
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, People's Republic of China
| | - Yali Zheng
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, People's Republic of China
| | - Ying Shang
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, People's Republic of China
| | - Bing Zhou
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, People's Republic of China
| | - Zhancheng Gao
- Department of Respiratory & Critical Care Medicine, Peking University People's Hospital, Beijing, People's Republic of China.
| |
Collapse
|
39
|
Peeters SA, Engelen L, Buijs J, Chaturvedi N, Fuller JH, Jorsal A, Parving HH, Tarnow L, Theilade S, Rossing P, Schalkwijk CG, Stehouwer CDA. Circulating matrix metalloproteinases are associated with arterial stiffness in patients with type 1 diabetes: pooled analysis of three cohort studies. Cardiovasc Diabetol 2017; 16:139. [PMID: 29070037 PMCID: PMC5657128 DOI: 10.1186/s12933-017-0620-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/15/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Altered regulation of extracellular matrix (ECM) composition by matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinase (TIMPs) may contribute to arterial stiffening. We investigated associations between circulating MMP-1, -2, -3, -9, -10 and TIMP-1, and carotid-femoral pulse wave velocity (cfPWV) and pulse pressure (PP), as markers of arterial stiffness in type 1 diabetic patients. METHODS Individuals with type 1 diabetes from three different cohorts were included in this study: EURODIAB Prospective Complications study (n = 509), LEACE (n = 370) and PROFIL (n = 638). Linear regression analyses were used to investigate cross-sectional associations between circulating levels of MMP-1, -2, -3, -9, -10, and TIMP-1 and cfPWV (n = 614) as well as office PP (n = 1517). Data on 24-h brachial and 24-h central PP were available in 638 individuals from PROFIL. Analyses were adjusted for age, sex, duration of diabetes, HbA1c, mean arterial pressure (MAP), and eGFR, and additionally for other cardiovascular risk factors and presence of vascular complications. RESULTS After adjustment for potential confounders and presence of vascular complications, circulating MMP-3 was associated with cfPWV [β per 1 SD higher lnMMP3 0.29 m/s (0.02; 0.55)]. In addition, brachial and central 24-h PP measurements in PROFIL were significantly associated with MMP-2 [(1.40 (0.47:2.33) and 1.43 (0.63:2.23)]. Pooled data analysis showed significant associations of circulating levels of MMP-1 and MMP-2 with office PP [β per 1 SD higher lnMMP-1 and lnMMP-2 = - 0.83 mmHg (95% CI - 1.50; - 0.16) and = 1.33 mmHg (0.55; 2.10), respectively]. CONCLUSIONS MMPs-1, -2, and -3 are independently associated with markers of arterial stiffening in patients with type 1 diabetes and may become therapeutic targets.
Collapse
Affiliation(s)
- Stijn A. Peeters
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- Department of Internal Medicine, Zuyderland hospital, Heerlen, The Netherlands
| | - Lian Engelen
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- Centraal Bureau voor de Statistiek, Heerlen, The Netherlands
| | - Jacqueline Buijs
- Department of Internal Medicine, Zuyderland hospital, Heerlen, The Netherlands
| | - Nish Chaturvedi
- Institute of Cardiovascular Sciences, University College London, London, UK
| | - John H. Fuller
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Anders Jorsal
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Hans-Henrik Parving
- Department of Medical Endocrinology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lise Tarnow
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Faculty of Health, Aarhus University, Aarhus, Denmark
| | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - Casper G. Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Coen D. A. Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
40
|
Mossanen JC, Pracht J, Jansen TU, Buendgens L, Stoppe C, Goetzenich A, Struck J, Autschbach R, Marx G, Tacke F. Elevated Soluble Urokinase Plasminogen Activator Receptor and Proenkephalin Serum Levels Predict the Development of Acute Kidney Injury after Cardiac Surgery. Int J Mol Sci 2017; 18:ijms18081662. [PMID: 28758975 PMCID: PMC5578052 DOI: 10.3390/ijms18081662] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022] Open
Abstract
Acute kidney injury (AKI) develops in up to 40% of patients after cardiac surgery. The soluble urokinase plasminogen activator receptor (suPAR) has been identified as a biomarker for incident chronic kidney disease (CKD). Proenkephalin (proENK) also has been shown to be a biomarker for renal dysfunction. We hypothesized that pre-surgery suPAR and proENK levels might predict AKI in patients undergoing cardiac surgery. Consecutive patients (n = 107) undergoing elective cardiac surgery were studied prospectively. Clinical data, laboratory parameters, suPAR and proENK serum levels were assessed before operation, after operation and days one and four post-operatively. A total of 21 (19.6%) patients developed AKI within the first four days after elective surgery. Serum levels of suPAR and proENK, but not of creatinine, were significantly higher before surgery in these patients compared to those patients without AKI. This difference remained significant for suPAR, if patients with or without AKI were matched for risk factors (hypertension, diabetes, CKD). If cardiac surgery patients with pre-existing CKD (n = 10) were excluded, only pre-operative suPAR but not proENK serum levels remained significantly elevated in patients with subsequent AKI. Thus, our findings indicate that suPAR may be a predictive biomarker for AKI in the context of cardiac surgery, even in patients without underlying CKD.
Collapse
Affiliation(s)
- Jana C Mossanen
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany.
- Department of Intensive and Intermediate Care, University Hospital Aachen, 52074 Aachen, Germany.
| | - Jessica Pracht
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany.
| | - Tobias U Jansen
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany.
| | - Lukas Buendgens
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany.
| | - Christian Stoppe
- Department of Intensive and Intermediate Care, University Hospital Aachen, 52074 Aachen, Germany.
| | - Andreas Goetzenich
- Department of Thoracic and Cardiovascular Surgery, University Hospital Aachen, 52074 Aachen, Germany.
| | | | - Rüdiger Autschbach
- Department of Thoracic and Cardiovascular Surgery, University Hospital Aachen, 52074 Aachen, Germany.
| | - Gernot Marx
- Department of Intensive and Intermediate Care, University Hospital Aachen, 52074 Aachen, Germany.
| | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, 52074 Aachen, Germany.
| |
Collapse
|
41
|
Svenningsen P, Hinrichs GR, Zachar R, Ydegaard R, Jensen BL. Physiology and pathophysiology of the plasminogen system in the kidney. Pflugers Arch 2017; 469:1415-1423. [DOI: 10.1007/s00424-017-2014-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/09/2017] [Accepted: 06/12/2017] [Indexed: 12/31/2022]
|
42
|
von Scholten BJ, Persson F, Rosenlund S, Eugen-Olsen J, Pielak T, Faber J, Hansen TW, Rossing P. Effects of liraglutide on cardiovascular risk biomarkers in patients with type 2 diabetes and albuminuria: A sub-analysis of a randomized, placebo-controlled, double-blind, crossover trial. Diabetes Obes Metab 2017; 19:901-905. [PMID: 28105731 DOI: 10.1111/dom.12884] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/12/2017] [Accepted: 01/15/2017] [Indexed: 12/28/2022]
Abstract
We assessed the effects of liraglutide treatment on five cardiovascular risk biomarkers, reflecting different pathophysiology: tumour necrosis factor (TNF)-α; soluble urokinase plasminogen activator receptor (suPAR); mid-regional pro-adrenomedullin (MR-proADM); mid-regional pro-atrial natriuretic peptide (MR-proANP); and copeptin, in people with type 2 diabetes with albuminuria. In a randomized, double-blind, placebo-controlled, crossover trial we enrolled people with type 2 diabetes and persistent albuminuria (urinary albumin-to-creatinine ratio [UACR] >30 mg/g) and estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2 . Participants received liraglutide (1.8 mg/d) and matched placebo for 12 weeks, in random order. The primary endpoint was change in albuminuria; this was a prespecified sub-study. A total of 32 participants were randomized, of whom 27 completed the study. TNF-α level was 12% (95% confidence interval [CI] 3; 20) lower after liraglutide treatment compared with placebo (P = .012); MR-proADM level was 4% (95% CI 0; 8) lower after liraglutide treatment compared with placebo (P = .038), and MR-proANP level was 13% (95% CI 4; 21) lower after liraglutide treatment compared with placebo (P = .006). In the present study, we showed anti-inflammatory effects of liraglutide treatment, reflected in reductions in levels of TNF-α and MR-proADM, while the reduction in MR-proANP levels may represent a clinically relevant benefit with regard to heart failure.
Collapse
Affiliation(s)
- Bernt Johan von Scholten
- Department of Diabetes Complications Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Frederik Persson
- Department of Diabetes Complications Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Signe Rosenlund
- Department of Diabetes Complications Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Jesper Eugen-Olsen
- Department of Clinical Biochemistry, Clinical Research Centre, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | | | - Jens Faber
- Department of Endocrinology, Herlev Hospital, Herlev, Denmark
| | - Tine W Hansen
- Department of Diabetes Complications Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Peter Rossing
- Department of Diabetes Complications Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
43
|
Hayek SS, Ko YA, Awad M, Ahmed H, Gray B, Hosny KM, Aida H, Tracy MJ, Wei C, Sever S, Reiser J, Quyyumi AA. Cardiovascular Disease Biomarkers and suPAR in Predicting Decline in Renal Function: A Prospective Cohort Study. Kidney Int Rep 2017; 2:425-432. [PMID: 29142970 PMCID: PMC5678674 DOI: 10.1016/j.ekir.2017.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 11/02/2016] [Accepted: 02/02/2017] [Indexed: 02/01/2023] Open
Abstract
INTRODUCTION Soluble urokinase-type plasminogen activator receptor (suPAR) strongly predicts outcomes and incident chronic kidney disease (CKD) in patients with cardiovascular disease (CVD). Whether the association between suPAR and CKD is a reflection of its overall association with chronic inflammation and poor CVD outcomes is unclear. We examined whether CVD biomarkers, including high-sensitivity C-reactive protein (hs-CRP), fibrin-degradation products (FDPs), heat-shock protein 70 (HSP-70), and high-sensitivity troponin I (hs-TnI) were associated with a decline in kidney function in the Emory Cardiovascular Biobank cohort, in which suPAR levels were shown to be predictive of both incident CKD and CVD outcomes. METHODS We measured suPAR, hs-CRP, HSP-70, FDP, and hs-TnI plasma levels in 3282 adults (mean age 63 years, 64% male, 75% estimated glomerular filtration rate [eGFR] >60 ml/min per 1.73 m2). Glomerular filtration rate was estimated using Chronic Kidney Disease-Epidemiology Collaboration (eGFR) at enrollment (n = 3282) and follow-up (n = 2672; median 3.5 years). Urine protein by dipstick at baseline was available for 1335 subjects. RESULTS There was a weak correlation among biomarkers (r range: 0.17-0.28). hs-CRP, FDPs, hs-TnI, and suPAR were independently associated with baseline eGFR and proteinuria. The median yearly decline in eGFR was -0.6 ml/min per 1.73 m2. hs-CRP (β: -0.04; P = 0.46), FDPs (β: -0.13; P = 0.08), HSP-70 (β: 0.05; P = 0.84), or hs-TnI (β: -0.01; P = 0.76) were associated with eGFR decline. suPAR remained predictive of eGFR decline even after adjusting for all biomarkers. DISCUSSION hs-CRP, FDP, HSP-70, and hs-TnI were not associated with eGFR decline. The specific association of suPAR with eGFR decline supported its involvement in pathways specific to the pathogenesis of kidney disease.
Collapse
Affiliation(s)
- Salim S. Hayek
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yi-An Ko
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, Georgia, USA
| | - Mosaab Awad
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hina Ahmed
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Brandon Gray
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Hiroshi Aida
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Melissa J. Tracy
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Sanja Sever
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Arshed A. Quyyumi
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
44
|
Soluble urokinase receptor (suPAR) predicts microalbuminuria in patients at risk for type 2 diabetes mellitus. Sci Rep 2017; 7:40627. [PMID: 28091558 PMCID: PMC5238426 DOI: 10.1038/srep40627] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/08/2016] [Indexed: 01/29/2023] Open
Abstract
Early identification of patients at risk of developing diabetic nephropathy is essential. Elevated serum concentrations of soluble urokinase receptor (suPAR) associate with diabetes mellitus and predict onset and loss of renal function in chronic kidney disease. We hypothesize, that suPAR may be an early risk indicator for diabetic nephropathy, preceding microalbuminuria. The relationship of baseline suPAR and incident microalbuminuria was assessed in a prospective long-term cohort of subjects at increased risk for type 2 diabetes (TULIP, n = 258). Association with albuminuria at later stages of disease was studied in a cross-sectional cohort with manifest type 2 diabetes (ICEPHA, n = 266). A higher baseline suPAR was associated with an increased risk of new-onset microalbuminuria in subjects at risk for type 2 diabetes (hazard ratio 5.3 (95% CI 1.1-25.2, p = 0.03) for the highest vs. lowest suPAR quartile). The proportion of subjects with prediabetes at the end of observation was higher in subjects with new-onset microalbuminuria. suPAR consistently correlated with albuminuria in a separate cohort with manifest type 2 diabetes. Elevated baseline suPAR concentrations independently associate with new-onset microalbuminuria in subjects at increased risk of developing type 2 diabetes. suPAR may hence allow for earlier risk stratification than microalbuminuria.
Collapse
|
45
|
Desmedt S, Desmedt V, Delanghe JR, Speeckaert R, Speeckaert MM. The intriguing role of soluble urokinase receptor in inflammatory diseases. Crit Rev Clin Lab Sci 2017; 54:117-133. [DOI: 10.1080/10408363.2016.1269310] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - J. R. Delanghe
- Department of Clinical Chemistry, Ghent University Hospital, Gent, Belgium
| | - R. Speeckaert
- Department of Clinical Chemistry, Ghent University Hospital, Gent, Belgium
| | | |
Collapse
|
46
|
Fisher VL, Tahrani AA. Cardiac autonomic neuropathy in patients with diabetes mellitus: current perspectives. Diabetes Metab Syndr Obes 2017; 10:419-434. [PMID: 29062239 PMCID: PMC5638575 DOI: 10.2147/dmso.s129797] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiac autonomic neuropathy (CAN) is a common and often-underdiagnosed complication of diabetes mellitus (DM). CAN is associated with increased mortality, cardiovascular disease, chronic kidney disease, and morbidity in patients with DM, but despite these significant consequences CAN often remains undiagnosed for a prolonged period. This is commonly due to the disease being asymptomatic until the later stages, as well as a lack of easily available screening strategies. In this article, we review the latest developments in the epidemiology, pathogenesis, diagnosis, consequences, and treatments of CAN in patients with DM.
Collapse
Affiliation(s)
| | - Abd A Tahrani
- Institute of Metabolism and Systems Research, University of Birmingham
- Department of Diabetes and Endocrinology, Birmingham Heartlands Hospital
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Correspondence: Abd A Tahrani, Institute of Metabolism and Systems Research, Medical School, University of Birmingham, Birmingham B15 2TT, UK, Email
| |
Collapse
|
47
|
Dande RR, Peev V, Altintas MM, Reiser J. Soluble Urokinase Receptor and the Kidney Response in Diabetes Mellitus. J Diabetes Res 2017; 2017:3232848. [PMID: 28596971 PMCID: PMC5449757 DOI: 10.1155/2017/3232848] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 03/19/2017] [Indexed: 12/20/2022] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease (ESRD) worldwide. DN typically manifests by glomerular hyperfiltration and microalbuminuria; then, the disease progresses to impaired glomerular filtration rate, which leads to ESRD. Treatment options for DN include the strict control of blood glucose levels and pressure (e.g., intraglomerular hypertension). However, the search for novel therapeutic strategies is ongoing. These include seeking specific molecules that contribute to the development and progression of DN to potentially interfere with these "molecular targets" as well as with the cellular targets within the kidney such as podocytes, which play a major role in the pathogenesis of DN. Recently, podocyte membrane protein urokinase receptor (uPAR) and its circulating form (suPAR) are found to be significantly induced in glomeruli and sera of DN patients, respectively, and elevated suPAR levels predicted diabetic kidney disease years before the occurrence of microalbuminuria. The intent of this review is to summarize the emerging evidence of uPAR and suPAR in the clinical manifestations of DN. The identification of specific pathways that govern DN will help us build a more comprehensive molecular model for the pathogenesis of the disease that can inform new opportunities for treatment.
Collapse
Affiliation(s)
| | - Vasil Peev
- Rush University Medical Center, Chicago, IL, USA
| | - Mehmet M. Altintas
- Rush University Medical Center, Chicago, IL, USA
- *Mehmet M. Altintas: and
| | - Jochen Reiser
- Rush University Medical Center, Chicago, IL, USA
- *Jochen Reiser:
| |
Collapse
|
48
|
Persson F, Theilade S, Eugen-Olsen J, Rossing P, Parving HH. Renin angiotensin system blockade reduces urinary levels of soluble urokinase plasminogen activator receptor (suPAR) in patients with type 2 diabetes. J Diabetes Complications 2016; 30:1440-1442. [PMID: 27475262 DOI: 10.1016/j.jdiacomp.2016.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/22/2016] [Accepted: 07/06/2016] [Indexed: 11/18/2022]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is associated with faster decline in kidney function and the pathogenesis of diabetic nephropathy. However, little is known about the impact of treatment on plasma and urinary levels of suPAR. We aimed to investigate the impact of renin angiotensin system (RAS) single and dual blockade on suPAR levels in patients with type 2 diabetes and albuminuria. We conducted a post-hoc analysis of a randomized controlled crossover trial. Urine and plasma samples were analyzed for suPAR levels. The placebo period was considered reference and all treatment periods were compared to placebo. Patients (n = 22) were treated for 2-month periods with either placebo, irbesartan 300 mg once daily, aliskiren 300 mg once daily or irbesartan/aliskiren combination in random order. Placebo geometric mean plasma (SEM) levels of suPAR were 3.3 ng/mL (1.1) and urine levels were 4.0 ng/mL (1.1). None of the treatments had significant effects on plasma levels of suPAR compared to placebo. Compared to placebo, irbesartan and combination treatment decreased urinary levels of suPAR significantly (-1.3 ng/mL), while aliskiren did not. In patients with type 2 diabetes urinary levels of suPAR were reduced during RAS blockade treatment, which may contribute to renoprotection.
Collapse
Affiliation(s)
| | | | | | - Peter Rossing
- Steno Diabetes Center, Gentofte, Denmark; NNF Center for Basic and Metabolic Research, Copenhagen University, Denmark; HEALTH, Aarhus University, Aarhus, Denmark
| | - Hans-Henrik Parving
- HEALTH, Aarhus University, Aarhus, Denmark; Dept. of Medical Endocrinology, Rigshospitalet, Copenhagen University Hospital, Denmark
| |
Collapse
|
49
|
Theilade S, Rossing P, Eugen-Olsen J, Jensen JS, Jensen MT. suPAR level is associated with myocardial impairment assessed with advanced echocardiography in patients with type 1 diabetes with normal ejection fraction and without known heart disease or end-stage renal disease. Eur J Endocrinol 2016; 174:745-53. [PMID: 26951602 DOI: 10.1530/eje-15-0986] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/07/2016] [Indexed: 12/17/2022]
Abstract
AIM Heart disease is a common fatal diabetes-related complication. Early detection of patients at particular risk of heart disease is of prime importance. Soluble urokinase plasminogen activator receptor (suPAR) is a novel biomarker for development of cardiovascular disease. We investigate if suPAR is associated with early myocardial impairment assessed with advanced echocardiographic methods. METHODS In an observational study on 318 patients with type 1 diabetes without known heart disease and with normal left ventricular ejection fraction (LVEF) (biplane LVEF >45%), we performed conventional, tissue Doppler and speckle tracking echocardiography, and measured plasma suPAR levels. Associations between myocardial function and suPAR levels were studied in adjusted models including significant covariates. RESULTS Patients were 55±12 years (mean±s.d.) and 160 (50%) males. Median (interquartile range) suPAR was 3.4 (1.7) ng/mL and LVEF was 58±5%. suPAR levels were not associated with LVEF (P=0.11). In adjusted models, higher suPAR levels were independently associated with both impaired systolic function assessed with global longitudinal strain (GLS) and tissue velocity s', and with impaired diastolic measures a' and e'/a' (all P=0.034). In multivariable analysis including cardiovascular risk factors and both systolic and diastolic measures (GLS and e'/a'), both remained independently associated with suPAR levels (P=0.012). CONCLUSIONS In patients with type 1 diabetes with normal LVEF and without known heart disease, suPAR is associated with early systolic and diastolic myocardial impairment. Our study implies that both suPAR and advanced echocardiography are useful diagnostic tools for identifying patients with diabetes at risk of future clinical heart disease, suited for intensified medical therapy.
Collapse
MESH Headings
- Adult
- Aged
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/diagnostic imaging
- Diabetes Mellitus, Type 1/physiopathology
- Echocardiography
- Female
- Heart/diagnostic imaging
- Heart/physiopathology
- Humans
- Kidney Failure, Chronic/blood
- Kidney Failure, Chronic/complications
- Kidney Failure, Chronic/diagnostic imaging
- Kidney Failure, Chronic/physiopathology
- Male
- Middle Aged
- Receptors, Urokinase Plasminogen Activator/blood
- Risk Factors
- Ventricular Dysfunction, Left/blood
- Ventricular Dysfunction, Left/complications
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/physiopathology
Collapse
Affiliation(s)
| | - Peter Rossing
- Steno Diabetes CenterGentofte, Denmark Aarhus UniversityAarhus, Denmark University of CopenhagenCopenhagen, Denmark
| | | | - Jan S Jensen
- University of CopenhagenCopenhagen, Denmark Department of CardiologyGentofte Hospital, Gentofte, Denmark
| | - Magnus T Jensen
- Steno Diabetes CenterGentofte, Denmark Department of CardiologyGentofte Hospital, Gentofte, Denmark Department of Internal MedicineHolbaek Sygehus, Holbaek, Denmark
| |
Collapse
|
50
|
Hayek SS, Sever S, Ko YA, Trachtman H, Awad M, Wadhwani S, Altintas MM, Wei C, Hotton AL, French AL, Sperling LS, Lerakis S, Quyyumi AA, Reiser J. Soluble Urokinase Receptor and Chronic Kidney Disease. N Engl J Med 2015; 373:1916-25. [PMID: 26539835 PMCID: PMC4701036 DOI: 10.1056/nejmoa1506362] [Citation(s) in RCA: 315] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Relatively high plasma levels of soluble urokinase-type plasminogen activator receptor (suPAR) have been associated with focal segmental glomerulosclerosis and poor clinical outcomes in patients with various conditions. It is unknown whether elevated suPAR levels in patients with normal kidney function are associated with future decline in the estimated glomerular filtration rate (eGFR) and with incident chronic kidney disease. METHODS We measured plasma suPAR levels in 3683 persons enrolled in the Emory Cardiovascular Biobank (mean age, 63 years; 65% men; median suPAR level, 3040 pg per milliliter) and determined renal function at enrollment and at subsequent visits in 2292 persons. The relationship between suPAR levels and the eGFR at baseline, the change in the eGFR over time, and the development of chronic kidney disease (eGFR <60 ml per minute per 1.73 m(2) of body-surface area) were analyzed with the use of linear mixed models and Cox regression after adjustment for demographic and clinical variables. RESULTS A higher suPAR level at baseline was associated with a greater decline in the eGFR during follow-up; the annual change in the eGFR was -0.9 ml per minute per 1.73 m(2) among participants in the lowest quartile of suPAR levels as compared with -4.2 ml per minute per 1.73 m(2) among participants in the highest quartile (P<0.001). The 921 participants with a normal eGFR (≥ 90 ml per minute per 1.73 m(2)) at baseline had the largest suPAR-related decline in the eGFR. In 1335 participants with a baseline eGFR of at least 60 ml per minute per 1.73 m(2), the risk of progression to chronic kidney disease in the highest quartile of suPAR levels was 3.13 times as high (95% confidence interval, 2.11 to 4.65) as that in the lowest quartile. CONCLUSIONS An elevated level of suPAR was independently associated with incident chronic kidney disease and an accelerated decline in the eGFR in the groups studied. (Funded by the Abraham J. and Phyllis Katz Foundation and others.).
Collapse
Affiliation(s)
- Salim S Hayek
- From the Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine (S.S.H., Y.-A.K., M.A., L.S.S., S.L., A.A.Q.), and the Department of Biostatistics and Bioinformatics, Emory University (Y.-A.K.) - both in Atlanta; the Department of Medicine, Harvard Medical School, Boston, and Division of Nephrology, Massachusetts General Hospital, Charlestown - both in Massachusetts (S.S.); the Department of Pediatrics, NYU Langone Medical Center, New York (H.T.); and the Department of Medicine, Rush University Medical Center (S.W., M.M.A., C.W., A.L.F., J.R.), and the Women's Interagency HIV Study/CORE Center of Cook County (A.L.H., A.L.F.) - both in Chicago
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|