1
|
Belvederi F, Leggeri S, Urbani A, Baroni S. suPAR as a biomarker of support in different clinical settings. Clin Chim Acta 2025; 573:120303. [PMID: 40222544 DOI: 10.1016/j.cca.2025.120303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
The urokinase-type plasminogen activator receptor (uPAR) system, which includes protease, receptor and inhibitors, is essential for key cellular functions like immune activation, cell migration, and tissue remodeling. Soluble uPAR (suPAR), released into circulation, serves as a valuable biomarker for systemic inflammation and immune activation. Elevated suPAR levels are associated with disease severity in conditions such as infections, sepsis, cardiovascular diseases, renal injury, cancer, and autoimmune diseases providing prognostic value especially in acute settings. Recent advancements in diagnostic methods, have enhanced the accuracy of suPAR measurement in serum and plasma. New rapid tests, such as suPARnostic Quick Triage, as well as turbidimetric assays, further expand its clinical applicability. In this review, we discuss the suPAR biomarker, focusing on its biochemical structure, biological functions, measurement methods and areas of clinical interest in different fields of medicine.
Collapse
Affiliation(s)
- Fabio Belvederi
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Simone Leggeri
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy; Unit of Chemistry, Biochemistry and Molecular Biology, "A. Gemelli" Hospital Foundation IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy
| | - Silvia Baroni
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy; Unit of Chemistry, Biochemistry and Molecular Biology, "A. Gemelli" Hospital Foundation IRCCS, Largo A. Gemelli, 8, 00168 Rome, Italy.
| |
Collapse
|
2
|
Scalise E, Costa D, Gallelli G, Ielapi N, Turchino D, Accarino G, Faga T, Michael A, Bracale UM, Andreucci M, Serra R. Biomarkers and Social Determinants in Atherosclerotic Arterial Diseases: A Scoping Review. Ann Vasc Surg 2025; 113:41-63. [PMID: 39863282 DOI: 10.1016/j.avsg.2024.12.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/27/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Arterial diseases like coronary artery disease (CAD), carotid stenosis (CS), peripheral artery disease (PAD), and abdominal aortic aneurysm (AAA) have high morbidity and mortality, making them key research areas. Their multifactorial nature complicates patient treatment and prevention. Biomarkers offer insights into the biochemical and molecular processes, while social factors also significantly impact patients' health and quality of life. This scoping review aims to search the literature for studies that have linked the biological mechanisms of arterial diseases through biomarkers with social issues and to analyze them, supporting the interdependence of biological and social sciences. METHODS After a rigorous selection process, adhering to the Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines for Scoping Reviews, 30 articles were identified through Scopus, Web of Science, and PubMed. Inclusion and exclusion criteria were based on the population, intervention, comparator, outcome, time, and setting framework. Inclusion criteria were studies involving human subjects that explored the relationships among arterial diseases, biomarkers, and psychosocial factors, with no restrictions on publication date. Nonhuman studies, purely biological or medical analyses without psychosocial dimensions, and non-English publications were excluded. Eligible study types included experimental, observational, and review articles published in peer-reviewed journals. Data extraction focused on study characteristics, such as authors, publication year, country, methods, population, and findings. Results were synthesized narratively, as this format was deemed the most suitable for summarizing diverse findings. The quality or methodological rigor of the included studies was not formally assessed, consistent with the scoping review methodology. RESULTS In CAD, biomarkers such as high-sensitivity C-reactive protein are strongly associated with psychological stress, whereas lipoprotein (a) and the apolipoprotein B/apolipoprotein A1 ratio reflect lipid profiles that are influenced by socioeconomic factors and ethnicity. In CS, increased carotid intima-media thickness is linked to psychiatric conditions like attention deficit/hyperactivity disorder, and heat shock protein-70 levels are associated with socioeconomic status and gender. In PAD, inflammatory markers, including interleukin-6, intracellular adhesion molecule-1, and high-sensitivity C-reactive protein, mediate the connection between depression and disease severity, with gender and ethnicity influencing the expression of biomarkers and clinical outcomes. In AAA, factors like smoking and exposure to air pollution have increased matrix metalloproteinase levels and other inflammatory markers. Additionally, estradiol provides partial protection in women, underscoring the role of hormones and environmental influences in disease progression. Social determinants such as socioeconomic status, healthcare access, and ethnicity significantly affect biomarker levels and arterial disease progression. CONCLUSIONS These findings are crucial for the assumption that social determinants of health modulate the levels of inflammatory biomarkers involved in the progression of arterial diseases such as CAD, CS, PAD, and AAA. This highlights the need to integrate highly predictive mathematical systems into clinical practice, combining biological sciences with social sciences to achieve advanced standards in precision medicine. However, further studies are needed to validate these approaches fully.
Collapse
Affiliation(s)
- Enrica Scalise
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy; Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, Catanzaro, Italy
| | - Davide Costa
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy; Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, Catanzaro, Italy
| | - Giuseppe Gallelli
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy; Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, Catanzaro, Italy
| | - Nicola Ielapi
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, Roma, Italy
| | - Davide Turchino
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Giulio Accarino
- Department of Public Health, University Federico II of Naples, Naples, Italy; Vascular Surgery Unit, Struttura Ospedaliera ad Alta Specialità Mediterranea, Naples, Italy
| | - Teresa Faga
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Ashour Michael
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | | | - Michele Andreucci
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy; Interuniversity Center of Phlebolymphology (CIFL), "Magna Graecia" University, Catanzaro, Italy.
| |
Collapse
|
3
|
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
|
4
|
Ruzanovic A, Saric-Matutinovic M, Milinkovic N, Jovicic S, Dimic A, Matejevic D, Kostic O, Koncar I, Ignjatovic S. Significance of myeloperoxidase, pentraxin-3 and soluble urokinase plasminogen activator receptor determination in patients with moderate carotid artery stenosis. Scand J Clin Lab Invest 2024; 84:486-492. [PMID: 39508179 DOI: 10.1080/00365513.2024.2422404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/30/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024]
Abstract
We investigated serum concentrations of specific inflammatory parameters in patients with significant carotid artery stenosis (CAS) of 50-99%, with an additional focus on patients with moderate stenosis (50-69%), in terms of both symptomatic status and plaque morphology, to determine whether there are certain parameters that can be associated with plaque instability before the progression of CAS to a high degree. The study included 119 CAS patients, 29 of whom had moderate stenosis, and 46 controls. Ultrasonography of the carotid arteries was performed using color flow Doppler and B-mode duplex ultrasound, and serum inflammatory parameters were measured using commercially available enzyme immunoassays. When comparing patients with 50-99% stenosis, only serum amyloid A (SAA) was higher in symptomatic patients, while in the group of patients with 50-69% stenosis, myeloperoxidase (MPO) was higher and pentraxin-3 (PTX-3) was lower in symptomatic compared to asymptomatic patients, and soluble urokinase plasminogen activator receptor (suPAR) was higher in patients with carotid plaque of unstable compared to stable morphology. Our results suggest that the importance of different inflammatory parameters in patients with moderate CAS is not the same as in CAS patients in general, and therefore their separate investigation in patients with high and moderate stenosis may be beneficial. SAA has the potential to be further considered in research to predict CAS symptom risk. There is a possibility that MPO and PTX-3 play a role in the development of CAS symptoms originating from less stenotic plaques and that suPAR is involved in the destabilisation of such plaques.
Collapse
Affiliation(s)
- Ana Ruzanovic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | | | - Neda Milinkovic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Snezana Jovicic
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Andreja Dimic
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - David Matejevic
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
| | - Ognjen Kostic
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
| | - Igor Koncar
- University Clinical Center of Serbia, Clinic for Vascular and Endovascular surgery, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
5
|
Christensen LWS, Iversen E, Andersen AL, Walls AB, Rasmussen LJH, Andersen O, Kallemose T, Houlind MB. A characterization of patients with low soluble urokinase plasminogen activator receptor who died within 90 days of hospital discharge. Basic Clin Pharmacol Toxicol 2024; 135:364-371. [PMID: 38988231 DOI: 10.1111/bcpt.14050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/29/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is a marker of systemic chronic inflammation. Elevated suPAR levels are associated with adverse clinical outcomes, but a small subset of patients with low suPAR also experience poor outcomes. Therefore, we aimed to characterize patients presenting to the emergency department with low suPAR (<3 ng/mL) who died within 90 days after discharge in a registry-based study. Compared to patients with low suPAR who survived (n = 15 122), those who died within 90 days (n = 87) had higher age (75.4 years), higher medication use (7.0; 71.3% with polypharmacy) and more blood tests outside reference intervals (5.0) (including C-reactive protein, neutrophils and albumin), and the most common diagnoses were chronic pulmonary disease (27.6%), cerebrovascular disease (18.4%) and dementia (11.5%). Patients with low suPAR were more morbid than what was reflected by suPAR alone. Future studies must determine which factors that contribute the most to potential algorithms when stratifying patients based on their risk of adverse clinical outcomes. These data indicate that inclusion of medication data could be relevant.
Collapse
Affiliation(s)
- Louise Westberg Strejby Christensen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Aino Leegaard Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Anne Byriel Walls
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Emergency Department, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
- The Capital Region Pharmacy, Herlev, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Chen Y, Gue Y, McDowell G, Gorog DA, Lip GYH. Impaired endogenous fibrinolysis status: a potential prognostic predictor in ischemic stroke. Minerva Med 2024; 115:364-379. [PMID: 38727704 DOI: 10.23736/s0026-4806.24.09133-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Stroke confers a severe global healthcare burden, hence exploring risk factors for stroke occurrence and prognosis is important for stroke prevention and post-stroke management strategies. Endogenous fibrinolysis is a spontaneous physiological protective mechanism that dissolves thrombus to maintain vascular patency. Recently, impaired endogenous fibrinolysis has been considered as a potential novel cardiovascular risk factor, but its link with ischaemic stroke in the past has been underappreciated. In this review, we summarize the latest mechanisms of endogenous fibrinolysis, review the current evidence and data on endogenous fibrinolysis in ischemic stroke. It includes the structure of thrombus in ischemic stroke patients, the effect of fibrin structure on the endogenous fibrinolytic efficiency, and the association between intravenous thrombolytic therapy and endogenous fibrinolysis in ischemic stroke. It also includes the single factors (tissue plasminogen activator, urokinase plasminogen activator, plasminogen activator inhibitor-1, thrombin activatable fibrinolysis inhibitor, complement component 3, complement component 5, alpha-2-antiplasmin, plasmin-alpha-2-antiplasmin complex, and lipoprotein[a]), and the global assessments of endogenous fibrinolysis status (thromboelastography, rotational thromboelastometry, and global thrombosis test), and their potential as predictors to identify occurrence or unfavorable functional outcomes of ischemic stroke. All of these assessments present advantages and limitations, and we suggest that the global thrombosis test may be more appropriate for detecting impaired endogenous fibrinolysis status in ischemic stroke patients.
Collapse
Affiliation(s)
- Yang Chen
- Liverpool Center for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Ying Gue
- Liverpool Center for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK -
| | - Garry McDowell
- Liverpool Center for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Diana A Gorog
- School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Hatfield, UK
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, UK
| | - Gregory Y H Lip
- Liverpool Center for Cardiovascular Science at University of Liverpool, Liverpool John Moores University, Liverpool Heart and Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
7
|
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
|
8
|
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: 11] [Impact Index Per Article: 11.0] [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
|
9
|
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
|
10
|
Ismail A, Hayek SS. Role of Soluble Urokinase-Type Plasminogen Activator Receptor in Cardiovascular Disease. Curr Cardiol Rep 2023; 25:1797-1810. [PMID: 37948017 DOI: 10.1007/s11886-023-01991-7] [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] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
PURPOSE OF REVIEW Chronic inflammation is a major contributor to cardiovascular disease (CVD) risk. Soluble urokinase plasminogen activator receptor (suPAR) is an immune-derived glycoprotein that is strongly associated with atherosclerotic disease. This review summarizes evidence on suPAR's role in CVD pathogenesis and its potential as a prognostic indicator and therapeutic target. RECENT FINDINGS Clinical, genetic, and experimental evidence supports suPAR's role as a pathogenic factor in atherosclerosis. suPAR promotes atherosclerosis through modulation of monocyte activation and function. Clinically, elevated suPAR levels are linked to increased cardiovascular risk across diverse populations. Ongoing clinical trials are evaluating therapies targeting suPAR signaling. Current evidence positions suPAR as a regulator of myeloid cell function that contributes to vascular inflammation and subsequent cardiovascular events. Additional research is needed to determine whether suPAR measurement can improve CVD risk prediction and enable personalized management. Overall, suPAR is a promising immune-derived biomarker and target for reducing inflammation and cardiovascular risk.
Collapse
Affiliation(s)
- Anis Ismail
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, 48109, MI, USA
| | - Salim S Hayek
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, 48109, MI, USA.
| |
Collapse
|
11
|
Zimmermann P, Schierbauer J, Kopf N, Sourij H, Oliver N, Aberer F, Wachsmuth NB, Moser O. Speckle-Tracking Analysis of the Right and Left Heart after Peak Exercise in Healthy Subjects with Type 1 Diabetes: An Explorative Analysis of the AppEx Trial. J Cardiovasc Dev Dis 2023; 10:467. [PMID: 37998525 PMCID: PMC10672090 DOI: 10.3390/jcdd10110467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/06/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
In eight healthy participants with Type 1 diabetes (T1D) exercise-related dynamic cardiac remodeling was analyzed by performing two-dimensional echocardiography, including deformation analysis of the left-ventricular (LV) global longitudinal strain (LV-GLS), and the deformation pattern of the left atrium (LA) and right ventricle (RV) at rest and post-peak performance on a bicycle. The feasibility echocardiographic speckle-tracking analysis was performed on eight asymptomatic participants with T1D (n = 8, male n = 5, age: 23-65 years). The obtained echocardiographic data were compared for various echocardiographic parameters at rest and post exercise. Across our participating T1D individuals no structural echocardiographic abnormalities of concern could be revealed. All participating T1D subjects showed preserved contractile reserve of the LV and no significant diastolic dysfunction. Significant differences were found for the phasic LA contractile strain pattern at rest and post exercise (p < 0.001), whereby the dynamic RV (p = 0.5839 and p = 0.7419) and LV strain pattern (p = 0.5952) did not reveal significant differences in comparison to resting conditions. This descriptive secondary outcome analysis describes preserved contractile reserve of the LV and elucidates dynamic modification of the phasic LA contractile deformation pattern in asymptomatic T1D individuals after exhaustive exercise on a bicycle.
Collapse
Affiliation(s)
- Paul Zimmermann
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (J.S.); (N.K.); (N.B.W.)
- Interdisciplinary Center of Sportsmedicine Bamberg, Klinikum Bamberg, 96049 Bamberg, Germany
- Department of Cardiology, Klinikum Bamberg, 96049 Bamberg, Germany
| | - Janis Schierbauer
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (J.S.); (N.K.); (N.B.W.)
| | - Niklas Kopf
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (J.S.); (N.K.); (N.B.W.)
| | - Harald Sourij
- Interdisciplinary Metabolic Medicine Research Group, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (H.S.); (F.A.)
| | - Nick Oliver
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK;
| | - Felix Aberer
- Interdisciplinary Metabolic Medicine Research Group, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (H.S.); (F.A.)
| | - Nadine B. Wachsmuth
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (J.S.); (N.K.); (N.B.W.)
| | - Othmar Moser
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (P.Z.); (J.S.); (N.K.); (N.B.W.)
- Interdisciplinary Metabolic Medicine Research Group, Division of Endocrinology and Diabetology, Medical University of Graz, 8036 Graz, Austria; (H.S.); (F.A.)
| |
Collapse
|
12
|
Mohammed MS, Ahmed HS. Plasminogen activator urokinase receptor as a diagnostic and prognostic biomarker in type 2 diabetic patients with cardiovascular disease. J Cardiovasc Thorac Res 2023; 15:154-160. [PMID: 38028718 PMCID: PMC10590464 DOI: 10.34172/jcvtr.2023.32895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/05/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Cardiovascular diseases are the main cause of death among type 2 diabetic patients. Higher levels of plasminogen activator urokinase receptor have been found to predict morbidity and mortality across acute and chronic diseases in the common populace. This study aims to explore the role of serum plasminogen activator urokinase receptor levels as a cardiometabolic risk factor among type 2 diabetic Iraqi patients. Methods Seventy type 2 diabetic patients (40 male and 30 female) (mean age: 46.20±7.56 years) participated in this study; 35 patients were with cardiovascular disease and 35 were without cardiovascular disease; their ages range was 40-55 years. In addition, 30 individuals who apparently healthy were selected as the control group. Results There were significant increases (P<0.05) in glycemic and lipid profiles in diabetic patients with cardiovascular disease as compared to those without cardiovascular disease and control group. The present results reveal high levels of plasminogen activator urokinase receptor (2500.72±12.36 ρg/mL versus 2255.32±10.15 ρg/mL) with OR=1.80, 95%CI 1.2, and P=0.0001 in type 2 diabetic patients with and without cardiovascular disease respectively as compared to healthy control (229.00±14.48 ρg/mL). Conclusion It has been concluded that serum plasminogen activator urokinase receptor showed higher levels among type 2 diabetic patients with cardiovascular disease, this revealed it's critical role in cardiac disease. Therefore, it could be considered a more sensitive biomarker for the detection of cardiovascular events among type 2 diabetic patients who were at high-risk.
Collapse
Affiliation(s)
| | - Hind Shakir Ahmed
- Department of Chemistry, College of Education for Pure Science (Ibn Al-Haitham), University of Baghdad, Baghdad, Iraq
| |
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
|
Kapoor PM, Prakash M, Mujahid OM. suPAR as a risk Prediction Biomarker in Extracorporeal Membrane Oxygenation. JOURNAL OF CARDIAC CRITICAL CARE TSS 2023. [DOI: 10.25259/jccc_5_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
suPAR is a promising biomarker of cardiovascular diseases, as it reflects “low-grade inflammation” and is associated with lifestyle factors such as smoking, alcohol, and an inactive lifestyle. suPAR is expressed in various cells involved in the development of atherosclerosis, including macrophages, endothelial cells, and smooth muscle cells, and an accumulation of suPAR in the atheroma has also been found. suPAR plays a role in the coagulation cascade during plasminogen activation and fibrinolysis. This abstract provides description of three case series showing the utility of suPAR as a risk prediction biomarker on VA extracorporeal membrane oxygenation (ECMO). We used SUPAR in 15 patients undergoing cardiac surgery of which three went on VA ECMO. Herein, we describe in detail three such patients and discuss each with good review of the literature.
Collapse
Affiliation(s)
| | - Mohit Prakash
- Department of Cardiac Anesthesia and Critical Care, AIIMS, New Delhi, India,
| | | |
Collapse
|
15
|
Tavenier J, Rasmussen LJH, Tolstrup J, Petersen J, Sobocki J, Pisinger C, Eugen-Olsen J, Gamst-Jensen H. Self-rated health and chronic inflammation are related and independently associated with hospitalization and long-term mortality in the general population. Sci Rep 2022; 12:19761. [PMID: 36396700 PMCID: PMC9670062 DOI: 10.1038/s41598-022-24422-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
The subjective indicator of health self-rated health (SRH) and the chronic inflammation biomarker soluble urokinase plasminogen activator receptor (suPAR) are both robust predictors of healthcare use and mortality. However, the possible relationship between SRH and suPAR in the assessment of hospitalization and mortality risk is unknown. We used data from the Danish population-based Inter99 cohort to examine the association between SRH and suPAR and test their individual and combined associations with 2-year risk of acute hospitalization and 5- and 15-year mortality. SRH and serum suPAR levels were measured in 5490 participants (median age 45.1 years, 48.7% men). Poorer SRH was associated with elevated suPAR. In unadjusted analyses, SRH and suPAR were individually associated with higher risks of acute hospitalization and mortality, and both measures remained independently associated with higher risks of hospitalization and 15-year mortality after mutual adjustments. The association of suPAR with mortality was stronger in poorer SRH categories, and when combined, SRH and suPAR could identify different groups of individuals with increased risk of acute hospitalization and mortality. Both SRH and suPAR were independently associated with risk of acute hospitalization and mortality, and different combinations of the two measures could identify different groups of individuals at increased risk.
Collapse
Affiliation(s)
- Juliette Tavenier
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Janne Tolstrup
- National Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Janne Petersen
- Copenhagen Phase 4 Unit, Department of Clinical Pharmacology and Center for Clinical Research and Prevention, Copenhagen University Hospital Bispebjerg and Frederiksberg, Frederiksberg, Denmark
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Sobocki
- Emergency Department, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Charlotta Pisinger
- Center for Clinical Research and Prevention, Copenhagen University Hospital Bispebjerg and Frederiksberg, Frederiksberg, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Hejdi Gamst-Jensen
- Department of Clinical Research, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Emergency Department, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| |
Collapse
|
16
|
Koschitzky M, Navrazhina K, Garshick MS, Gonzalez J, Han J, Garcet S, Krueger JG. Ustekinumab reduces serum protein levels associated with cardiovascular risk in psoriasis vulgaris. Exp Dermatol 2022; 31:1341-1351. [PMID: 35474520 PMCID: PMC9869081 DOI: 10.1111/exd.14582] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/30/2022] [Accepted: 04/22/2022] [Indexed: 01/26/2023]
Abstract
Psoriasis increases the risk of cardiovascular disease (CVD). Biomarkers for cardiovascular (CV) risk stratification in psoriasis are lacking, and the effects of psoriasis biologics on CV risk reduction remain unclear. The goal of this study was to identify biomarkers of CV risk in psoriasis blood that are reduced by ustekinumab. We quantified 276 inflammatory and CV-related serum proteins with Olink's multiplex assay in 10 psoriasis patients (vs. 18 healthy controls) and after 12 weeks of ustekinumab treatment. For each protein down-regulated after treatment, the literature was reviewed for studies assessing the protein's association with CVD. Data were collected from each study to calculate CV risk thresholds for each protein, which were compared with protein levels in psoriasis patients before and after treatment. Our results showed that 43 out of 276 proteins were down-regulated after treatment, 25 of which were initially up-regulated at baseline (vs. controls, all p-values ≤0.1). 8 down-regulated proteins were initially elevated above thresholds associated with enhanced CV risk in the literature (myeloperoxidase, C-X-C motif chemokine 10, E-selectin, interleukin-6, cystatin B, von Willebrand factor, tumor necrosis factor receptor 1 and N-terminal prohormone brain natriuretic peptide). Treatment lowered these proteins to below their risk thresholds, except for IL-6, which was lowered but remained at its risk threshold despite successful psoriasis skin treatment. In summary, 12 weeks of ustekinumab treatment reduced serum proteins present at levels associated with CV risk in psoriasis patients. Further studies can evaluate these proteins as potential ustekinumab-modulated biomarkers of CV risk in psoriasis and the impact of ustekinumab on CV risk reduction.
Collapse
Affiliation(s)
- Merav Koschitzky
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, New York, USA,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristina Navrazhina
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, New York, USA,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, Weill Cornell University, New York, New York, USA
| | - Michael S. Garshick
- Center for the Prevention of Cardiovascular Disease and Leon H. Charney Division of Cardiology, Department of Medicine, Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Juana Gonzalez
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Joseph Han
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sandra Garcet
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - James G. Krueger
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
17
|
Tarkin JM, Gonçalves I. Could targeting the macrophage urokinase-type plasminogen activator receptor be a bullseye for PET imaging of atherosclerotic plaque inflammation? Atherosclerosis 2022; 352:80-82. [DOI: 10.1016/j.atherosclerosis.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022]
|
18
|
Śmiłowska K, Śmiłowski M, Partyka R, Kokocińska D, Jałowiecki P. Personalised Approach to Diagnosing and Managing Ischemic Stroke with a Plasma-Soluble Urokinase-Type Plasminogen Activator Receptor. J Pers Med 2022; 12:jpm12030457. [PMID: 35330458 PMCID: PMC8953259 DOI: 10.3390/jpm12030457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 02/01/2023] Open
Abstract
Background: The increasing incidence of ischemic stroke has led to the search for a novel biomarker to predict the course of disease and the risk of mortality. Recently, the role of the soluble urokinase plasminogen activator receptor (suPAR) as a biomarker and indicator of immune system activation has been widely examined. Therefore, the aim of the current study was to assess the dynamics of changes in serum levels of suPAR in ischemic stroke and to evaluate the prognostic value of suPAR in determining mortality risk. Methods: Eighty patients from the Department of Neurology, diagnosed with ischemic stroke, were enrolled in the study. Residual blood was obtained from all the patients on the first, third and seventh days after their ischemic stroke and the concentrations of suPAR and C-reactive protein (CRP), as well as the number of leukocytes and National Institute of Health’s Stroke Scale (NIHSS) scores, were evaluated. Results: On the first day of ischemic stroke, the average suPAR concentration was 6.55 ng/mL; on the third day, it was 8.29 ng/mL; on the seventh day, it was 9.16 ng/mL. The average CRP concentration on the first day of ischemic stroke was 4.96 mg/L; on the third day, it was 11.76 mg/L; on the seventh day, it was 17.17 mg/L. The number of leukocytes on the first day of ischemic stroke was 7.32 × 103/mm3; on the third day, it was 9.27 × 103/mm3; on the seventh day, it was 10.41 × 103/mm3. Neurological condition, which was assessed via the NIHSS, on the first day of ischemic stroke, was scored at 10.71 points; on the third day, it was scored at 12.34 points; on the seventh day, it was scored at 13.75 points. An increase in the values of all the evaluated parameters on the first, third and seventh days of hospitalisation was observed. The patients with hypertension, ischemic heart disease and type 2 diabetes showed higher suPAR and CRP concentrations at the baseline as well as on subsequent days of hospitalisation. The greatest sensitivity and specificity were characterised by suPAR-3, where a value above 10.5 ng/mL resulted in a significant increase in mortality risk. Moreover, an NIHSS-1 score above 12 points and a CRP-3 concentration above 15.6 mg/L significantly increased the risk of death in the course of the disease. Conclusions: The plasma suPAR concentration after ischemic stroke is strongly related to the patient’s clinical status, with a higher concentration on the first and third days of stroke resulting in a poorer prognosis at a later stage of treatment. Therefore, assessing the concentration of this parameter has important prognostic value.
Collapse
Affiliation(s)
- Katarzyna Śmiłowska
- Department of Emergency Medicine, Faculty of Medical Sciences, Medical University of Silesia, 40-055 Katowice, Poland; (R.P.); (D.K.); (P.J.)
- Department of Neurology, 5th Regional Hospital in Sosnowiec, Plac Medyków 1, 41-200 Sosnowiec, Poland
- Correspondence:
| | - Marek Śmiłowski
- Department of Hematology and Bone Marrow Transplantation, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Robert Partyka
- Department of Emergency Medicine, Faculty of Medical Sciences, Medical University of Silesia, 40-055 Katowice, Poland; (R.P.); (D.K.); (P.J.)
| | - Danuta Kokocińska
- Department of Emergency Medicine, Faculty of Medical Sciences, Medical University of Silesia, 40-055 Katowice, Poland; (R.P.); (D.K.); (P.J.)
| | - Przemysław Jałowiecki
- Department of Emergency Medicine, Faculty of Medical Sciences, Medical University of Silesia, 40-055 Katowice, Poland; (R.P.); (D.K.); (P.J.)
| |
Collapse
|
19
|
Walter JE, Amrein MLF, Schäfer I, Zimmermann T, Lopez-Ayala P, Boeddinghaus J, Twerenbold R, Puelacher C, Nestelberger T, Wussler D, Honegger U, Badertscher P, Eugen-Olsen J, Koechlin L, Fahrni G, Jeger R, Kaiser C, Zellweger M, Mueller C. Soluble urokinase plasminogen activator receptor and functionally relevant coronary artery disease: a prospective cohort study. Biomarkers 2022; 27:278-285. [PMID: 35112976 DOI: 10.1080/1354750x.2022.2038269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Soluble urokinase plasminogen activator receptor (suPAR) is an emerging biomarker associated with anatomical CAD burden and cardiovascular outcomes including myocardial infarction (MI) and death. We aimed at validating previous findings of the prognostic value of suPAR and evaluated its diagnostic potential for functional relevant CAD (fCAD). METHODS Consecutive patients with suspected fCAD were enrolled. Adjudication of fCAD was performed blinded to suPAR concentrations by myocardial perfusion single photon emission tomography (MPI-SPECT) and coronary angiography. Prognostic outcome measures included all-cause, cardiovascular death, and incident MI during 2-year follow-up. RESULTS Among consecutive 968 patients, SuPAR concentrations were higher in patients with fCAD compared to those without (3.45ng/mL versus 3.20ng/mL, p = 0.007), without acceptable diagnostic accuracy (area under the curve [AUC]: 0.56, 95%CI 0.52-0.60). SuPAR correlated with high-sensitivity cardiac-troponin (hs-cTn) T (Spearman's rho (ρ) 0.393, p < 0.001), NT-proBNP (ρ = 0.327, p < 0.001), age (ρ = 0.364, p < 0.001) and very weakly with coronary atherosclerosis (ρ = 0.123, p < 0.001). Prognostic discrimination of suPAR was moderate for cardiovascular death (AUC =0.72, 95%CI 0.62-0.81) and all-cause death (AUC =0.72, 95%CI 0.65-0.79) at 2-years. SuPAR remained a significant predictor for all-cause death in the full model (HR =1.96, p = 0.001). CONCLUSIONS SuPAR was an independent predictor of all-cause death, without diagnostic utility for fCAD.
Collapse
Affiliation(s)
- Joan Elias Walter
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Department of Radiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Melissa Lee Fen Amrein
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Ibrahim Schäfer
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Tobias Zimmermann
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Pedro Lopez-Ayala
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Jasper Boeddinghaus
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Raphael Twerenbold
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,University Heart & Vascular Center Hamburg, Germany
| | - Christian Puelacher
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Department of Radiology, University Hospital Zurich, University of Zurich, Switzerland
| | - Thomas Nestelberger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Vancouver General Hospital, University of British Columbia, Canada
| | - Desiree Wussler
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Universitäts-Herzzentrum Bad Krozingen, Germany
| | - Ursina Honegger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Patrick Badertscher
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Copenhagen University Hospital Hvidovre, Denmark
| | - Luca Koechlin
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland.,Department of Cardiac Surgery, University Hospital Basel, University of Basel, Switzerland
| | - Gregor Fahrni
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Raban Jeger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Christoph Kaiser
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Michael Zellweger
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| | - Christian Mueller
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology, University Hospital Basel, University of Basel, Switzerland
| |
Collapse
|
20
|
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
|
21
|
Różański D, Szlufik S, Tomasiuk R, Milanowski Ł, Figura M, Saramak K, Myrcha P, Koziorowski D. Soluble Urokinase Plasminogen Activator Receptor Levels Correlation with Other Inflammatory Factors in Prognosis of Disability and Death in Patients with Ischemic Stroke. Brain Sci 2021; 12:39. [PMID: 35053782 PMCID: PMC8774014 DOI: 10.3390/brainsci12010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/18/2021] [Accepted: 12/24/2021] [Indexed: 12/03/2022] Open
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is an inflammatory biomarker elevated in cardiovascular diseases. The aim of this 3-year follow-up prospective study was to evaluate suPAR levels in patients with a first ischemic stroke in correlation with CRP, PCT, NT-proCNP and endothelin 1-21 and to investigate the impact of suPAR on the outcome. Fifty-one patients (mean age 73.7+ = 11.9 years, 26 female and 25 male) were included. Samples were collected on the first (suPAR 1), third (suPAR 3) and seventh days after stroke onset (suPAR 7). Plasma samples were analyzed using ELISA. A phone interview was conducted to collect follow-up information after 24 and 36 months (modified Rankin Scale, mRS). A positive correlation between suPAR levels and other inflammatory biomarkers (except endothelin 3) was observed. A positive correlation between suPAR 3 and mRS score at 24 months was observed (p = 0.042). The logistic regression model revealed no significant effect of suPAR on death occurrence in the first 24 months: suPAR 1 (p = 0.8794), suPAR 3 (p = 0.2757), and suPAR 7 (p = 0.3652). The suPAR level is a potential inflammatory marker in ischemic stroke, and there is a correlation with other markers. There is no major impact on mortality. However, the suPAR level is associated with a degree of disability or dependence in daily activities 2 years after a stroke.
Collapse
Affiliation(s)
- Dorota Różański
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 02-091 Warsaw, Poland; (Ł.M.); (M.F.); (K.S.); (D.K.)
| | - Stanisław Szlufik
- Faculty of Medical Sciences and Health Sciences, Kazimierz Pulaski University of Technology and Humanities, 26-600 Radom, Poland; (S.S.); (R.T.)
| | - Ryszard Tomasiuk
- Faculty of Medical Sciences and Health Sciences, Kazimierz Pulaski University of Technology and Humanities, 26-600 Radom, Poland; (S.S.); (R.T.)
| | - Łukasz Milanowski
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 02-091 Warsaw, Poland; (Ł.M.); (M.F.); (K.S.); (D.K.)
| | - Monika Figura
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 02-091 Warsaw, Poland; (Ł.M.); (M.F.); (K.S.); (D.K.)
| | - Kamila Saramak
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 02-091 Warsaw, Poland; (Ł.M.); (M.F.); (K.S.); (D.K.)
| | - Piotr Myrcha
- Department of General and Vascular Surgery, Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Department of General, Vascular and Oncological Surgery, Masovian Brodnowski Hospital, 03-242 Warsaw, Poland
| | - Dariusz Koziorowski
- Department of Neurology, Faculty of Health Sciences, Medical University of Warsaw, 02-091 Warsaw, Poland; (Ł.M.); (M.F.); (K.S.); (D.K.)
| |
Collapse
|
22
|
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
|
23
|
Fagerberg B, Barregard L. Review of cadmium exposure and smoking-independent effects on atherosclerotic cardiovascular disease in the general population. J Intern Med 2021; 290:1153-1179. [PMID: 34157165 DOI: 10.1111/joim.13350] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Exposure to cadmium (Cd) via food and smoking is associated with an increased risk of atherosclerotic cardiovascular disease (ASCVD). Blood and urine levels of Cd are established biomarkers of exposure. OBJECTIVES To review (1) the smoking-independent associations between Cd exposure and ASCVD, including the possible presence of a nonlinear dose-response relationship with Cd exposure and (2) the causal effects of Cd exposure on different stages of atherosclerosis. METHODS Narrative review. RESULTS Cd confers increased risk of ASCVD and asymptomatic atherosclerosis in the carotid and coronary arteries above B-Cd >0.5 μg/L or U-Cd >0.5 μg/g creatinine, but it has not been shown below a threshold of these exposure levels. Adjustment for smoking does not exclude the possibility of residual confounding, but several studies in never-smoking cohorts have shown associations between Cd and ASCVD, and experimental studies have demonstrated pro-atherosclerotic effects of Cd. Cd accumulates in arterial walls and atherosclerotic plaques, reaching levels shown to have proatherosclerotic effects. Suggested early effects are increased subendothelial retention of atherogenic lipoproteins, which become oxidized, and endothelial dysfunction and damage with increased permeability for monocytes, which in the intima turn to macrophages and then to foam cells. Later, Cd may contribute to plaque rupture and erosion by endothelial apoptosis and degradation of the fibrous cap. Finally, by having prothrombotic and antifibrinolytic effects, the CVD risk may be further increased. CONCLUSIONS There is strong evidence that Cd causes ASCVD above a suggested exposure level via mechanisms in early as well as the late stages of atherosclerotic disease.
Collapse
Affiliation(s)
- Björn Fagerberg
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lars Barregard
- Occupational and Environmental Medicine, Department of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
24
|
Olson NC, Raffield LM, Moxley AH, Miller-Fleming TW, Auer PL, Franceschini N, Ngo D, Thornton TA, Lange EM, Li Y, Nickerson DA, Zakai NA, Gerszten RE, Cox NJ, Correa A, Mohlke KL, Reiner AP. Soluble Urokinase Plasminogen Activator Receptor: Genetic Variation and Cardiovascular Disease Risk in Black Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003421. [PMID: 34706549 PMCID: PMC8692389 DOI: 10.1161/circgen.121.003421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 08/30/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND suPAR (Soluble urokinase plasminogen activator receptor) has emerged as an important biomarker of coagulation, inflammation, and cardiovascular disease (CVD) risk. The contribution of suPAR to CVD risk and its genetic influence in Black populations have not been evaluated. METHODS We measured suPAR in 3492 Black adults from the prospective, community-based JHS (Jackson Heart Study). Cross-sectional associations of suPAR with lifestyle and CVD risk factors were assessed, whole-genome sequence data were used to evaluate genetic associations of suPAR, and relationships of suPAR with incident CVD outcomes and overall mortality were estimated over follow-up. RESULTS In Cox models adjusted for traditional CVD risk factors, estimated glomerular filtration rate, and CRP (C-reactive protein), each 1-SD higher suPAR was associated with a 21% to 31% increased risk of incident coronary heart disease, heart failure, stroke, and mortality. In the genome-wide association study, 2 missense (rs399145 encoding p.Thr86Ala, rs4760 encoding p.Phe272Leu) and 2 noncoding regulatory variants (rs73935023 within an enhancer element and rs4251805 within the promoter) of PLAUR on chromosome 19 were each independently associated with suPAR and together explained 14% of suPAR phenotypic variation. The allele frequencies of each of the four suPAR-associated genetic variants differ considerably across African and European populations. We further show that PLAUR rs73935023 can alter transcriptional activity in vitro. We did not find any association between genetically determined suPAR and CVD in JHS or a larger electronic medical record-based analyses of Blacks or Whites. CONCLUSIONS Our results demonstrate the importance of ancestry-differentiated genetic variation on suPAR levels and indicate suPAR is a CVD biomarker in Black adults.
Collapse
Affiliation(s)
- Nels C. Olson
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Anne H. Moxley
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Tyne W. Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Paul L. Auer
- Joseph J. Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Debby Ngo
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Timothy A. Thornton
- Departments of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Ethan M. Lange
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Deborah A. Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Neil A. Zakai
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Nancy J. Cox
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Alex P. Reiner
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
25
|
Muhammad IF, Borné Y, Zaigham S, Söderholm M, Johnson L, Persson M, Melander O, Engström G. Comparison of risk factors for ischemic stroke and coronary events in a population-based cohort. BMC Cardiovasc Disord 2021; 21:536. [PMID: 34772344 PMCID: PMC8588679 DOI: 10.1186/s12872-021-02344-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 10/25/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Although coronary events (CE) and ischemic stroke share many risk factors, there are also some important differences. The aim of this paper was to assess the association of risk factors in relation to incident CE and ischemic stroke and to evaluate the heterogeneity in patterns of risk factors between the two outcomes. METHOD Traditional risk factors and inflammatory markers associated with coronary events and ischemic stroke were measured in the Malmö Diet and Cancer Cohort (MDCS, n = 26 519), where a total of 2270 incident ischemic stroke and 3087 incident CE occurred during a mean follow up time 19 ± 6 years, and in relation to inflammatory markers in the cardiovascular sub-cohort (MDC-CV, n = 4795). Cox regression analysis was used to obtain hazard ratios. A modified Lunn-McNeil competing risk analysis was conducted to assess the significance of any differences in risk profiles of these outcomes. RESULTS Most cardiovascular risk factors were associated both with incident CE and ischemic stroke. However, current smoking, ApoB, low ApoA1, male sex and education level of ≤ 9 years of schooling were preferentially associated with CE compared to ischemic stroke. Conversely, age showed a stronger association with ischemic stroke than with CE. CONCLUSION CE and ischemic stroke have broadly similar risk factors profiles. However, there are some important differential associations, as well as substantial differences in the magnitude of the association. These could reflect the distinct biology of atherogenesis in different vascular beds. The difference in the determinants highlights the importance of looking at CE and ischemic stroke, two manifestations of cardiovascular disease, separately.
Collapse
Affiliation(s)
- Iram Faqir Muhammad
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden.
| | - Yan Borné
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Suneela Zaigham
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Martin Söderholm
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden.,Skåne University Hospital, Malmö, Sweden
| | - Linda Johnson
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden.,Skåne University Hospital, Malmö, Sweden
| | - Margaretha Persson
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden.,Skåne University Hospital, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, CRC 60:13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| |
Collapse
|
26
|
Bourassa KJ, Rasmussen LJH, Danese A, Eugen-Olsen J, Harrington H, Houts R, Poulton R, Ramrakha S, Sugden K, Williams B, Moffitt TE, Caspi A. Linking stressful life events and chronic inflammation using suPAR (soluble urokinase plasminogen activator receptor). Brain Behav Immun 2021; 97:79-88. [PMID: 34224821 PMCID: PMC8453112 DOI: 10.1016/j.bbi.2021.06.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/27/2022] Open
Abstract
Stressful life events have been linked to declining health, and inflammation has been proposed as a physiological mechanism that might explain this association. Using 828 participants from the Dunedin Longitudinal Study, we tested whether people who experienced more stressful life events during adulthood would show elevated systemic inflammation when followed up in midlife, at age 45. We studied three inflammatory biomarkers: C-reactive protein (CRP), interleukin-6 (IL-6), and a newer biomarker, soluble urokinase plasminogen activator receptor (suPAR), which is thought to index systemic chronic inflammation. Stressful life events were not associated with CRP or IL-6. However, people who experienced more stressful life events from age 38 to 44 had elevated suPAR at age 45, and had significantly greater increases in suPAR from baseline to follow-up across the same period. When examining stressful life events across the lifespan, both adverse childhood experiences (ACEs) and adult stressful life events were independently associated with suPAR at age 45. ACEs moderated the association of adult stressful life events and suPAR at age 45-children with more ACEs showed higher suPAR levels after experiencing stressful life events as adults. The results suggest systemic chronic inflammation is one physiological mechanism that could link stressful life events and health, and support the use of suPAR as a useful biomarker for such research.
Collapse
Affiliation(s)
- Kyle J. Bourassa
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC,Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Line J. H. Rasmussen
- Department of Psychology & Neuroscience, Duke University, Durham, NC,Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Andrea Danese
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, King’s College London, London, United Kingdom,Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom,National and Specialist Child and Adolescent Mental Health Services Trauma, Anxiety, and Depression Clinic, South London and Maudsley National Health Service Foundation Trust, London, United Kingdom
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | | | - Renate Houts
- Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Richie Poulton
- Department of Psychology, University of Otago, Otago, New Zealand
| | - Sandhya Ramrakha
- Department of Psychology, University of Otago, Otago, New Zealand
| | - Karen Sugden
- Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Ben Williams
- Department of Psychology & Neuroscience, Duke University, Durham, NC
| | - Terrie E. Moffitt
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC,Department of Psychology & Neuroscience, Duke University, Durham, NC,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC,Center for Genomic and Computational Biology, Duke University, Durham, NC
| | - Avshalom Caspi
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC,Department of Psychology & Neuroscience, Duke University, Durham, NC,Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC,Center for Genomic and Computational Biology, Duke University, Durham, NC
| |
Collapse
|
27
|
Sun J, Singh P, Österlund J, Orho-Melander M, Melander O, Engström G, Edsfeldt A. Hyperglycaemia-associated Caspase-3 predicts diabetes and coronary artery disease events. J Intern Med 2021; 290:855-865. [PMID: 34309093 PMCID: PMC7612448 DOI: 10.1111/joim.13327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/05/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Apoptosis is central in both diabetes and atherosclerosis, linked to pancreatic beta cell death and plaque progression. Circulating Caspase-3 has also been associated with diabetes and coronary calcium score. Here, we explored if soluble Caspase-3 (sCaspase-3) is associated with cardio-metabolic risk factors and predicts incidence of diabetes and coronary artery disease (CAD). METHODS Clinical data and plasma from 4637 individuals from the Malmö Diet and Cancer cohort were studied. Plasma sCaspase-3 was measured by a Proximity Extension Assay. National registers were used to identify diabetes and CAD events during follow-up. Type 2 diabetes risk variants and expression quantitative trait loci (eQTL) for sCaspase-3 were retrieved from the DIAGRAM consortium and the Genotype-Tissue Expression project. RESULTS HbA1c was the factor with the strongest association with sCaspase-3 (r = 0.18, P = 1.3x10-36 ). During follow-up 666 individuals developed diabetes and 648 individuals suffered from CAD. Increasing sCaspase-3 was associated with a higher risk of developing diabetes (hazard ratio (HR) 1.18 per 1unit; P = 7 × 10-5 ) and CAD (HR 1.2 per 1 unit, P = 1 × 10-4 ) during follow-up. A genetic variant rs60780116, located upstream of CASP3, showed strong association with type 2 diabetes (OR 1.06, 95%CI 1.04-1.07, P = 8.4 × 10-11 ). An eQTL was identified between this variant and gene expression of CASP3, where the allele positively correlated with type 2 diabetes was associated with increased CASP3 expression in blood. CONCLUSIONS The present study provides evidence for plasma sCaspase-3 as a marker of cardio-metabolic risk factors and as a predictor of future diabetes and CAD in a cohort without cardiovascular disease or diabetes at baseline.
Collapse
Affiliation(s)
- Jiangming Sun
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Pratibha Singh
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Johan Österlund
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Marju Orho-Melander
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Olle Melander
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Gunnar Engström
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Andreas Edsfeldt
- From the, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Department of Cardiology, Skåne University Hospital, Malmö, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Fernandez-Botran R, Szabo YZ, Lyle KB, Newton TL. The levels of soluble urokinase plasminogen activator receptor (suPAR) in saliva are influenced by acute stress. Biol Psychol 2021; 165:108147. [PMID: 34492333 DOI: 10.1016/j.biopsycho.2021.108147] [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: 01/21/2021] [Revised: 05/14/2021] [Accepted: 07/03/2021] [Indexed: 11/29/2022]
Abstract
Although elevations in systemic suPAR levels have been associated with inflammatory conditions and with exposure to life stress and adversity, it is not yet clear whether acute psychological stress influences suPAR levels, either systemically and/or in saliva. The aim of this study was to investigate whether salivary suPAR levels are increased following exposure to acute psychological stress. Healthy subjects, aged 18-40 years, completed a laboratory psychological stressor and provided saliva samples before and after the stress test (60 min apart). Levels of suPAR as well as those of cytokines increased in the post-stress samples (all ps < .001). Baseline and post-stress IL-1β and TNF-α as well as post-stress IL-6 correlated significantly with suPAR (all ps < .01), but IL-10 and baseline IL-6 did not. These results show that suPAR levels in saliva are stress-reactive and suggest a potential application as stress biomarkers in saliva, particularly given the advantage of easily detectable concentrations.
Collapse
Affiliation(s)
- Rafael Fernandez-Botran
- Department of Pathology & Laboratory Medicine, University of Louisville, Louisville, KY, United States.
| | - Yvette Z Szabo
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, KY, United States; Department of Veteran Affairs, VISN 17 Center of Excellence for Research on Returning War Veterans, Waco, TX, United States.
| | - Keith B Lyle
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, KY, United States.
| | - Tamara L Newton
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, KY, United States.
| |
Collapse
|
29
|
Reisinger AC, Niedrist T, Posch F, Hatzl S, Hackl G, Prattes J, Schilcher G, Meißl AM, Raggam RB, Herrmann M, Eller P. Soluble urokinase plasminogen activator receptor (suPAR) predicts critical illness and kidney failure in patients admitted to the intensive care unit. Sci Rep 2021; 11:17476. [PMID: 34471146 PMCID: PMC8410930 DOI: 10.1038/s41598-021-96352-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Soluble urokinase plasminogen activator receptor (suPAR) is an inflammatory biomarker and risk factor for kidney diseases, with a potential prognostic value in critically ill patients. In this monocentric prospective study, we measured plasma suPAR levels immediately after ICU admission in unselected 237 consecutive patients using a turbidimetric assay. Primary objective was the prognostic value for ICU- and 28-day mortality. Secondary objectives were association with sequential organ failure assessment (SOFA) score, coagulation and inflammation markers, AKI-3 and differences in prespecified subgroups. Median suPAR levels were 8.0 ng/mL [25th-75th percentile 4.3-14.4], with lower levels in ICU survivors than non-survivors (6.7 vs. 11.6 ng/mL, p < 0.001). SuPAR levels were higher in COVID-19, kidney disease, moderate-to-severe liver disease, and sepsis. ICU mortality increased by an odds ratio (OR) of 4.7 in patients with the highest compared to lowest quartile suPAR. Kaplan-Meier overall survival estimates at 3 months were 63% and 49%, in patients with suPAR below/above 12 ng/mL (log-rank p = 0.027). Due to an observed interaction between SOFA score and suPAR, we performed a random forest method identifying cutoffs. ICU mortality was 53%, 17% and 2% in patients with a SOFA score > 7, SOFA ≤ 7 & suPAR > 8 ng/mL, and SOFA score ≤ 7 & suPAR ≤ 8 ng/mL, respectively. suPAR was a significant predictor for AKI-3 occurrence (OR per doubling 1.89, 95% CI: 1.20-2.98; p = 0.006). suPAR levels at ICU admission may offer additional value for risk stratification especially in ICU patients with moderate organ dysfunction as reflected by a SOFA score ≤ 7.
Collapse
Affiliation(s)
- Alexander C. Reisinger
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Tobias Niedrist
- grid.11598.340000 0000 8988 2476Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Florian Posch
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Stefan Hatzl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria ,grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Gerald Hackl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Juergen Prattes
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Section of Infectious Diseases and Tropical Medicine, Medical University of Graz, Graz, Austria
| | - Gernot Schilcher
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Anna-Maria Meißl
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Nephrology, Medical University of Graz, Graz, Austria
| | - Reinhard B. Raggam
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Division of Angiology, Medical University of Graz, Graz, Austria
| | - Markus Herrmann
- grid.11598.340000 0000 8988 2476Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- grid.11598.340000 0000 8988 2476Department of Internal Medicine, Intensive Care Unit, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| |
Collapse
|
30
|
Rasmussen LJH, Caspi A, Ambler A, Danese A, Elliott M, Eugen-Olsen J, Hariri AR, Harrington H, Houts R, Poulton R, Ramrakha S, Sugden K, Williams B, Moffitt TE. Association Between Elevated suPAR, a New Biomarker of Inflammation, and Accelerated Aging. J Gerontol A Biol Sci Med Sci 2021; 76:318-327. [PMID: 32766674 PMCID: PMC7812430 DOI: 10.1093/gerona/glaa178] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background To understand and measure the association between chronic inflammation, aging, and age-related diseases, broadly applicable standard biomarkers of systemic chronic inflammation are needed. We tested whether elevated blood levels of the emerging chronic inflammation marker soluble urokinase plasminogen activator receptor (suPAR) were associated with accelerated aging, lower functional capacity, and cognitive decline. Methods We used data from the Dunedin Study, a population-representative 1972–1973 New Zealand birth cohort (n = 1037) that has observed participants to age 45 years. Plasma suPAR levels were analyzed at ages 38 and 45 years. We performed regression analyses adjusted for sex, smoking, C-reactive protein, and current health conditions. Results Of 997 still-living participants, 875 (88%) had plasma suPAR measured at age 45. Elevated suPAR was associated with accelerated pace of biological aging across multiple organ systems, older facial appearance, and with structural signs of older brain age. Moreover, participants with higher suPAR levels had greater decline in physical function and cognitive function from childhood to adulthood compared to those with lower suPAR levels. Finally, improvements in health habits between ages 38 and 45 (smoking cessation or increased physical activity) were associated with less steep increases in suPAR levels over those years. Conclusions Our findings provide initial support for the utility of suPAR in studying the role of chronic inflammation in accelerated aging and functional decline.
Collapse
Affiliation(s)
- Line Jee Hartmann Rasmussen
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina.,Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina.,Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK
| | - Antony Ambler
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Andrea Danese
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK.,National and Specialist Child and Adolescent Mental Health Services Trauma, Anxiety, and Depression Clinic, South London and Maudsley National Health Service Foundation Trust, London, UK
| | - Maxwell Elliott
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - HonaLee Harrington
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Renate Houts
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Richie Poulton
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Sandhya Ramrakha
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Benjamin Williams
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina.,Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK
| |
Collapse
|
31
|
Bengaard AK, Iversen E, Kallemose T, Juul-Larsen HG, Rasmussen LJH, Dalhoff KP, Andersen O, Eugen-Olsen J, Houlind MB. Using soluble urokinase plasminogen activator receptor to stratify patients for medication review in the emergency department. Br J Clin Pharmacol 2021; 88:1679-1690. [PMID: 34242432 DOI: 10.1111/bcp.14982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 11/30/2022] Open
Abstract
AIMS To investigate whether the association between levels of medication use (including polypharmacy and potentially inappropriate medications [PIMs]) and health outcomes such as readmission and mortality is dependent on baseline soluble urokinase plasminogen activator receptor (suPAR). METHODS This registry-based cohort study included medical patients admitted to the emergency department at Copenhagen University Hospital Hvidovre, Denmark. Patients were grouped according to their admission suPAR levels: low (0-3 ng/mL), intermediate (3-6 ng/mL), or high (>6 ng/mL). Hyper-polypharmacy was defined as ≥10 prescribed medications. PIMs were identified based on the EU(7)-PIM list, and data on admissions and mortality were obtained from national registries. Risk of 90-day readmission and mortality was assessed by Cox regression analysis adjusted for sex, age and Charlson comorbidity index. Results were reported as hazard ratios within 90 days of index discharge. RESULTS In total, 26 291 patients (median age 57.3 y; 52.7% female) were included. Risk of 90-day readmission and mortality increased significantly for patients with higher suPAR or higher number of medications. Among patients with low suPAR, patients with ≥10 prescribed medications had a hazard ratio of 2.41 (95% confidence interval = 2.09-2.78) for 90-day readmission and 8.46 (95% confidence interval = 2.53-28.28) for 90-day mortality compared to patients with 0 medications. Patients with high suPAR generally had high risk of readmission and mortality, and the impact of medication use was less pronounced in this group. Similar, but weaker, association patterns were observed between suPAR and PIMs. CONCLUSION The association between levels of medication use and health outcomes is dependent on baseline suPAR.
Collapse
Affiliation(s)
- Anne Kathrine Bengaard
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,The Capital Region Pharmacy, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Esben Iversen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thomas Kallemose
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Helle Gybel Juul-Larsen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Line Jee Hartmann Rasmussen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
| | - Kim Peder Dalhoff
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Emergency Department, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Morten Baltzer Houlind
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.,The Capital Region Pharmacy, Herlev, Denmark.,Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Nilsson Wadström B, Persson M, Engström G, Nilsson PM. Aortic Stiffness, Inflammation, and Incidence of Cardiovascular Events in Elderly Participants From the General Population. Angiology 2021; 73:51-59. [PMID: 34013787 DOI: 10.1177/00033197211017406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Low-grade inflammation and arterial stiffness are key factors in the development of vascular aging. However, the interplay between arterial stiffness and inflammation for cardiovascular (CV) disease is unclear. Aortic pulse wave velocity (aPWV) and the inflammatory markers, high-sensitivity C-reactive protein (CRP) and orosomucoid, were measured in 2710 participants (median age: 72 years). These participants were followed up for a mean of 7.6 years for a composite CV disease end point. Per 1 interquartile range increment of CRP and orosomucoid, respectively, aPWV increased by 0.19 m/s (95% CI: 0.07-0.32) and 0.19 m/s (0.11-0.27), after multifactorial adjustment. Mediation analysis showed that aPWV, after multifactorial adjustment, mediated 8% (-4, 20) of the CV disease risk associated with CRP and 8% (-4, 18) of orosomucoid risk. The associated risk increased with combinations of high aPWV and high CRP or orosomucoid. We found no evidence that arterial PWV acted as an important mediator of the relationship between systemic inflammation and CV disease risk in this elderly population. The results instead indicate an additive effect. Our study supports the view that arterial stiffness and chronic inflammation affects CV risk mainly through separate causal pathways.
Collapse
Affiliation(s)
| | - Margaretha Persson
- Department of Clinical Sciences, 5193Lund University, Skåne University Hospital, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, 5193Lund University, Skåne University Hospital, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences, 5193Lund University, Skåne University Hospital, Malmö, Sweden.,Clinical Research Unit, Department of Internal medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
33
|
Lind L, Gigante B, Borné Y, Feldreich T, Leppert J, Hedberg P, Östgren CJ, Nyström FH, Sundström J, Ärnlöv J, Baldassarre D, Tremoli E, Veglia F, Hamsten A, O'Donnell CJ, Franceschini N, Orho-Melander M, Nilsson J, Melander O, Engström G, Mälarstig A. Plasma Protein Profile of Carotid Artery Atherosclerosis and Atherosclerotic Outcomes: Meta-Analyses and Mendelian Randomization Analyses. Arterioscler Thromb Vasc Biol 2021; 41:1777-1788. [PMID: 33657885 DOI: 10.1161/atvbaha.120.315597] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lars Lind
- Department of Medical Sciences, Uppsala University, Sweden (L.L., J.S.)
| | - Bruna Gigante
- Unit of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Sweden (B.G., A.H., A.M.)
| | - Yan Borné
- Department of Clinical Sciences Malmö, Lund University, Sweden (Y.B., M.O.-M., J.N., O.M., G.E.)
| | - Tobias Feldreich
- School of Health and Social Sciences, Dalarna University, Falun, Sweden (T.F., J.A.)
| | - Jerzy Leppert
- Centre for Clinical Research, Uppsala University (J.L., P.H.), Västmanland County Hospital, Västerås, Sweden
| | - Pär Hedberg
- Centre for Clinical Research, Uppsala University (J.L., P.H.), Västmanland County Hospital, Västerås, Sweden.,Department of Clinical Physiology (P.H.), Västmanland County Hospital, Västerås, Sweden
| | - Carl Johan Östgren
- Department of Health, Medicine and Caring Sciences, Linköping University, Sweden (C.J.O., F.H.N.).,Department of Medicine, Boston University, MA (C.J.O.)
| | - Fredrik H Nyström
- Department of Health, Medicine and Caring Sciences, Linköping University, Sweden (C.J.O., F.H.N.)
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Sweden (L.L., J.S.).,The George Institute for Global Health, University of New South Wales, Sydney, Australia (J.S.)
| | - Johan Ärnlöv
- School of Health and Social Sciences, Dalarna University, Falun, Sweden (T.F., J.A.)
| | - Damiano Baldassarre
- Damiano Baldassarre, Department of Medical Biotechnology and Translational Medicine, Università di Milano (D.B.).,Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., E.T., F.V.)
| | - Elena Tremoli
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., E.T., F.V.)
| | - Fabrizio Veglia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., E.T., F.V.)
| | - Anders Hamsten
- Unit of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Sweden (B.G., A.H., A.M.)
| | - Christopher J O'Donnell
- Department of Health, Medicine and Caring Sciences, Linköping University, Sweden (C.J.O., F.H.N.).,Department of Medicine, Boston University, MA (C.J.O.)
| | - Nora Franceschini
- Department of Epidemiology, University of North Caroline, Capel Hill (N.F.)
| | - Marju Orho-Melander
- Department of Clinical Sciences Malmö, Lund University, Sweden (Y.B., M.O.-M., J.N., O.M., G.E.)
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Sweden (Y.B., M.O.-M., J.N., O.M., G.E.)
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Sweden (Y.B., M.O.-M., J.N., O.M., G.E.)
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, Sweden (Y.B., M.O.-M., J.N., O.M., G.E.)
| | - Anders Mälarstig
- Unit of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Sweden (B.G., A.H., A.M.)
| |
Collapse
|
34
|
Sipos B, Jirak P, Paar V, Rezar R, Mirna M, Kopp K, Hoppe UC, Berezin AE, Lichtenauer M. Promising Novel Biomarkers in Cardiovascular Diseases. APPLIED SCIENCES 2021; 11:3654. [DOI: 10.3390/app11083654] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
Abstract
Cardiovascular diseases remain the most common causes of death globally, according to the World Health Organization. In recent years, a great number of biomarkers have been investigated, whereas only some have gained value in the diagnosis, prognosis, and risk stratification of different cardiovascular illnesses. As numerous studies have investigated the diagnostic yield of novel biomarkers in various disease entities every year, this review aims to provide an overview of the current status of four promising representatives. In particular, this manuscript refers to soluble suppression of tumorigenicity 2 (sST2), heart-type fatty acid binding protein (H-FABP), growth differentiation factor (GDF-15) and soluble urokinase-type plasminogen activator receptor (suPAR). These markers are of special interest as they are thought to provide an accurate estimate of cardiovascular risk in certain patient populations, especially those with pre-existing diseases, such as obesity or diabetes mellitus. We sought to give an overview of their function, individual diagnostic and predictive value and determination in the laboratory. A review of the literature regarding the aforementioned cardiovascular biomarkers yielded manifold results with respect to their individual diagnostic and prognostic value. Yet, the clinical relevance of these findings remains unclear, warranting further studies to identify their optimal use in clinical routine.
Collapse
Affiliation(s)
- Brigitte Sipos
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Peter Jirak
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Vera Paar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Richard Rezar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Moritz Mirna
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Kristen Kopp
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Uta C. Hoppe
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | - Alexander E. Berezin
- Internal Medicine Department, State Medical University, 69000 Zaporozhye, Ukraine
| | - Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
35
|
Pan J, Borné Y, Orho-Melander M, Nilsson J, Melander O, Engström G. The associations between red cell distribution width and plasma proteins in a general population. Clin Proteomics 2021; 18:12. [PMID: 33781199 PMCID: PMC8008679 DOI: 10.1186/s12014-021-09319-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/12/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND High red cell distribution width (RDW) has been increasingly recognized as a risk factor for cardiovascular diseases (CVDs), but the underlying mechanisms remain unknown. Our aim was to explore the associations between RDW and plasma proteins implicated in the pathogenesis of CVD using a targeted proteomics panel. METHODS RDW and 88 plasma proteins were measured in a population-based cohort study (n = 4726), Malmö Diet and Cancer-Cardiovascular Cohort (MDC-CC). A random 2/3 of the cohort was used as discovery sample and remaining 1/3 was used for replication. Multiple linear regression was used to assess the associations between RDW and plasma proteins, with adjustments for age, sex, and other potential confounders. Proteins with Bonferroni-corrected significant associations with RDW in the discovery sub-cohort were validated in the replication cohort. RESULTS Thirteen of 88 plasma proteins had significant associations with RDW in the discovery sample, after multivariate adjustments. Eleven of them were also significant in the replication sample, including SIR2-like protein 2 (SIRT2), stem cell factor (SCF, inversely), melusin (ITGB1BP2), growth differentiation factor-15 (GDF-15), matrix metalloproteinase-7 (MMP-7), hepatocyte growth factor (HGF), chitinase-3-like protein 1 (CHI3L1), interleukin-8 (IL-8), CD40 ligand (CD40-L), urokinase plasminogen activator surface receptor (U-PAR) and matrix metalloproteinase-3 (MMP-3). CONCLUSIONS Several proteins from this targeted proteomics panel were associated with RDW in this cohort. These proteins could potentially be linked to the increased cardiovascular risk in individuals with high RDW.
Collapse
Affiliation(s)
- Jingxue Pan
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden.
| | - Yan Borné
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, CRC Hus 60 plan 13, Jan Waldenströms gata 35, 20502, Malmö, Sweden
| |
Collapse
|
36
|
Aharaz A, Rasmussen JH, McNulty HBØ, Cyron A, Fabricius PK, Bengaard AK, Sejberg HRC, Simonsen RRL, Treldal C, Houlind MB. A Collaborative Deprescribing Intervention in a Subacute Medical Outpatient Clinic: A Pilot Randomized Controlled Trial. Metabolites 2021; 11:204. [PMID: 33808080 PMCID: PMC8066016 DOI: 10.3390/metabo11040204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/11/2021] [Accepted: 03/25/2021] [Indexed: 12/03/2022] Open
Abstract
Medication deprescribing is essential to prevent inappropriate medication use in multimorbid patients. However, experience of deprescribing in Danish Subacute Medical Outpatient Clinics (SMOCs) is limited. The objective of our pilot study was to evaluate the feasibility and sustainability of a collaborative deprescribing intervention by a pharmacist and a physician to multimorbid patients in a SMOC. A randomized controlled pilot study was conducted, with phone follow-up at 30 and 365+ days. A senior pharmacist performed a systematic deprescribing intervention using the Screening Tool of Older Persons' potentially inappropriate Prescriptions (STOPP) criteria, the Danish deprescribing list, and patient interviews. A senior physician received the proposed recommendations and decided which should be implemented. The main outcome was the number of patients having ≥1 medication where deprescribing status was sustained 30 days after inclusion. Out of 76 eligible patients, 72 (95%) were included and 67 (93%) completed the study (57% male; mean age 73 years; mean number of 10 prescribed medications). Nineteen patients (56%) in the intervention group and four (12%) in the control group had ≥1 medication where deprescribing status was sustained 30 days after inclusion (p = 0.015). In total, 37 medications were deprescribed in the intervention group and five in the control group. At 365+ days after inclusion, 97% and 100% of the deprescribed medications were sustained in the intervention and control groups, respectively. The three most frequently deprescribed medication groups were analgesics, cardiovascular, and gastrointestinal medications. In conclusion, a collaborative deprescribing intervention for multimorbid patients was feasible and resulted in sustainable deprescribing of medication in a SMOC.
Collapse
Affiliation(s)
- Anissa Aharaz
- The Capital Region Pharmacy, 2730 Herlev, Denmark; (H.B.Ø.M.); (A.K.B.); (H.R.C.S.); (R.R.L.S.); (C.T.); (M.B.H.)
- Multidisciplinary Outpatient Clinic (Fællesambulatoriet, subakutte patientforløb), Copenhagen University Hospital—Amager and Hvidovre, 2300 Copenhagen, Denmark; (J.H.R.); (A.C.)
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark;
| | - Jens Henning Rasmussen
- Multidisciplinary Outpatient Clinic (Fællesambulatoriet, subakutte patientforløb), Copenhagen University Hospital—Amager and Hvidovre, 2300 Copenhagen, Denmark; (J.H.R.); (A.C.)
- Department of Emergency Medicine, Copenhagen University Hospital—Bispebjerg and Frederiksberg, 2400 Copenhagen, Denmark
| | - Helle Bach Ølgaard McNulty
- The Capital Region Pharmacy, 2730 Herlev, Denmark; (H.B.Ø.M.); (A.K.B.); (H.R.C.S.); (R.R.L.S.); (C.T.); (M.B.H.)
| | - Arne Cyron
- Multidisciplinary Outpatient Clinic (Fællesambulatoriet, subakutte patientforløb), Copenhagen University Hospital—Amager and Hvidovre, 2300 Copenhagen, Denmark; (J.H.R.); (A.C.)
| | - Pia Keinicke Fabricius
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark;
| | - Anne Kathrine Bengaard
- The Capital Region Pharmacy, 2730 Herlev, Denmark; (H.B.Ø.M.); (A.K.B.); (H.R.C.S.); (R.R.L.S.); (C.T.); (M.B.H.)
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Rikke Rie Løvig Simonsen
- The Capital Region Pharmacy, 2730 Herlev, Denmark; (H.B.Ø.M.); (A.K.B.); (H.R.C.S.); (R.R.L.S.); (C.T.); (M.B.H.)
| | - Charlotte Treldal
- The Capital Region Pharmacy, 2730 Herlev, Denmark; (H.B.Ø.M.); (A.K.B.); (H.R.C.S.); (R.R.L.S.); (C.T.); (M.B.H.)
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark;
| | - Morten Baltzer Houlind
- The Capital Region Pharmacy, 2730 Herlev, Denmark; (H.B.Ø.M.); (A.K.B.); (H.R.C.S.); (R.R.L.S.); (C.T.); (M.B.H.)
- Department of Clinical Research, Copenhagen University Hospital—Amager and Hvidovre, 2650 Copenhagen, Denmark;
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
37
|
Velissaris D, Zareifopoulos N, Koniari I, Karamouzos V, Bousis D, Gerakaris A, Platanaki C, Kounis N. Soluble Urokinase Plasminogen Activator Receptor as a Diagnostic and Prognostic Biomarker in Cardiac Disease. J Clin Med Res 2021; 13:133-142. [PMID: 33854652 PMCID: PMC8016523 DOI: 10.14740/jocmr4459] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 12/26/2022] Open
Abstract
This review summarizes the published literature referring to the use and validity of the biomarker soluble urokinase plasminogen activator receptor (suPAR) when used for the assessment of patients with cardiac diseases. It is measured by enzyme-linked immunosorbent assay (ELISA) in plasma samples. In cardiology a cut-off value range of 3.5 - 4.5 ng/mL has been commonly utilized. Different cut-off values may be applied based on the measuring kit used, the patient population and the clinical setting. A PubMed/Medline search was conducted aiming to identify all publications relevant to the use of suPAR in patients with cardiac diseases. A total of 39 studies were included in this review. suPAR as a marker of inflammation has been used more extensively in recent years, alone or in combination with other biomarkers of inflammation and cardiac pathology in the assessment of patients with acute and chronic cardiac diseases. suPAR is closely related to the pathophysiology of cardiac disease, and a number of publications encourages its use as a valuable biomarker in the assessment of patients presenting to the cardiology service. It may be most valuable in the risk assessment of patients with acute coronary syndromes and congestive heart failure, as suPAR elevation may be an independent predictor of mortality in these conditions. In conclusion, among several biomarkers used for clinical entities with underlying inflammatory pathophysiology including cardiac diseases, suPAR is a novel attractive index for the prognostic risk stratification of cardiac patients. More research is warranted to confirm its diagnostic and prognostic validity, alone or combined with other cardiac and inflammatory biomarkers.
Collapse
Affiliation(s)
- Dimitrios Velissaris
- Department of Internal and Emergency Medicine, University of Patras, Patras, Greece
| | | | - Ioanna Koniari
- Department of Cardiology, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | | | - Dimitris Bousis
- Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Andreas Gerakaris
- Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Christina Platanaki
- Department of Internal Medicine, University Hospital of Patras, Patras, Greece
| | - Nicholas Kounis
- Department of Cardiology, University of Patras, Patras, Greece
| |
Collapse
|
38
|
Bruno RM, Nilsson PM, Engström G, Wadström BN, Empana JP, Boutouyrie P, Laurent S. Early and Supernormal Vascular Aging: Clinical Characteristics and Association With Incident Cardiovascular Events. Hypertension 2020; 76:1616-1624. [PMID: 32895017 DOI: 10.1161/hypertensionaha.120.14971] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pulse wave velocity is an established marker of early vascular aging but may also help identifying individuals with supernormal vascular aging. We tested the hypothesis that individuals with the largest difference (Δ-age) between chronological and vascular age show the lowest rate of cardiovascular events and may thus be defined as supernormal vascular aging. Vascular age was defined as the predicted age in the best fitting multivariable regression model including classical risk factors and treatment and pulse wave velocity, in a subset of the Reference Values for Arterial Stiffness Collaboration Database (n=3347). Δ-age was then calculated as chronological age minus vascular age, and the 10th and 90th percentiles were used to define early (Δ-age<-5.7 years), normal (Δ-age -5.7 to 6.8 years) and supernormal vascular aging (Δ-age>6.8 years). The risk for fatal and nonfatal cardiovascular events associated with vascular aging categories was investigated in the Malmö Diet and Cancer Study cohort (n=2642). In the Malmö Diet and Cancer Study Cohort (6.6-year follow-up, 286 events), Δ-age was significantly (P<0.01) and inversely associated with cardiovascular events. Compared with normal vascular aging, supernormal vascular aging had lower risk (hazard ratio, 0.59 [95% CI, 0.41-0.85]), whereas early vascular aging had higher risk (hazard ratio, 2.70 [95% CI, 1.55-4.70]) of cardiovascular events, in particular coronary events. There was no significant association with all-cause mortality. This study represents the first validation of the clinical significance of the supernormal vascular aging concept, based on prospective data. Its further characterization may help discovering novel protective molecular pathways and providing preventive strategies for successful vascular aging.
Collapse
Affiliation(s)
- Rosa Maria Bruno
- From the INSERM, U970, Paris Cardiovascular Research Center-PARCC, France (R.M.B., J.-P.E., P.B., S.L.)
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden (P.M.N., G.E., B.N.W.)
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden (P.M.N., G.E., B.N.W.)
| | - Benjamin Nilsson Wadström
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden (P.M.N., G.E., B.N.W.)
| | - Jean-Philippe Empana
- From the INSERM, U970, Paris Cardiovascular Research Center-PARCC, France (R.M.B., J.-P.E., P.B., S.L.).,Université de Paris, France (RM.B., J.-P.E., P.B., S.L.)
| | - Pierre Boutouyrie
- From the INSERM, U970, Paris Cardiovascular Research Center-PARCC, France (R.M.B., J.-P.E., P.B., S.L.).,Université de Paris, France (RM.B., J.-P.E., P.B., S.L.).,Assistance Publique-Hopitaux de Paris, France (P.B., S.L.)
| | - Stephane Laurent
- From the INSERM, U970, Paris Cardiovascular Research Center-PARCC, France (R.M.B., J.-P.E., P.B., S.L.).,Université de Paris, France (RM.B., J.-P.E., P.B., S.L.).,Assistance Publique-Hopitaux de Paris, France (P.B., S.L.)
| |
Collapse
|
39
|
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
|
40
|
Nilsson Wadström B, Engström G, Nilsson PM. Exploring and comparing definitions of healthy vascular ageing in the population: characteristics and prospective cardiovascular risk. J Hum Hypertens 2020; 35:428-436. [PMID: 32415282 DOI: 10.1038/s41371-020-0353-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 11/09/2022]
Abstract
Different methods can be used to define healthy vascular ageing (HVA). In this prospective cohort study, we explored three different definitions in order to provide guidance for which to use. 2718 subjects were included from the Cardiovascular (CV) arm of the Malmö Diet Cancer Study (MDCS; median age 71.9 years, 62.2% females). Three different definitions of HVA were used: HVA-1 (<15th percentile of aortic pulse wave velocity (aPWV) distribution from age-quintiles); HVA-2 (<35th percentile of aPWV+ <35th percentile of carotid Intima-Media Thickness. cIMT); and HVA-3 (aPWV < 7.6 m/s + no hypertension). The HVA-1 and HVA-2 groups were compared with the HVA-3, and to the corresponding groups without HVA (non-HVA), in cross-sectional analyses for baseline characteristics and using Cox regressions for prospective risk, yielding hazard ratios (HRs) adjusted for conventional risk factors. A composite CVD endpoint was used, consisting of myocardial infarctions, ischemic heart disease mortality, and coronary artery procedures. The baseline characteristics were, with minor exceptions, similar across HVA groups. In the fully adjusted model, the HRs (95%CI) were 0.62 (0.41-0.93), 0.45 (0.26-0.76), and 0.56 (0.34-0.91) for HVA-1, HVA-2, and HVA-3, respectively. In summary, this observational study of elderly subjects provides examples of integrating hypertension and cIMT in the definition of HVA, as compared with only using aPWV. As aPWV itself is such a robust marker of HVA, it is demanding to find additional markers which improve the definition. There is a need to create a generally accepted definition of HVA.
Collapse
Affiliation(s)
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden. .,Clinical Research Unit, Department of Internal medicine, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
41
|
Al‐Badri A, Tahhan AS, Sabbak N, Alkhoder A, Liu C, Ko Y, Vaccarino V, Martini A, Sidoti A, Goodwin C, Ghazzal B, Beshiri A, Murtagh G, Mehta PK, Quyyumi AA. Soluble Urokinase-Type Plasminogen Activator Receptor and High-Sensitivity Troponin Levels Predict Outcomes in Nonobstructive Coronary Artery Disease. J Am Heart Assoc 2020; 9:e015515. [PMID: 32301366 PMCID: PMC7428519 DOI: 10.1161/jaha.119.015515] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Multiple biomarkers have been independently and additively associated with major adverse cardiovascular events in patients with coronary artery disease. We investigated the prognostic value of suPAR (soluble urokinase-type plasminogen activator receptor) and hsTnI (high-sensitivity troponin I) levels in symptomatic patients with no obstructive coronary artery disease. We hypothesized that high levels of these biomarkers will be associated with the risk of future adverse outcomes. Methods and Results Plasma levels of suPAR and hsTnI were measured in 556 symptomatic patients with no obstructive coronary artery disease. A biomarker risk score was calculated by counting the number of biomarkers above the median in this cohort (suPAR>2523 pg/mL and hsTnI>2.7 pg/mL). Survival analyses were performed with models adjusted for traditional risk factors. All-cause death and major adverse cardiovascular events (cardiovascular death, myocardial infarction, stroke, and heart failure) served as clinical outcomes over a median follow-up of 6.2 years. Mean age was 57±10 years, 49% of the cohort patients were female, and 68% had a positive stress test. High suPAR and hsTnI levels were independent predictors of all-cause death (hazard ratio=3.2 [95% CI, 1.8-5.7] and 1.3 [95% CI, 1.0-1.7], respectively; both P<0.04) and major adverse cardiovascular events (hazard ratio=2.7 [95% CI, 1.4-5.4] and 1.5 [95% CI, 1.2-2.0], respectively; both P<0.002). Compared with a biomarker risk score of 0, biomarker risk scores of 1 and 2 were associated with 19- and 14-fold increased risk of death and development of major adverse cardiovascular events, respectively. Conclusions Among symptomatic patients with no obstructive coronary artery disease, higher levels of suPAR and hsTnI were independently and additively associated with an increased risk of adverse events. Whether modification of these biomarkers will improve risk in these patients needs further investigation.
Collapse
Affiliation(s)
- Ahmed Al‐Badri
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Ayman Samman Tahhan
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Nabil Sabbak
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Ayman Alkhoder
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Chang Liu
- Departments of Biostatistics and BioinformaticsEmory University School of MedicineAtlantaGA
| | - Yi‐An Ko
- Departments of Biostatistics and BioinformaticsEmory University School of MedicineAtlantaGA
| | - Viola Vaccarino
- Department of Epidemiology and Rollins School of Public HealthEmory UniversityAtlantaGA
| | - Afif Martini
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Arianna Sidoti
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Cydney Goodwin
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Bahjat Ghazzal
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Agim Beshiri
- Diagnostics DivisionAbbott LaboratoriesNorth ChicagoIL
| | | | - Puja K. Mehta
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Arshed A. Quyyumi
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| |
Collapse
|
42
|
Sandø A, Schultz M, Eugen-Olsen J, Køber L, Engstrøm T, Kelbæk H, Jørgensen E, Saunamäki K, Holmvang L, Pedersen F, Tilsted HH, Høfsten D, Helqvist S, Clemmensen P, Iversen K. Soluble urokinase receptor as a predictor of non-cardiac mortality in patients with percutaneous coronary intervention treated ST-segment elevation myocardial infarction. Clin Biochem 2020; 80:8-13. [PMID: 32213303 DOI: 10.1016/j.clinbiochem.2020.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 03/16/2020] [Accepted: 03/19/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Identification of patients at high risk of non-cardiac mortality following ST-segment elevation myocardial infarction (STEMI) could guide clinicians to identify patients who require attention due to serious non-cardiac conditions after the acute phase of STEMI. The purpose of this study was to evaluate if the non-specific and prognostic biomarker of inflammation and comorbidity, soluble urokinase receptor (suPAR), could predict non-cardiac mortality in a cohort of STEMI patients. METHODS SuPAR was measured in 1,190 STEMI patients who underwent primary percutaneous coronary intervention (pPCI). The primary endpoint was non-cardiac mortality, secondary endpoints were cardiac mortality, all-cause mortality, reinfarction and periprocedural acute kidney injury. Backwards elimination of potential confounders significantly associated with the respective outcome was used to adjust associations. RESULTS Patients were followed for a median of 3.0 years (interquartile range 2.5- 3.6 years). Multivariate cox regression revealed that a plasma suPAR level above 3.70 ng mL-1 was associated with non-cardiac and cardiac mortality at hazard ratios 3.33 (95% confidence interval 1.67-6.63, p = 0.001, adjusted for age) and 0.99 (0.18-5.30, p = 0.98, adjusted for previous myocardial infarction and left ventricular ejection fraction), respectively. CONCLUSION In patients with pPCI treated STEMI, suPAR was an independent prognostic biomarker of non-cardiac but not cardiac mortality and may identify patients with high risk of non-cardiac mortality.
Collapse
Affiliation(s)
- Andreas Sandø
- Department of Cardiology, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - Martin Schultz
- Department of Cardiology, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Copenhagen University Hospital Hvidovre, Kettegård Alle 30, 2650 Hvidovre, Denmark
| | - Lars Køber
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Thomas Engstrøm
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Henning Kelbæk
- Department of Cardiology, Zealand University Hospital, Sygehusvej 10, 4000 Roskilde, Denmark.
| | - Erik Jørgensen
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Kari Saunamäki
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Lene Holmvang
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Frants Pedersen
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Hans Henrik Tilsted
- Department of Cardiology, Aalborg University Hospital, Hobrovej 18, 9000 Aalborg, Denmark.
| | - Dan Høfsten
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Steffen Helqvist
- The Heart Center, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Peter Clemmensen
- Department of General and Interventional Cardiology, University Heart Center, Hamburg-Eppendorf, Hamburg, Germany and Department of Medicine, Division of Cardiology, Nykøbing Falster Hospital, University of Southern Denmark, Fjordvej 15, 4800 Nykøbing Falster, Denmark.
| | - Kasper Iversen
- Department of Cardiology, Copenhagen University Hospital Herlev, Herlev Ringvej 75, 2730 Herlev, Denmark.
| |
Collapse
|
43
|
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
|
44
|
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
|
45
|
Ishikawa H, Izumiya Y, Shibata A, Ichikawa Y, Yamaguchi T, Yamaguchi Y, Kitada R, Iwata S, Ehara S, Tomita S, Hanatani A, Yoshiyama M. Soluble urokinase-type plasminogen activator receptor represents exercise tolerance and predicts adverse cardiac events in patients with heart failure. Heart Vessels 2019; 35:681-688. [PMID: 31741050 DOI: 10.1007/s00380-019-01538-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/08/2019] [Indexed: 10/25/2022]
Abstract
Soluble urokinase-type plasminogen activator receptor (suPAR) is a membrane-binding protein that is released into the blood stream by immune activation. Recent reports suggest that circulating suPAR levels are associated with adverse cardiovascular outcomes. Exercise tolerance is an independent predictor of prognosis in patients with heart failure (HF); however, the relationship between serum suPAR level and exercise tolerance is unclear. We prospectively enrolled 94 patients who were hospitalized for worsening of HF. All patients underwent a symptom-limited cardiopulmonary exercise test to evaluate exercise tolerance. The median value of serum suPAR was 4848 pg/ml. During follow up, 44 patients (47%) were admitted for all-cause mortality and re-hospitalization for HF. Median serum suPAR was significantly higher in the patients with cardiac events than in the patients with non-event group. Patients were divided into two groups according to circulating suPAR levels. Kaplan-Meier analysis demonstrated that adverse cardiac events were significantly higher in the high suPAR group (log-rank p = 0.023). Multivariate analysis revealed that suPAR was independently correlated with the parameters of exercise tolerance such as anaerobic threshold (p = 0.007) and peak oxygen uptake (p = 0.005). suPAR levels predicted adverse cardiac events and independently correlated with the parameters of exercise tolerance. suPAR could be a useful surrogate biomarker of exercise tolerance in patients with HF.
Collapse
Affiliation(s)
- Hirotoshi Ishikawa
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan.
| | - Atsushi Shibata
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Yoshitaka Ichikawa
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Takehiro Yamaguchi
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yumi Yamaguchi
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Ryoko Kitada
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Shinichi Iwata
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Shoichi Ehara
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Shuhei Tomita
- Department of Pharmacology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Akihisa Hanatani
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| | - Minoru Yoshiyama
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8586, Japan
| |
Collapse
|
46
|
Ahmed RM, Khalil MA, Ibrahim AH, Eid HM, Abdelbasset WK, Soliman GS. Clinical value of soluble urokinase type plasminogen activator receptors in chronic kidney disease. Medicine (Baltimore) 2019; 98:e17146. [PMID: 31567954 PMCID: PMC6756685 DOI: 10.1097/md.0000000000017146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED Chronic kidney disease (CKD) will progress to end stage without treatment, the decline off renal function may not linear. A sensitive marker such as soluble urokinase-type plasminogen activator receptors (suPARs) may allow potential intervention and treatment in earlier stages of CKD. OBJECTIVES This study was designed to measure plasma (suPAR) in patients with CKD with different stages and to find its correlation with the disease severity. METHODS This study was conducted on 114 subjects, 84 were patients with different stages and different causes of CKD, and 30 healthy subjects as controls. Blood urea, serum creatinine, serum high-sensitive C-reactive protein, estimated glomerular filtration rate, and 24 hours proteinuria were measured, renal biopsy was done for all patients, and plasma (suPAR) was measured using enzyme-linked immunosorbent assay. RESULTS suPAR plasma levels were significantly higher in patients with CKD (7.9 ± 3.82 ng/mL) than controls (1.76 ± 0.77 ng/mL, P < .001). suPAR correlated with the disease severity. In stage 1 to 2 group, it was 3.7 ± 1.5 ng/mL, in stage 3 to 4, it was 10.10 ± 1.22 ng/mL, and in stage 5 group, it was 12.34 ± 0.88 ng/mL; the difference between the 3 groups was highly significant (P < .001). A cutoff point 2.5 ng/mL of suPAR was found between controls and stage 1 group. According to the cause of CKD, although patients with obstructive cause and those with focal glomerulosclerosis had the higher levels 9.11 ± 3.32 ng/mL and 8.73 ± 3.19 ng/mL, respectively, but there was no significant difference between patients with CKD according to the cause of the CKD. CONCLUSION Plasma (suPAR) increased in patients with CKD and correlated with disease severity.
Collapse
Affiliation(s)
| | | | - Amal H. Ibrahim
- Department of Internal Medicine, Faculty of Medicine, Al-Azhar university, Cairo, Egypt
| | - Hanaa M. Eid
- Department of Internal Medicine, Faculty of Medicine, Al-Azhar university, Cairo, Egypt
| | - Walid Kamal Abdelbasset
- Department of Physical Therapy and Health Rehabilitation, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital
| | - Gaber S. Soliman
- Department of Physical Therapy for Cardiovascular/Respiratory Disorder and Geriatrics, Faculty of Physical Therapy, Cairo University, Giza, Egypt
- Department of Physical Therapy and Health Rehabilitation, College of Applied Medical Sciences in Al-Qurayyat, Jouf University, Al-Jawf, Saudi Arabia
| |
Collapse
|
47
|
Swart C, Lammertyn L, Welsh PI, Botha-Le Roux S. Vascular Structure and Inflammation in a South African Population: The SABPA Study. Pulse (Basel) 2019; 7:60-68. [PMID: 32399438 DOI: 10.1159/000500899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/09/2019] [Indexed: 02/03/2023] Open
Abstract
Background and Objective Inflammation plays a role in the early onset of cardiovascular disease. However, longitudinal studies on this topic, especially in South African populations, are scant. We explored whether early changes in vascular structure are associated with changes in inflammation. Methods We investigated 303 South African teachers aged 20-65 years at two intervals, three years apart. Standardised methods were used to determine carotid intima-media thickness (IMT) and cross-sectional wall area (CSWA) as measures of vascular structure, as well as the inflammatory markers soluble urokinase plasminogen activator receptor (suPAR), C-reactive protein (CRP) and interleukin-6 (IL-6) at baseline and follow-up. Results IMT and CSWA were higher, while CRP was lower at follow-up than at baseline. After adjusting for confounding factors, percent change in IMT was inversely associated with percent change in suPAR (β = -0.12, p = 0.036; adjusted R<sup>2</sup> = 0.16) only, and only in the highest tertile of percent change in suPAR (r = -0.31; p = 0.002). Conclusion The early structural changes observed are not related to higher inflammatory levels in this South African population. Future studies are needed to investigate the possible protective effect of suPAR on early changes in vascular structure, especially with the focus on cardiovascular disease prevention.
Collapse
Affiliation(s)
- Carla Swart
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa
| | - Leandi Lammertyn
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Paul I Welsh
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Shani Botha-Le Roux
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom, South Africa.,MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
48
|
Nilsson Wadström B, Fatehali AAH, Engström G, Nilsson PM. A Vascular Aging Index as Independent Predictor of Cardiovascular Events and Total Mortality in an Elderly Urban Population. Angiology 2019; 70:929-937. [PMID: 31234636 DOI: 10.1177/0003319719857270] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The morphology and function of the arteries can be directly measured using different established methods. This prospective cohort study aimed to translate 2 of these, aortic pulse wave velocity (aPWV) and carotid intima-media thickness (cIMT), into a combined Vascular Aging Index (VAI) and then evaluate the predictive power of aPWV, cIMT, and VAI. Patients (n = 2718) were included from the cardiovascular arm of the Malmö Diet and Cancer Study (median age 71.9 years, 62.2% females). Total follow-up time was 16 448 person-years and a composite cardiovascular disease (CVD) end point was used. Cox regressions yielded adjusted hazard ratios (95% confidence interval) per 1 standard deviation increment of loge aPWV, loge cIMT, and loge VAI of 1.25 (1.08-1.45, P = .003), 1.27 (1.13-1.44, P < .001), and 1.45 (1.26-1.68, P < .001), respectively. The C-statistics increased from 0.714 to 0.734 when adding aPWV and cIMT to a model of conventional risk factors. Net Reclassification Index also showed a significant (P < .001) improvement for the classification of event-free patients and no change for patients with events. A VAI based on aPWV and cIMT had a good predictive performance. Used together, aPWV and cIMT incrementally and significantly improve the prediction of CVD events by correctly down-adjusting the predicted risk for noncases.
Collapse
Affiliation(s)
| | | | - Gunnar Engström
- 2 Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Peter M Nilsson
- 2 Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
49
|
Schenk M, Eichelmann F, Schulze MB, Rudovich N, Pfeiffer AF, di Giuseppe R, Boeing H, Aleksandrova K. Reproducibility of novel immune-inflammatory biomarkers over 4 months: an analysis with repeated measures design. Biomark Med 2019; 13:639-648. [PMID: 31157547 DOI: 10.2217/bmm-2018-0351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Assessment of the feasibility and reliability of immune-inflammatory biomarker measurements. Methods: The following biomarkers were assessed in 207 predominantly healthy participants at baseline and after 4 months: MMF, TGF-β, suPAR and clusterin. Results: Intraclass correlation coefficients (95% CIs) ranged from good for TGF-β (0.75 [95% CI: 0.33-0.90]) to excellent for MMF (0.81 [95% CI: 0.64-0.90]), clusterin (0.83 [95% CI: 0.78-0.87]) and suPAR (0.91 [95% CI: 0.88-0.93]). Measurement of TGF-β was challenged by the large number of values below the detection limit. Conclusion: Single measurements of suPAR, clusterin and MMF could serve as feasible and reliable biomarkers of immune-inflammatory pathways in biomedical research.
Collapse
Affiliation(s)
- Matthew Schenk
- Senior Scientist Group Nutrition, Immunity & Metabolism, Department of Nutrition & Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Fabian Eichelmann
- Senior Scientist Group Nutrition, Immunity & Metabolism, Department of Nutrition & Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.,German Centre for Diabetes Research, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,University of Potsdam, Institute of Nutritional Science, Potsdam, Germany
| | - Natalia Rudovich
- German Centre for Diabetes Research, Germany.,Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.,Department of Endocrinology, Diabetes & Nutrition, Campus Benjamin Franklin, Charité University Medicine, Berlin, Germany.,Division of Endocrinology & Diabetes, Department of Internal Medicine, Spital Bülach, Bülach, Switzerland
| | - Andreas F Pfeiffer
- German Centre for Diabetes Research, Germany.,Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.,Department of Endocrinology, Diabetes & Nutrition, Campus Benjamin Franklin, Charité University Medicine, Berlin, Germany
| | - Romina di Giuseppe
- Institute of Epidemiology, Christian-Albrechts University Kiel, Kiel, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Krasimira Aleksandrova
- Senior Scientist Group Nutrition, Immunity & Metabolism, Department of Nutrition & Gerontology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.,University of Potsdam, Institute of Nutritional Science, Potsdam, Germany
| |
Collapse
|
50
|
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
|